188
Bayesian Adaptive Methods for Clinical Trials Pfizer Research Technology Center, Cambridge, MA March 2, 2012 presented by Bradley P. Carlin University of Minnesota [email protected] Bayesian Adaptive Methods for Clinical Trials – p. 1/100

Bayesian Adaptive Methods for Clinical Trialsbrad/7440/slidePfizer-UofM2012.pdfBayesian Adaptive Methods for Clinical Trials Pfizer Research Technology Center, Cambridge, MA March

  • Upload
    dinhdan

  • View
    218

  • Download
    1

Embed Size (px)

Citation preview

Bayesian Adaptive Methods forClinical Trials

Pfizer Research Technology Center, Cambridge, MA

March 2, 2012

presented by

Bradley P. Carlin

University of Minnesota

[email protected]

Bayesian Adaptive Methods for Clinical Trials – p. 1/100

Textbooks for this courseRecommended

(“BCLM”): Bayesian Adaptive Methods for ClinicalTrials (ISBN 978-1-4398-2548-8) by S.M. Berry, B.P.Carlin, J.J. Lee, and P. Müller, Boca Raton, FL:Chapman and Hall/CRC Press, 2010.

Other books of interest:Your favorite math stat and linear models booksBayesian Methods for Data Analysis, 3rd ed., by B.P.Carlin and T.A. Louis, Boca Raton, FL: Chapmanand Hall/CRC Press, 2009.Bayesian Approaches to Clinical Trials andHealth-Care Evaluation, by D.J. Spiegelhalter, K.R.Abrams, and J.P. Myles: Chichester: Wiley, 2004.

Bayesian Adaptive Methods for Clinical Trials – p. 4/100

Software for Bayesian CTs IExpensive but incredibly cool commercial software: FACTS(Fixed and Adaptive Clinical Trials Simulator) software formany Phase I and II trial designs

permits dose finding in the presence of both safety andefficacy endpoints; stopping for success or futility

supports continuous, dichotomous, or time-to-event(TITE) endpoints; also longitudinal data (e.g.biomarkers)

joint venture between Berry Consultants (statistics,algorithms) and Tessella Technology and Consulting(software interface)

Website:http://www.smarterclinicaltrials.com/what-we-offer/facts/– probably best for large companies with ongoingcommitments to Bayesian CT development

Bayesian Adaptive Methods for Clinical Trials – p. 38/100

Software for Bayesian CTs IINoncommercial but still very professional software: freelyavailable from the M.D. Anderson Cancer CenterDepartment of Biostatistics Software page,https://biostatistics.mdanderson.org/SoftwareDownload/

All are stand-alone packages, free for download andlocal install

easy to learn: menu- and dialog box-driven, nicegraphics where appropriate

accompanied by extensive tutorials, typically withguidelines, exercises, and solutions

remarkably broad coverage of areas from all phases ofthe regulatory process...

Bayesian Adaptive Methods for Clinical Trials – p. 39/100

Software for Bayesian CTs IIPartial list of MD Anderson Phase I software packages:

CRMSimulator: Simulates power and Type I error ofContinual Reassessment Method (CRM) dose-findingdesigns, offering improvements in power and/or samplesize over “3 + 3" designs; see BCLM Section 3.2.

bCRM: handles bivariate dose-finding with twocompeting outcomes (say, toxicity and progression) anda single agent; see BCLM Section 3.4.2.

EffTox: finds a best dose when efficacy must be tradedoff against toxicity, both assumed increasing in dose;see BCLM Section 3.3.

ToxFinder: for combination therapy, i.e., two drugsbeing administered in combination, with only oneoutcome (usually toxicity); see BCLM Section 3.4.4.

Bayesian Adaptive Methods for Clinical Trials – p. 40/100

Software for Bayesian CTs IIPartial list of MD Anderson Phase II software packages:

Phase II PP Design: computes stopping boundaries fora single-arm Phase II predictive probability design witha binary endpoint; see BCLM Section 4.2.

MultcLean: for monitoring toxicity and efficacy in singlearm phase II clinical trials with binary data; see BCLMSection 4.3.2.

Adaptive Randomization (AR): for designing andsimulating outcome-adaptive randomized trials with upto 10 arms, using binary or time-to-event (TITE)outcomes; more patients are treated with the bettertreatment while retaining the benefits of randomization;see BCLM Section 4.4.

Bayesian Adaptive Methods for Clinical Trials – p. 41/100

Software for Bayesian CTs IIINoncommercial and not-all-that-professional software, butwith R and BUGS source code freely available: from theBCLM book’s data and software page,http://www.biostat.umn.edu/∼brad/data3.html

organized by chapter in the book

similar range of models/problems as the MDACCsoftware

continuing to grow

Mostly written in R, but those that require MCMCtypically call OpenBUGS using the BRugs package,whose installation and exemplification are given here:http://www.biostat.umn.edu/∼brad/software/BRugs/

Bayesian Adaptive Methods for Clinical Trials – p. 42/100

Software for Bayesian CTs IIIPartial list of BCLM Phase I software packages:

betabinHM.R: elementary BRugs metaanalysis programfor a single success proportion; see BCLM Section 2.4.

Power_BRugs.txt: slightly more advanced BRugs powerand Type I error-simulating program, Weibull survivalmodel; see BCLM Section 2.5.4.

titecrm.R: a basic R implementation of the TITE-CRMmethod; see BCLM Section 3.2.3.

324.R: an R program for dose-finding based on toxicityintervals (rather than fixed target levels); see BCLMSection 3.2.4.

354.R: a basic R implementation of the 2-agentcombination therapy dose-escalation method (similar toToxFinder); see BCLM Section 3.4.4.

Bayesian Adaptive Methods for Clinical Trials – p. 43/100

Software for Bayesian CTs IIIPartial list of BCLM Phase II-III software packages:

431.R: an R program for binary stopping for futilty,efficacy, or toxicity (similar to MultcLean); see BCLMSection 4.3.2.

443.R: R code for outcome adaptive randomization withdelayed survival response ; see BCLM Section 4.4.4.

adapt.R: R code to compute the simulated Type I errorand other operating characteristics of a basic one-armbinary response confirmatory trial; see BCLM Section5.2.1.

example5.4.R: an R program to simulate operatingcharacteristics of the basic confirmatory trial withdelayed outcomes; see BCLM Section 5.2.3.

Bayesian Adaptive Methods for Clinical Trials – p. 44/100

MCMC-based Bayesian designSimulating the power or other operating characteristics (say,Type I error) in this setting works as follows:

Sample “true” β values from an assumed “true prior”(skeptical, enthusiastic, or in between)

Given these, sample fake survival times ti (say, N fromeach study group) from the Weibull

We may also wish to sample fake censoring times cifrom a particular distribution (e.g., a normal truncatedbelow 0); for all i such that ti > ci, replace ti by “NA”

Compute (βL, βU ) by calling BUGS from R using BRugs

Determine the simulated trial’s outcome based onlocation of (βL, βU ) relative to the indifference zone

Repeat this process Nrep times; report empiricalfrequencies of the six possible outcomes

Bayesian Adaptive Methods for Clinical Trials – p. 68/100

Results from Power.BRugsAssuming:

Weibull shape r = 2, and N = 50 in each groupmedian survival of 36 days with 50% improvement inthe treatment groupa N(80, 20) censoring distributionthe enthusiastic prior as the “truth”

We obtain the following output from Nrep = 100 reps:

Here are simulated outcome frequencies for N= 50accept control: 0reject treatment: 0.07equivalence: 0reject control: 0.87accept treatment: 0.06no decision: 0End of BRugs power simulation

Bayesian Adaptive Methods for Clinical Trials – p. 69/100

Bayesian power calculationWe will likely wish to repeat the entire process for severalsample sizes N and several priors.

A Bayesian power calculation here might arise fromusing the enthusiastic prior as the “truth”

For Nrep = 1000 (and using 100 burn-in and 1000production MCMC iterations in each WinBUGS call), weobtained the following probabilities of rejecting thecontrol when the enthusiastic prior is true:

N Skeptical Reference Enthusiastic25 .014 .207 .47550 .087 .352 .61575 .191 .378 .652100 .288 .472 .682

Power increases with N and/or prior enthusiasm!Bayesian Adaptive Methods for Clinical Trials – p. 70/100

Type I error rate calculationA Bayesian version of this calculation would arisesimilar to the method of the previous slide, but nowassuming the skeptical prior is true

A true frequentist Type I error calculation is alsopossible: simply fix β1 = 0, and generate only the ti andci for each of the Nrep iterations.

Note that while Bayesians are free to look at their dataat any time without affecting the inference, multiplelooks will alter the frequentist Type I error behavior ofthe procedure. If this is of interest, the algorithm mustbe modified to explicitly include these multiple looks,checking for early stopping after each look.

Early stopping for futility based on predictivedistributions (“Bayesian stochastic curtailment”) mayalso be of interest – see Berry and Berry (2004)!

Bayesian Adaptive Methods for Clinical Trials – p. 71/100

Ch 3: Phase I studiesThe first application of a new drug to humans

Typically small (20-50 patients)

Main goal: to establish the safety of a proposed drug,often through determining an appropriate dosingschedule (dose-finding)

For cytotoxic agents (cancer), we assume the drugbenefit (as well as the severity of its toxicity and otherside effects) increases with dose, and thus seek themaximum tolerated dose (MTD)

Key elements:

a starting dose (often 110LD10,mice)

a definition of dose limiting toxicity (DLT)a target toxicity level (TTL) (say, 20-30%)a dose escalation scheme

Bayesian Adaptive Methods for Clinical Trials – p. 72/100

Sec 3.1: Rule-based MTD designsAlter the dose based on the toxicity observed in theprevious cohort. Most common: the 3+3 design:

1. Enter 3 patients at the lowest dose level

2. Observe the toxicity outcome:if 0/3 DLT ⇒ Treat next 3 patients at next higher doseif 1/3 DLT ⇒ Treat next 3 patients at the same dose;

if 0/3 DLT ⇒ Treat next 3 at next higher doseif 1/3 ⇒ Define this dose as MTDif 2/3 or 3/3 DLT ⇒ dose exceeds MTD

if 2/3 or 3/3 DLT ⇒ dose exceeds MTD

3. Repeat Step 2. If the last dose exceeds MTD, definethe previous dose as MTD provided 6 or more patientshave been treated at that dose.

4. MTD is defined as a dose with ≤ 2/6 DLTBayesian Adaptive Methods for Clinical Trials – p. 73/100

3+3 Design: Example

In Panel 1, dose level 3 is chosen as the MTD withestimated DLT rate 16.7%

In Panel 2, the same dose level is chosen, but withestimated DLT rate 0% (note 33.3% is also possible)

⇒ MTD choice is ad hoc and fairly imprecise

Bayesian Adaptive Methods for Clinical Trials – p. 74/100

Sec 3.2: Model-based MTD designs

Here we assume there is a monotonic relationshipbetween dose and P(DLT), typically in the form of asimple one- or two-parameter model.

In the figure, for a target toxicity level (TTL) of 33%,dose level 4 is the MTD

Bayesian Adaptive Methods for Clinical Trials – p. 75/100

Continual Reassessment Method (CRM)The first Bayesian model-based design introduced inthe literature (O’Quigley et al., 1990).

Often characterizes the dose-toxicity relationship viasimple one-parameter parametric models. That is,letting p(d) = P (DLT |dose = d), 3 possible models are:

Hyperbolic tangent: p(d) =

[exp(d)

exp(d) + exp(−d)

]a

Logistic: p(d) =exp(3 + ad)

1 + exp(3 + ad)

Power: p(d) = dexp(a)

The Bayesian posterior distribution of a induces aposterior for p(d), and hence that of the MTD for anygiven TTL!

Bayesian Adaptive Methods for Clinical Trials – p. 76/100

CRM algorithm1. Assume a vague or fully non-informative prior for a.

2. Treat 1 patient at the level closest to the currentestimate of the MTD, and observe the toxicity outcome

3. Update the posterior distribution of a, proportional to theprior for a times the likelihood,

L(a;d,y) ∝n∏

i=1

p(di)yi [1− p(di)]

1−yi ,

where di is the dose level for patient i, and where yi = 1if a DLT is observed for i and yi = 0 if not.

4. Treat the next patient at the level closest to the updatedestimate of MTD based on the posterior distribution of a.

5. Repeat these steps until a sufficiently precise estimateof a is achieved or the maximum sample size is reached

Bayesian Adaptive Methods for Clinical Trials – p. 77/100

Properties of the CRMAdvantages

Model-based method with a clearly defined objectiveTreats more patients at close to the target MTD level,hence reduces the number of patients treated at lowor ineffective dose levelsUses all the data to model the dose-toxicity curve

DisadvantagesThe dose assignment may be too aggressiveSuccess depends on a proper choice of thedose-toxicity curve and the prior distribution on a

Need special a computer program to implement thedesign

In order to address some of these safety concerns...

Bayesian Adaptive Methods for Clinical Trials – p. 78/100

Modified CRMc.f. Faries (1994, J. Biopharm. Stat.); Korn et al. (1994,Statist. in Med.); Goodman et al. (1995, Statist. in Med.)

Use CRM but with the following modifications:

Start at the lowest dose level, and do not skip doses

No dose escalation to new doses until all treatment inpatients from the previous doses are completed

Use asymmetric metrics to determine the current MTD,e.g., the level closest to but no higher than the TTL

Use cohort size of 2 or 3

Possibly use a more conservative stopping rule, e.g., nomore than 2 of 6 developed MTD at any given dose level

Bayesian Adaptive Methods for Clinical Trials – p. 79/100

Software for Modified CRM1. MD Anderson option: The CRMSimulator package from

biostatistics.mdanderson.org/SoftwareDownload/

can do simulations for the power model...

2. BCLM book-related option: The phaseIsim.R function fromwww.biostat.umn.edu/∼brad/software/BCLM ch3.html

The following code corresponds to the example on the next 2 pages!

p.tox0 <- c(.05,.15,.3,.45,.6)

s.dose <- log(p.tox0/(1-p.tox0)) - 3

phaseIsim(nsim=10000,npat=30,sdose=s.dose,prob.tox=p.tox0,

design=’3+3’, outfile=’3plus3.txt’) # 3+3 results

phaseIsim(nsim=100,npat=30,sdose=s.dose,prob.tox=p.tox0,

outfile=’CRM1.txt’) # CRM1 results

phaseIsim(nsim=100,npat=30,sdose=s.dose,prob.tox=p.tox0,

crm.group.size=3,outfile=’CRM3.txt’) # CRM3 resultsBayesian Adaptive Methods for Clinical Trials – p. 80/100

Modified CRM outperforms 3+3Suppose that in developing a new agent, we have fivepotential dose levels, and our target toxicity level is30%. We wish to simulate and compare the operatingcharacteristics of the 3+3 and two CRM designs using10,000 simulated trials.

Suppose the true probabilities of DLT at dose levels 1 to5 are 0.05, 0.15, 0.30, 0.45, and 0.60, respectively, sothat dose level 3 is the true MTD.

The table on the next page shows that the CRM designwith a cohort size of 1 (CRM 1) treats more patients atthe true MTD level, but also more patients at dose levelsabove the MTD. The overall percent of DLT for the 3+3and CRM 1 designs are 21.1 and 27.0, respectively.

By increasing the CRM cohort size from 1 to 3 (CRM 3),we treat fewer patients at levels above the MTD.

Bayesian Adaptive Methods for Clinical Trials – p. 81/100

Modified CRM outperforms 3+3Dose Ave %

1 2 3 4 5 N DLT

Scenario 1 P(DLT): 0.05 0.15 0.30 0.45 0.60

3+3 % patients 26.0 32.5 27.2 12.1 2.3 15.2 21.1

% MTD 20.5 42.7 27.5 5.7 0

CRM 1 % patients 15.6 24.1 34.7 19.0 6.7 18.5 27.0

% MTD 1.0 21.4 52.4 23.0 2.2

CRM 3 % patients 21.3 31.4 29.1 15.8 2.5 19.0 23.3

% MTD 1.5 22.6 49.8 23.7 2.4

CRM designs are much more likely to find the true MTD

CRM 3 offers protection for just a bit higher Ave N

CRM also beats 3+3 when the assigned doses are lessand more toxic than anticipated, respectively.

Bayesian Adaptive Methods for Clinical Trials – p. 82/100

Escalation w/ Overdose Control (EWOC)Same as CRM, except when choosing the next dose usethe αth quantile of the MTD’s posterior, instead of the mean

For dose x, P (DLT |x = MTD) ≡ θ, the TTL

Start at the lowest dose level, i.e., set x1 = d1

For any patient k, let πk(γ) be the MTD posterior cdf,

πk(γ) = P (MTD ≤ γ|yk)

where yk is the data currently available

Ideally, select the next dose level xk such that

πk(xk) = α

To restrict to our dose levels {d1, . . . , dr}, takex∗k = max{d1, . . . , dr : di − xk ≤ T1 and πk(xk)− α ≤ T2}for prespecified tolerances T1, T2 > 0

Bayesian Adaptive Methods for Clinical Trials – p. 83/100

Bayesian EWOCThe EWOC doses xk minimize risk with respect to theasymmetric loss function,

L(x, γ) =

{α(γ − x) for x ≤ γ (i.e., x is an underdose)

(1− α)(x− γ) for x > γ (i.e., x is an overdose).

Choosing the feasibility bound α < 0.5 corresponds toplacing a higher penalty on overdosing than onunderdosing

Choosing α = 0.5 implies a symmetric loss function, andproduces the MTD posterior median as the new dose

When α << 0.5, the final dose recommended for PhaseII study (e.g., MTD posterior median) may besignificantly larger than the dose any Phase I patienthas received ⇒ use a varying feasibility bound?

Bayesian Adaptive Methods for Clinical Trials – p. 84/100

EWOC ImplementationConsider EWOC under the logistic model,

Prob(DLT |dose = x) ≡ p(x) =exp(β0 + β1x)

1 + exp(β0 + β1x).

To ease prior specification, reparametrize from (β0, β1) to(ρ0, γ), where ρ0 = p(Xmin), the probability of DLT at theminimum dose, Xmin, and γ is the MTD. Then

β0 =1

γ −Xmin[γ logit(ρ0)−Xminlogit(θ)]

and β1 =1

γ −Xmin[logit(θ)− logit(ρ0)] .

Note: We assume that γ ∈ (Xmin, Xmax) with probability 1;we would typically take the starting dose d1 = Xmin.

Bayesian Adaptive Methods for Clinical Trials – p. 85/100

EWOC Code and Extensionswww.biostat.umn.edu/∼brad/data3.html offersWinBUGS EWOC code, using the model above andadopting independent uniform priors on γ and ρ0 on theranges (Xmin, Xmax) and (0, θ), respectively.

sisyphus.emory.edu/software ewoc.php is “the"EWOC website, featuring papers and relevant programs

Covariate adjustment = “individualized patient dosing"

Example: a phase I non-small-cell lung cancer trial ofPNU-214936, a monoclonal antibody, with a covariate(anti-SEA) previously shown to have a neutralizingeffect on PNU. A convenient dose-toxicity model:

P (DLT |x, c) = exp[β0 + β1 log(x) + β2 log(c)]

1 + exp[β0 + β1 log(x) + β2 log(c)]

where c denotes anti-SEA level.Bayesian Adaptive Methods for Clinical Trials – p. 86/100

EWOC ExtensionsRecent work by Zabor (2010) considers the case of twocovariates, one categorical and one continuous

Example: Trial of 852A, an “agonist" that can enhancethe tumor-inhibiting and immune response-boostingproperties of other oncologic agents. Let z be 0 formale, 1 for female, and let c ∈ [39, 80] be the patient’sage in years. Then our model for P (DLT ) is

P (DLT |x, c, z) = exp[β0 + β1x+ β2c+ β3z]

1 + exp[β0 + β1x+ β2c+ β3z].

Reparametrize to γmax = γ(c = 80, z = 0) and

ρ1 = P (DLT |X = Xmin, C = 39, Z = 0)

ρ2 = P (DLT |X = Xmin, C = 80, Z = 0)

ρ3 = P (DLT |X = Xmin, C = 39, Z = 1)

Bayesian Adaptive Methods for Clinical Trials – p. 87/100

EWOC Results

0.0

0.2

0.4

0.6

0.8

1.0

Dose

P(D

LT)

0.15

0.30

0.60

1.20

1.55

2.00

θ

Males

39−6465−7475−80

0.0

0.2

0.4

0.6

0.8

1.0

Dose

P(D

LT)

0.15

0.30

0.60

1.20

1.55

2.00

θ

Females

39−6465−7475−80

Figure 1: Probability of DLT by covariate group and dose

−0.5 0.0 0.5 1.0 1.5 2.0 2.5

0.0

0.2

0.4

0.6

0.8

1.0

MTD

Pos

terio

r de

nsity

True MTD=1.2n=3n=30n=60

Figure 2: The marginal posterior distribution of the maximum tolerated dose (MTD) for an 80 year old male as data accumulates

Left plot shows true dose-response curves by genderand grouping ages into 3 clinically meaningful groups

Simulated trials used a feasibility bound of α = 0.25,entered up to 20 cohorts of size 3, and stopped early if95% Bayesian CI had width < 1.2

BRugs results for posterior of γ(80, 0) in one trial (rightplot) reveal Bayesian learning as data accumulates!

Bayesian Adaptive Methods for Clinical Trials – p. 88/100

Outline

Session II (cont.)– TITE-CRM– Efficacy - Toxicity Trade-off (EffTox)– Combination Therapy

Session III (Phase II Studies)– Standard phase IIA Designs– Predictive Probability-Based Methods– Posterior Probability-Based Methods

Stopping for futility and efficacyStopping for futility, efficacy, and toxicity

– Adaptive Randomization (AR)Baseline ARResponse (Outcome-based) AR

– Biomarker-based Adaptive Design (BATTLE)1

TITE-CRM: CRM with Survival EndpointCheung and Chappell (2000, Biometrics)

Survival outcome with minimal model assumptions.For a patient observed (or censored) at time let and replace by

Using that's all!

at (calendar) time t for patient recruited at .Justification

2

TITE-CRM Algorithm

0. Initialization and cohort size k = 31. Initial dose escalation: initial dose escalation until the

first toxicity is observed or d = 62. Compute based on the

pseudo likelihood

3. Allocation: estimated toxicity probsselect dose

3

4. Next cohort:simulate (when evaluating operating characteristics)

or recruit (when carrying out the trial) the next cohort of k patients, i=n+1,…, n+k, allocated at

Record the recruitment times– When simulating, generate and save the (future)

response times – Increment and advance the calendar time

t = t + 0.5.5. Stopping:

– if stop and report posterior estimated toxicity probabilities (computed as in Step 2). Otherwise repeat with step 2.

4

Compute the Posterior of aEvaluating evaluate the posterior expectation as average over a grid:Posterior expectation:

sum over a grid.Grid:Evaluate the pointwise posterior

and compute

5

Software and Example

Software– SAS program: http://roadrunner.cancer.med.umich.edu/

wiki/index.php/TITE-CRM– R code: titecrm library

Example– Simulation truth: p0 = 0.05, 0.1, 0.2, 0.3, 0.5, 0.7 for

d = 0.05, 0.1, 0.2, 0.3, 0.5, 0.7 i.e., CRM model with a = 1.

– Target: p* = 20%.– Initial dose escalation: first two cohorts t = 0 and 6– Posterior updating: starting with t = 12.

6

A Simulated Trial Realization

(a) Allocation d* by t (b) Estimate by t

7

Dose-Finding Based On Efficacy-Toxicity Trade-OffsThall and Cook, Biometrics 60:684-693, 2004

Patient Outcome = {Efficacy, Toxicity}

The physician must specify – A Lower Limit pE* on πE = Pr(Efficacy)– An Upper Limit pT* on πT = Pr(Tox)– Three equally desirable(πE, πT) targets to construct an

Efficacy -Toxicity Trade-off ContourA dose x is acceptable if e.g.,

Pr{ πE(x,θ) > pE* | data } > .90

Pr{ πT(x,θ) < pT* | data } > .90

Given the current data, compute the posterior prob of πE and πT to determine the dose level of the next patient

8

Use the EffTox program from http://biostatistics.mdanderson.org/SoftwareDownload/

9

10

11

A Cohort-by-Cohort Illustration

AML patients relapsed within 6 months of CRRx = Fixed dose ara-C + one of 4 doses of a new “anti-sense” biological agent Res = Alive & in CR at day 35Tox = Grade 4 symptomatic tox within 35 daysNmax = 36, cohort size = 3pT* = .50 and pE* = .20 for Target pairs (.20, 0), (.60, .40), (1.00, .50)

(Courtesy of Dr. Peter Thall) 12

Dose-Finding /w the Combination of Two AgentsThall et al. (2003, Biometrics)

Binary toxicity responseBivariate dose for two agentsModel: with

Dose finding: climb up to reach target toxicity; then adjust dose combinations, keeping toxicity unchanged.– Stage 1: Dose escalation on a grid of until target

toxicity is reached.– Stage 2: Maintaining toxicity, adjust dose combination to

maximize cancer-cell killing and learning.

{0,1}iy ∈

13

Stage 1Define grid on a fixed line segmentStart with the lowest doseLet posterior mean toxicity surface after nth cohortAssign (n+1)st cohort at:

Subject to escalation controlWhen the first toxicity is observed atrefine the grid by introducing half-steps.Stop after patients. 14

Stage 2

Let set of doses with

Alternate between dose in

Each cohort, assign dose in

that maximizes expected learning and cell killing.

15

Example: Gemcitibine + Cyclophosphamide

Stage 1: Escalation on grid over line segmentStage 2: Adjust dose combination

16

As the Trial Progress …

17

Operating Characteristics

Dose combinations selected for the Gem/CTX (SD as subscripts).

selected dose; estimated and true toxicity,V = posterior uncertainty

18

Phase IIA Clinical Trial DesignH0: p ≤ p0 p0 -- an uninteresting response rateH1: p ≥ p1 p1 -- a target response rate

The observed responses: X ~ Binomial(n, p)

Simon’s Minimax/Optimal 2-Stage DesignStage 1: Enroll n1 patients,

If ≤ r1 responses, stop trial, reject H1;Otherwise, continue to Stage 2;

Stage 2: Enroll Nmax - n1 patients, If ≤ r responses, reject H1Otherwise, “accept” H0

PET(p0) = Prob(Early Termination | p0) = pbinom(r1, n1 , p0)E(N |p0) = n1 + [1 - PET(p0)] × (Nmax - n1)

Minimax Design: give smallest NmaxOptimal Design: yield smallest E(N|p0) 19

Presenter
Presentation Notes
In phase II cancer clinical trials, one of the focuses is on testing if a new treatment regimen has a promising response rate that warrants further investigation. In this single arm hypothesis testing setting, p0 is an uninteresting response rate and p1 is a desired target response rate.

Predictive Probability (PP) Design• PP is the prob of rej H0 at the end of study should the current trend continue, i.e., given the current data, the chance of a “positive” result at the end of study.

• If PP is very large or very small, essentially we know the answer can stop the trial and draw a conclusion.# of future pts after x responses in n pts: m=Nmax − n# of future responses: Y ~ beta-binomial(m, a0 + x, b0 + n − x)For each Y=i: f(p|x, Y=i)= beta(a0 + x + i, b0 + Nmax − x − i)Predictive Probability (PP) =∑ {Prob(Y=i | x) × [Prob(p > p0 | p ~ f(p|x,Y=i) , x, Y=i) > θT ]}

• If PP < θL, stop the trial, accept H0. Otherwise, continue to the next stage until Nmax

• At the end of trial, reject if Prob(p > p0 | p, x, Y=y) > θT. Otherwise, accept H0 20

Presenter
Presentation Notes
Given maximum accrual of Nmax patients, number of potential future patients is m, number of future responses in m patients Y has a beta-binomial distribution. For each observation of Y, posterior distribution of response rate p is again a beta distribution.

PP Designs

Goal: Find θL , θT , and Nmax to satisfying the type I and type II error rates constraints.Properties:

1. PP design can control type I and type II error rates while allow interim monitoring.

2. Under H0, PP design can yield a higher PET(p0), and smaller E(N|p0) or Nmax than Simon’s 2-stage design

3. PP design produces a flexible monitoring schedule with robust operating characteristics across a wide range of stages and cohort sizes.

4. Advantages of PP design compared to standard multi-stage design:

More flexible, More efficient, More robust21

Presenter
Presentation Notes
The decision rule is: at the end of each stage of the trial, If this quantity is less than a small number pL, which means it is unlikely the response rate will be larger than p0, we may as well stop the trial at this point and reject the alternative hypothesis Otherwise, Continue to the next stage until Nmax So, based on this setting, Given p0, prior, n, Nmax, pL, pT: (1) Stopping boundary can be determined. (2) Type I and Type II errors can be calculated.

Operating characteristics of designs with type I and type II error rates ≤ 0.10, prior for p = beta(0.2,0.8), p0=0.2, p1=0.4

Simon’s Minimax/Optimal 2-Satger1/n1 r/Nmax PET(p0) E(N| p0) α β

Minimax 3/19 10/36 0.46 28.26 0.086 0.098Optimal 3/17 10/37 0.55 26.02 0.095 0.097Predictive Probability

θL θT r/Nmax PET(p0) E(N| p0) α β

--- --- na/35+ --- --- 0.126 0.093--- --- na/35++ --- --- 0.074 0.1160.001 [0.852,0.922]* 10/36 0.86 27.67 0.088 0.0940.011 [0.830,0.908] 10/37 0.85 25.13 0.099 0.0840.001 [0.876,0.935] 11/39 0.88 29.24 0.073 0.0920.001 [0.857,0.923] 11/40 0.86 30.23 0.086 0.0750.003 [0.837,0.910] 11/41 0.85 30.27 0.100 0.0620.043 [0.816,0.895] 11/42 0.86 23.56 0.099 0.0830.001 [0.880,0.935] 12/43 0.88 32.13 0.072 0.0740.001 [0.862,0.924] 12/44 0.87 33.71 0.085 0.0590.001 [0.844,0.912] 12/45 0.85 34.69 0.098 0.048

22

prior for p = beta(0.2,0.8)Simon’s Optimal PP

n Rej Region PET(p0)

Rej Region PET(p0)

10 0 0.107417 3 0.55 1 0.056321 2 0.066324 3 0.081527 4 0.084329 5 0.1010

31 6 0.0996

33 7 0.0895

34 8 0.0946

35 9 0.0767

36 10 0.55 10 0.86

α = 0.088 β = 0.094E(N | p0) = 27.67PET(p0) = 0.86

Simon’s Optimal: α = 0.095 β = 0.097E(N | p0) = 26.02PET(p0) = 0.55

Simon’s MiniMax:α = 0.086β = 0.098E(N | p0) = 28.26PET(p0) = 0.46

Stopping Boundaries for p0=0.20, p1=0.40, α= β= 0.10

23

Presenter
Presentation Notes
 

Number of Patients

Rej

ectio

n R

egio

n in

Num

ber o

f Res

pons

es

0 10 20 30

02

46

810

2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 32 34 36

02

46

810 Simon's MiniMax

Stopping Boundaries

Simon's Optimal

PP

24

Posterior Probability-Based DesignsBayesian Sequential Monitoring Designs for

Single-arm Trials with Multiple OutcomesThall et al., Statistics in Medicine, 1995.

Denote experimental tx by E and standard tx (historical data) by SProb. of response is θR and Prob. of tox is θTStop the trial if

Prob(θR,S + δR > θR,E ) > π∗ orProb(θT,S + δT < θT,E ) > π∗

δR (typically 0 ≤ δR ≤ 0.1) is an offset of minimal response improvement of E over S. δT (typically −0.1 ≤ δT ≤ 0) is an offset of maximal toxicity allowance of E over S. Otherwise, continue

E has lower response rate than S

E has higher toxicity rate than S

25

26

27

Response Stopping Boundaries:

0 1 6 201 5 7 232 8 8 263 11 9 294 14 10 305 17

Toxicity Stopping Boundaries:

3 3 8 173 4 9 194 6 9 205 8 10 225 9 11 246 11 11 257 13 12 277 14 12 288 16 13 30

The following are less (greater) -than-or-equal boundaries: a pair (n, m) means to stop if the number of responses (toxicity) after treating m patients is less (greater) than or equal to n.

28

Stopping for Futility, Efficacy and ToxicityDefine a set of elementary events, e.g.– A1=(CR, TOX), A2=(no CR, TOX), A3=(CR, no TOX),

A4=(no CR, no TOX); thenProbability model:

Prior:

Posterior:

29

Decision RuleChoose thresholds on posterior prob to correspond to clinically meaningful events

Sequential Stopping Rule

Computation: Evaluation πn(CR) and πn(TOX) requires integration wrt two independent Beta r.v.’s

30

ExampleStudy: Phase II trial of post-transplant prophylaxis for GVHD in bone marrow transplant (BMT), Endpoint: – Efficacy: GVHD within 100 days post transplant,

G = no GVHD within 100 days (``CR'')

– Toxicity: transplant rejectionT = transplant rejection within 100 days (``TOX'')

Elementary outcomes:

Prior:

Design:

31

Simulation

32

Adaptive Randomization (AR)(Equal or Adaptive) Randomization Is A Good Thing– Result in comparable study groups wrt known/unknown

prognostic factors– removes investigator bias in the treatment allocation– guarantees the validity of statistical tests

Baseline AR– Ensure balance in prognostic factors among treatment

groups– Based on baseline covariates (static)– Treatment allocation method

Biased coin, urn designPocock-Simon (1975), Minimization

Response (Outcome-based) AR– Allocate more patients in superior treatments and less

patients in inferior treatment based on the observed data – Play the winner - deterministic– Model-based - probabilistic 33

Response Adaptive RandomizationPlay-the-winner (Deterministic)

Model-based (Probabilistic)– Urn model, Bandit problem, Probability-based randomization

Goal: max. # of pts assigned to the superior arm (total # of successes)Advantage:– Treat more pts in the better treatments– Obtain more precise estimate for the better treatments

Disadvantage/Limitation– Imbalance results in loss of efficiency– Require response to be measured quickly 34

Two-Arm Adaptive Randomization (AR)Consider two treatments, binary outcomeFirst n pts equally randomized (ER) into TX1 and TX2After ER phase, the next patient will be assigned to TX1 with probability , where

Note that the tuning parameter– λ = 0, ER– λ = ∞, “play the winner”– λ = 0.5 or 1– λ = n / Nmax (Thall and Wathen, Europ J. Cancer, 2007)

Continue until reaching early stopping criteria or Nmax35

Equal Randomization (ER) ER + Adaptive RandomizationAR rate to TX 1=

Demo for more two-arm AR designs36

Randomized Two-Arm TrialFrequentist’s approach– Ho: P1 = P2 vs. H1: P1 < P2

P1 = 0.3, P2 = 0.5, α=.025 (one-sided), 1−β = .8N1 = N2 = 103, N = 206

Bayesian approach with adaptive randomization– Consider P1 and P2 are random; Give a prior distribution;

Compute the posterior distribution after observing outcomes– Randomize more patients proportionally into the arm with

higher response rate– At the end of trial,

Prob(P1 > P2) > 0.975, conclude Tx 1 is betterProb(P2 > P1) > 0.975, conclude Tx 2 is better

– At interim,Prob(P1 > P2) > 0.999, Stop the trial early, conclude Tx 1 is betterProb(P2 > P1) > 0.999, Stop the trial early, conclude Tx 2 is better

37

AR Comparisons (4)

No AR ARAR w/ Early

Stopping (Nmax=200)

AR w/ Early Stopping

(Nmax=250)

Ho H1 Ho H1 Ho H1 Ho H1

N1 100 100 100 46 98 42 122 46

N2 100 100 100 154 97 125 121 150

N 200 200 200 200 195 167 243 196

P(Tx1 Better) .02 0 .04 0 .05 0 .05 0

P(Tx2 Better) .03 .83 .04 .75 .05 .76 .05 .85

P(Early Stopping) .04 .34 .04 .34 .04 .44

P(rand. in arm 2) .50 .50 .50 .77 .50 .75 .50 .77

Overall Resp. .30 .40 .30 .45 .30 .45 .30 .45

38

Demo for Multi-Arm AR Designs

39

Biomarker-Based Adaptive DesignsIdentify prognostic and predictive markers for treatments– Prognostic markers

markers that associate with the disease outcome regardless of the treatment: e.g., stage, performance status

– Predictive markersmarkers which predict differential treatment efficacy in different marker groups: e.g., In Marker (−), tx does not workbut in Marker (+), tx works

Test treatment efficacy– Control type I and II error rates– Maximize study power for testing efficacy between txs– Group ethics

Provide better treatment to patients enrolled in the trial– Assign patients into the better treatments with higher probs– Maximize total number of successes in the trial– Individual ethics

40

BATTLE (Biomarker-based Approaches of Targeted Therapy for Lung Cancer

Elimination)

Patient Population: Stage IV recurrent non-small cell lung cancer (NSCLC) Primary Endpoint: 8-week disease control rate [DCR] 4 Targeted treatments14 Biomarkers200 patients20% type I error rate and 80% power

Zhou X, Liu S, Kim ES, Lee JJ. Bayesian adaptive design for targeted therapy development in lung cancer - A step toward personalized medicine (Clin Trials, 2008).

41

Histology (H&E)

DNA extraction and Mutation Analysis (EGFR, K-RAS, B-RAF)

EGFR Exon 19 Mutation K-RAS Codon 12 Mutation

Histology Sections FISH and Immunohistochemistry

RXR-α RXR-β RXR-γVEGF VEGFR-2

Biomarker Analysis in Core Biopsies for Targeted Therapy Response Prediction

Baseline BiopsyBiomarker assessment:1. EGFR mutation2. EGFR over exp/amplification

4. KRAS mutation5.6. VEGF expression7. VEGFR-2 expression8. RXRα cytoplasm expression

10. RXRβ cytoplasm expression

12. RXRγ expression13. Cyclin D1 expression14. Cyclin D1 amplification

BRAF mutation

EGFR FISH

3. EGFR increased copy number

9. RXRα nucleus expression

11. RXRβ nucleus expression

42

Erlotinib Vandetanib Erlotinib + BexaroteneSorafenib

Biomarker Profile and Adaptive Randomization

Enrollment into BATTLE Umbrella Protocol

Four Molecular Pathways Targeted in the BATTLE Program

Endpoint: Progression-free survival at 8 weeks –Disease Control Rate (DCR)

K-ras and/or B-raf VEGF and/or VEGFR1 + x x x 0.15

2 - + x x 0.2

3 - - + x 0.3

4 - - - + 0.25

5 - - - - 0.1

MGBiomarker

Fre quencyRXR and/or cyclin D1EGFR

43

Bayesian Hierarchical Probit Model

• Notation-- ith : subject, i=1 , ..., njk-- jth : treatment arm, j=1 , …, 4-- kth : marker group, k=1 , …, 5-- yijk: 8-week progression-free survival status: 0(no) vs 1(yes)-- zijk : latent variable-- μjk : location parameter-- φj : hyper-prior on μjk-- γjk : disease control rate (DCR)-- σ2,τ2 : hyper-parameters control borrowing across MGs

within and between treatments

• Probit model with hyper prior (Albert et al, 1993)

44

Computation of the Posterior Probability via Full Conditional Distribution – Gibbs SamplingThe random variables are generated from their complete posterior conditional distributions as follows.

The latent variable zijk is sampled from a truncated normal distribution centering at μjk.The full conditional distribution of μjk and φj are the linear combination of the prior distribution and the sampling distribution.

45

ER is applied in the first stage for model developmentAR will be applied after enrolling at least one patient in each (Treatment x MG) subgroup.Adaptively assign the next patient into the treatment arms proportional to the marginal posterior disease control rates. Randomization Rate (RR): proportional to the marginal posterior DCR.

set a minimum RR to 10% to ensuring a reasonable probability of randomizing pts in each armSuspend randomization of a treatment in a biomarker group if – Probability(DCR > 0.5 | Data) < 10%

Declare a treatment is effective in a biomarker group if – Probability(DCR > 0.3 | Data) > 80%

ˆ ˆ/jk wkw

γ γ∀∑

Equal Randomization (ER) Followed By Adaptive Randomization (AR)

46

Simulation Results, Scenario 1(with early stopping)

47

Presenter
Presentation Notes
The additional row “None” indicates the average number of patients who do not receive any treatments due to lack of efficacy in all treatments. For MGs 1-4, almost all patients receive treatment, but for MG 5, about 11% of patients receive no treatment. (Patients will be treated off protocol.) For effective treatments, there is very little chance that the treatment will be suspended early. However, for ineffective treatments, there is an approximately 60% chance they will be stopped early when the DCR is 30%.

Demo of Biomarker-Based Multi-Arm AR Designs

48

49

BATTLE Results: Disease Control in % (n)

EGFR KRAS VEGFRXR/

CycD1None Total

Erlotinib 35% (17) 14% (7) 40% (25) 0% (1) 38% (8) 34% (58)

Vandetanib 41% (27) 0% (3) 38% (16) NA (0) 0% (6) 33% (52)

Erlotinib + Bexarotene 55% (20) 33% (3) 0% (3) 100% (1) 56% (9) 50% (36)

Sorafenib 39% (23) 79% (14) 64% (39) 25% (4) 61% (18) 58% (98)

Total 43% (87) 48% (27) 49% (83) 33% (6) 46% (41) 46% (244)

Trea

tmen

ts

Marker Groups

entation: http://app2.capitalreach.com/esp1204/servlet/tc?cn=aacr&c=10165&s=20435&e=12587&&m=1&br=80&audio=false

Presenter
Presentation Notes
These are the results of the percent of patients achieving disease control Each cell has the percent as well as the number of patients The primary endpoint result is 46% overall of patients achieving disease control. We had assumed 30% based on historical trials Across the top we have the biomarker groups (again the bundled individual markers) Along the side are the treatments and their overall disease control Pointing out several areas Sorafenib had a good disease control of 58%, also had the highest number of patients enrolled (98) Sorafenib had a 39% DC in the EGFR marker group Very intriguing was the Sorafenib disease control in the KRAS marker group (79%)

Lessons Learned from BATTLEBiomarker-based adaptive design is doable! It is well received by clinicians and patients.Prospective tissues collection & biomarkers analysis provide a wealth of informationTreatment effect and predictive markers are efficiently assessed. Pre-selecting markers is not a good idea. We don’t know what are the best predictive markers at get-go.Bundling markers into groups, although can reduce total number of marker patterns, is not the best way to use the marker information either.AR should kicks in early & needs to be closely monitored.AR works well only when we have good drugs and good predictive markers. 50

Section 4.4: Adaptive Randomization“Bayesians don’t need/like randomization”

plays no role in calculating posterior probabilities(whereas crucial for frequentist inference)

we can control for prognosis-related covariates anyway

ethically difficult for physicians

patients willing to be randomized are inherently different

Our take: Randomization IS still essential:

ensures pt prognosis is uncorrelated with trt assigned

balances trt assignment within patient subgroups

we can’t control for unknown/unmeasured covariates!

BUT: Note that we needn’t randomize patients with equalprobabilities to all arms...

Bayesian Adaptive Methods for Clinical Trials – p. 90/100

Principles of Adaptive RandomizationBy “adaptive,” we mean a procedure that alterssomething based on on the results of the trial so far=⇒ implications for Type I error!

Here we focus on outcome-adaptive designs, notcovariate-adaptive designs that seek to balancecovariates across treatments

Basic idea: Treatment arms Ak having responseprobabilities θj , j = 1, . . . ,m. Given data y, randomize totreatment Ak with probability

rk(y) ∝ {p(θk = maxj

θj | y)}c for some c ≥ 0 (1)

c = 0 =⇒ equal randomization

might take c = n/2N where n is number of currentlyenrolled pts and N is maximum enrollment

Bayesian Adaptive Methods for Clinical Trials – p. 91/100

MDACC software package:ARWindows application for trials having up to 10 arms

outcomes may be either binary or time-to-event (TITE),though latter case currently restricted to exponentialsurvival with a conjugate prior

easy-to-read user’s guide

583 registered downloads between 2005 and 2009

Binary case: Assign Beta(αk, βk) priors to the θk by

choosing the (αk, βk) pairs directly

specifying either two quantiles or the mean and thevariance; AR can then “back out” (αk, βk)

Assuming xk positive and nk − xk negative (independent)responses =⇒ θk|y ∼ Beta(xk + αk , nk − xk + βk) as usual=⇒ used to define a variety of stopping rules...

Bayesian Adaptive Methods for Clinical Trials – p. 92/100

Algorithm used by ARStep 1. Early loser: If for some prespecified probability pL,

P (θk > θj 6=k|y) < pL ,

then arm k is declared a loser and is suspended.Normally pL is fairly small (say, 0.10 or less)AR permits an arm to return later

Step 2. Early winner: If for some prespecified probability pU ,

P (θk > θj 6=k|y) > pU ,

then arm k is declared the winner and the trial isstopped early.

Normally pU is fairly large (say, 1− pL for a two-armtrial)

Bayesian Adaptive Methods for Clinical Trials – p. 93/100

Algorithm used by AR (cont’d)Step 3. Final winner: If, after all patients have been evaluated,

for some prespecified probability p∗U ,

P (θk > θj 6=k|y) > p∗U ,

then arm k is declared the winner.If no treatment arm can meet this criterion, AR doesnot make a final selection.Normally p∗U < pU (say, between 0.70 and 0.90)

Step 4. Futility: If for some θmin and some prespecified p∗L,

P (θk > θmin|y) < p∗L ,

then arm k is declared futile and its accrual is stopped.

Reactivation of a futile arm is not permittedNormally p∗L is quite small (say, 0.10 or less)

Bayesian Adaptive Methods for Clinical Trials – p. 94/100

Example: Sensitizer TrialGoal: Evaluate ability of a “sensitizer” (given concurrentlywith another chemotherapeutic agent) to produce completeremission (CR) at 28 days post-treatment

Classical two-arm comparison of drug-plus-sensitizervs. drug alone ⇒ sample size near 100 – too large forour accrual rate of just 30/year!

Instead, use some prior information with a Bayesian ARdesign having

maximum patient accrual = 60

minimum randomization probability = 0.10

first 14 patients randomized fairly (7 to each arm)before AR begins

AR tuning parameter c = 1 (i.e., modest deviation fromequal randomization after the first 14 patients)

Bayesian Adaptive Methods for Clinical Trials – p. 95/100

Priors for the Sensitizer Trial

0.0 0.2 0.4 0.6 0.8 1.0

01

23

45

6

θ

density

Arm 1 (control)

Arm 2 (sensitizer)

0.0 0.2 0.4 0.6 0.8 1.0

01

23

45

6

θdensity

Arm 1 (control)

Arm 2 (sensitizer)

Left: “standard" priors; right, “conservative" priors (bothsd’s doubled)

Both Arm 1 priors have mean 0.55; both Arm 2 priorshave mean 0.75

Bayesian Adaptive Methods for Clinical Trials – p. 96/100

Control Parameters for Sensitizer TrialBegin with a “standard" rule that sets

early loser selection probability pL = 0.025

early winner selection probability pU = 0.975

final winner selection probability p∗U = 0.90

futility parameters θmin = 0.50 and p∗L = 0.05

Use AR to compare results from three different scenarios:

Scenario 1: true response rates of .55 in both groups(the “null" scenario),

Scenario 2: true response rates of .55 control, .70sensitizer (the “most likely" scenario), and

Scenario 3: true response rates of .55 control, .80sensitizer (the “optimistic" scenario).

Bayesian Adaptive Methods for Clinical Trials – p. 97/100

100 Simulated Trials, Standard PriorScenario 1 (Average Trial Length: 22.5 months)

True Pr Pr Pr(select Pr(stop # Patients

Arm (success) (select) early) early) (2.5%, 97.5%)

Arm1 0.55 0.01 0 0.11 19.6 ( 5, 38 )

Arm2 0.55 0.16 0.11 0 35.6 ( 8, 53 )

Scenario 2 (Average Trial Length: 16.4 months)

Arm1 0.55 0 0 0.55 10.1 ( 4, 22 )

Arm2 0.7 0.74 0.55 0 30.8 ( 4, 51 )

Scenario 3 (Average Trial Length: 10.8 months)

Arm1 0.55 0 0 0.89 7.01 ( 4, 16 )

Arm2 0.8 0.96 0.89 0 20.1 ( 4, 51 )

Good Type I error (17% total) and power (74%, 96%)

But fairly high sample sizes & fairly long trial lengths

Bayesian Adaptive Methods for Clinical Trials – p. 98/100

100 Simulated Trials, Conservative PriorScenario 1 (Average Trial Length: 21.0 months

True Pr Pr Pr(select Pr(stop # Patients

Arm (success) (select) early) early) (2.5%, 97.5%)

Arm1 0.55 0.19 0.05 0.15 26.2 ( 5, 45 )

Arm2 0.55 0.16 0.13 0.09 25.4 ( 4, 47 )

Scenario 2 (Average Trial Length: 18.1 months)

Arm1 0.55 0.04 0.02 0.39 15.2 ( 4, 44 )

Arm2 0.7 0.52 0.39 0.02 29.4 ( 4, 49 )

Scenario 3 (Average Trial Length: 14.3 months)

Arm1 0.55 0 0 0.63 10.6 ( 2, 26 )

Arm2 0.8 0.80 0.63 0 25.6 ( 3, 50 )

is more conservative; higher Type I error, lower power

still fairly low average sample sizes (36.2, 44.6)

Bayesian Adaptive Methods for Clinical Trials – p. 99/100

Summary

MD Anderson software available athttps://biostatistics.mdanderson.org/SoftwareDownload/

BCLM text-related software available at:http://www.biostat.umn.edu/∼brad/data3.html

BRugs package installation and further examples:http://www.biostat.umn.edu/∼brad/software/BRugs/

Related design site for binary and Cox PH models:www.biostat.umn.edu/∼brianho/papers/2007/JBS/prac_bayes_design.html

Thanks for your attention!

Bayesian Adaptive Methods for Clinical Trials – p. 100/100

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Hierarchical Commensurate Prior Modelsfor Adaptive Incorporation of

Historical Information in Clinical Trials

Brian P. Hobbs1, Bradley P. Carlin2,Sumithra Mandrekar,3 and Daniel Sargent3

1 Department of Biostatistics, University of Texas M.D. Anderson Cancer Center,Houston, TX

2 Division of Biostatistics, School of Public Health, University of Minnesota,Minneapolis, MN

3 Mayo Clinic, Rochester, MN

University of Chicago, Department of Health Studies,December 7, 2011

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionBackground

Using Historical Data

I Email 8 Sep 2008 from Dr. Telba Irony, FDA: “When we tryto borrow strength from only one historical study (be it acontrol group or a treatment group) ... [the results] becomeVERY sensitive to the hyperprior [on the variance parametersthat control the amount of borrowing].”

I Borrowing from historical data offers advantages:I reduced sample size (at least in control group) hence lower

cost and ethical hazard, plus higher power

but also disadvantages:I higher Type I error, plus a possibly lengthier trial if the

informative prior turns out to be wrong

I Thus what is needed is a recipe for how much strength toborrow from the historical data

I One possibility: “back out” this amount based on Type I errorand power considerations. This is often done, but tends todefeat the historical data’s original purpose!

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

Proposed Solution: Power Priors

I Introduced by Ibrahim and Chen (2000, Statistical Science)

I Let D0 = (n0, x0) denote historical data, suppose θ is theparameter of interest, and let L(D0|θ) denote the generallikelihood

I Suppose π0(θ) is the prior distribution on θ before D0 isobserved, the initial prior

I The conditional power prior on θ for the current study is thehistorical likelihood, L(D0|θ), raised to power α0, whereα0 ∈ [0, 1], multiplied by the initial prior:

π(θ|D0, α0) ∝ L(D0|θ)α0π0(θ) ,

I α0 is the power parameter that controls the “degree ofborrowing” from D0

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

Power Priors (cont’d)

I The power parameter, α0, “can be interpreted as a relativeprecision parameter for the historical data” (IC, 2000, p.48)

I Certainly apparent for normal data, x0iiid∼ N(θ, σ20), since

under a flat initial prior,

π0(θ|D0, α0) = N(x0, σ

20/(α0n0)

)I Think of α0n0 as the “effective” historical sample size

I Given current data, D = (n, x), the conditional posterior

q(θ|D,D0, α0) ∝ L(D0|θ)α0L(D|θ)π0(θ)

I α0 → 1, q(θ|D,D0, α0)→ approaches full borrowing from D0

I α0 → 0, q(θ|D,D0, α0)→ approaches no borrowing from D0

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

Power Priors (cont’d)

I We could fix α0 ∈ [0, 1] and assume consistency among D0

and D is known, but if this is not the case, models with poorfrequentist operating characteristics may result

I Choosing a hyperprior, π(α0), for α0 enables the data to helpdetermine probable values for α0

I Ibrahim-Chen (2000) propose joint power priors of form

π(θ, α0|D0) ∝ L(D0|θ)α0π0(θ)π(α0)

I Duan et al. (2006), Neuenschwander et al. (2009), andPericchi (2009) propose modified joint power priors (MPP)which respect the Likelihood Principle,

π(θ, α0|D0) ∝ L(D0|θ)α0π0(θ)∫L(D0|θ)α0π0(θ)dθ

π(α0)

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

Location Commensurate Power Priors (LCPP)

I We propose an adaptive modification of the MPP

I Let x0iiid∼ Normal(µ0, σ

20) and xi

iid∼ Normal(µ, σ2)

I Different parameters in historical and current group, µ0 and µ

I Extend hierarchical model to include parameter, τ , thatdirectly measures similarity of µ and µ0

I Construct prior for µ dependent upon µ0 and τ

I τ parametrizes commensurability (precision)

I Use information in τ to guide prior on α0

πLCPP(µ, α0, τ |x0) ∝

∫ [N(x0| µ0 , σ

20)]α0∫ [

N(x0| µ0 , σ20)]α0 dµ0

dµ0×N(µ|µ0 ,1

τ)×Beta(α0|τ a , 1)×p(τ)

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

LCPP for Single Arm Trial

I Formalize commensurate as µ0 near µ by adopting Normalprior on µ with mean µ0 and precision τ

I Beta(τ a , 1) prior on α0 for some a > 0

I τ close to 0 corresponds to very low commensurability, whilevery large τ implies the two datasets may arise from similarpopulations

I τ →∞, point-mass prior on α0 at 1

I τ → 0 discourages incorporation of historical information

I Requires a fixed, known sampling historical variance σ20 (MLE)

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

LCPP for Single Arm Trial (cont’d)

I Both α0 and τ inflate the “prior” variance

πLCPP(µ, α0, τ |x0) ∝ N(µ| x0 ,1

τ+

σ20

α0n0)× Beta(α0|τ a , 1)× p(τ)

I Prior on τ : Mixture of two gammas with mixing probabilityω = 1/2 and with hyperparameter a = 1/2 provides sufficientflexibility:

p(τ) ∝(ωGamma(τ | 1 , 10) + (1− ω)Gamma(τ | 3/2 ,

1

1000)

)

I Posterior obtained after multiplying by current likelihoodN(x| µ , σ2) and vague (say reference) prior on σ2

I q(σ2|x) ∝ IG(n2 ,

n2

[s2 + (x − µ)2

])

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

Extension to Linear ModelsFormulate linear model to borrow adaptively from the identical covariates

I Suppose that both trials identically measure p − 1 covariates

I Let X0 and X be n0 × p and n × p design matrices

I Suppose y0 ∼ Nn0(X0β0, σ2) and y ∼ Nn(Xβ + Zλ, σ2) where Z is

an n × r design matrix containing variables relevant only to thecurrent trial, and an indicator for new treatment

I Let D0 = (y0,X0, n0, p), and D = (y ,X ,Z , n, p, r)

I Assume flat prior for λ

I Let β0 = (XT0 X0)−1XT

0 y0

πLCPP(β, λ, σ2, α0, τ2|D0) ∝

∫Nn0

(y0| X0β0, σ

20 In0

)Np

(β| β0,

1

τIp

)dβ0 × Beta (α0| τ a, 1)× 1

σ2× p(τ)

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

Randomized Controlled Colorectal Cancer Trials

I Two successive randomized controlled colorectal cancer trials onsubjects with previously untreated metastatic colorectal cancer:

Saltz et al. (2000) trial randomized N0 = 683: May 1996 and May 1998

1. Irinotecan alone (arm A)

2. Irinotecan and bolus Fluorouracil plus Leucovorin (arm B; IFL)significantly longer progression free survival

3. Fluorouracil and Leucovorin (arm C; 5FU/LV) standard therapy

Goldberg et al. (2004) trial randomized N = 795: May 1999 and April 2001

1. Irinotecan and bolus Fluorouracil plus Leucovorin (IFL) regulatorystandard in March 2000

2. Oxaliplatin and infused Fluorouracil plus Leucovorin (FOLFOX) newregimen

3. Irinotecan and Oxaliplatin (IROX) new regimen

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

Randomized Controlled Colorectal Cancer Trials (cont’d)

I Longest diameter (ld) in cm of 1 to 9 tumors measured every 6weeks for the first 42 weeks or until a response

I Compare FOLFOX to IFL for average reduction in ld sum from BL

I Covariate adjustments for baseline ld sum, age, and AST in units/L

I Historical: arm B (IFL) from the Saltz trial, n0 = 171

I Current: IFL, n = 129, and FOLFOX in the Goldberg trial, n = 141

Ordinary linear regression fits to colorectal cancer data:

Historical data Current dataestimate 95% CI estimate 95% CI

Intercept 0.880 (−1.977, 3.738) −0.467 (−2.275, 1.341)BL Tumor Sum −0.232 (−0.310, −0.154) −0.397 (−0.453, −0.340)

Age −0.022 (−0.067, 0.022) 0.014 (−0.014, 0.041)AST −0.001 (−0.017, 0.015) 0.005 (−0.007, 0.017)

FOLFOX – – −0.413 (−1.017, 0.190)

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

Randomized Controlled Colorectal Cancer Trials (cont’d)

Historical IFL

Fre

quen

cy

−20 −15 −10 −5 0 5

010

2030

4050

60

Concurrent FOLFOX

Fre

quen

cy

−20 −15 −10 −5 0 50

1020

3040

Histograms of average change in ld tumor sum from baseline: historicalIFL (left), FOLFOX (right); note FOLFOX results are slightly better (morenegative).

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionIntro Commensurate Power Priors Linear Model Example

Randomized Controlled Colorectal Cancer Trial (cont’d)

LCPP fit to colorectal cancer data:

estimate 95% BCI

β (Intercept) 0.180 (–1.11, 1.42)β (BL Tumor Sum) –0.39 (–0.44, –0.33)

β (Age) 0 (–0.02, 0.02)β (AST) 0 (–0.01, 0.01)

λ (FOLFOX) –0.46 (−0.82,−0.10)α0 0.86 (0.44, 1.00)

I High posterior estimate for α0 ⇒ LCPP analysis incorporatesvirtually all of the historical data

I Conclude FOLFOX resulted in a significant reduction in average ldsum when compared to the IFL

I Consistent with those of Goldberg et al. (2004), who determinedFOLFOX to have better times to progression and response rates

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

Competing Non-Power Prior Approaches

Let x0iiid∼ Normal(µ0, σ

20) and xi

iid∼ Normal(µ, σ2)

1. Cauchy prior on µ centered at historical sample mean x0I πcau ∝ Cauchy(µ|median = x0, scale = γ)

2. Location Commensurate Prior (LCP)I Adaptive mechanism based solely on the commensurability

parameter τ :

πLCP(µ, σ2, τ |x0) ∝ N(µ| x0 ,1

τ+σ20

n0)× 1

σ2× p(τ)

I We use a mixture prior on τ , often with ω = 1/2:

p(τ) ∝(ωGamma(τ | 1 , 10) + (1− ω)Gamma(τ | 3/2 ,

1

1000)

)

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

Non-Power Prior Approaches (cont’d)

3. Location-Scale Commensurate Mixture Prior (LSCMP)

I Borrowing depends upon commensurability of both the locationand scale parameters

I q0(µ0, σ20 |x0) ∝ Normal(x0| µ0 , σ

20)×

(1σ20

)I Commensurability Priors

I N(µ| µ0 ,

)I IG

(σ2| A = γσ4

0 + 2,B = σ20(γσ4

0 + 1))

πLSCMP(µ, σ2, σ20 | x0 , ν , γ) ∝∫

q0(µ0, σ20 |x0)× N

(µ| µ0 ,

1

ν

)× IG

(σ2| A(σ0, γ) ,B(σ0, γ)

)dµ0

∝ N

(µ| x0 ,

n0 + νσ20

νn0

)× IG

(σ2| A ,B

)× IG

(σ20 |

n0 − 1

2,

n0σ20

2

)

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

Non-Power Prior Approaches (cont’d)

3. Location-Scale Commensurate Mixture Prior (cont’d)

I Fix values of ν = (ν1, ν0) and γ = (γ1, γ0) corresponding to highand low precision

I ν = (1012, (10σ20)−1)

I γ = (102, 5−1)

I Formulate mixture prior with fixed mixing probability θ such thatπ?(µ, σ|x0, ν, γ, θ) is proportional to

θπ(µ, σ2, σ20| x0 , ν1 , γ1) + (1− θ)π(µ, σ2, σ20| x0 , ν0 , γ0) ,

where fixing θ = 0.5 seems to work well.

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 0, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 0.2, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 0.4, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 0.6, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 0.8, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 1, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 1.2, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 1.4, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 1.6, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 1.8, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = s20 = 1; x = 2, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 0, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 0.2, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 0.4, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 0.6, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 0.8, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 1, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 1.2, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 1.4, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 1.6, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 1.8, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionEqual Sampling Variances Unequal Sampling Variances

−1 0 1 2 3

Historical DataPooled DataModified Power PriorCauchy PriorLocation Commensurate Power PriorLocation Commensurate PriorLocation−Scale Commensurate Mixture PriorCurrent Data

95% BCI for µ given n = 10, n0 = 30; s2 = 3, s20 = 1; x = 2, x0 = 1

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Conventional Random-effects Meta-analytic ApproachRandom-effects meta-analysisa assumes exchangeability:

µ0,1, ..., µ0,H , µ ∼ N(ξ, η2)

I between-study heterogeneity and within-study variability

I ξ and η2 characterize the population mean and between-studyvariance

I shrinkage parameter

B = σ2/(σ2 + η2),

weight placed on the prior mean ξ for the posterior mean µ

I denote unknown bias by ∆h = µ− µ0,h,

I parameter vector θ = (λ,∆, σ2, σ20,1, . . . , σ

20,H), and

Y = (y, y0,1, ..., y0,H) denote the collection of response data

I estimate of ξ is a weighted average of the observed historical andcurrent study effects, with weights1/(σ2

0,1/n0,1 + η2), ..., 1/(σ20,H/n0,H + η2), 1/(σ2/n + η2)

a = Spiegelhalter et al., 2004

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Common “noninformative” and “weakly-informative” priors for η2

prior form

uniform variancea p(η2) = U(0, a), a = 100

inverse gammaa p(η2) = Γ−1(ε, ε), ε = 0.001

uniform standard deviationa p(η) = U(0,√

a)

half-Cauchyb p(η) ∝ (η2 + b)−1, b = 25

uniform shrinkagec p(η2) ∝ σ2/{(σ2 + η2)2}, σ20,h = σ2,

a = Spiegelhalter et al., 2004; b = Gelman, 2006; c = Daniels, 1999

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

−10 −5 0 5 10

rela

tive

prio

r de

nsity

prior

2ηlog

unif. varinv. gammaunif. sdhalf−Cauchyunif. shrink

−10 −5 0 5 10

0.0

0.1

0.2

0.3

0.4

post

erio

r de

nsity

posterior, H = 1

2ηlog

unif. var (v=30)inv. gamma (404890)unif. sd (21)half−Cauchy (209)unif. shrink (8)

−10 −5 0 5 10

posterior, H = 3

2ηlog

unif. var (4.19)inv. gamma (0.11)unif. sd (1.30)half−Cauchy (1.37)unif. shrink (0.41)

Prior & posteriors for log(η2) under full homogeneity: ∆tr = 0, for n =180, n0 = 60, and σ2 = σ2

0,h = 1. Parens = stand dev on the scale of η2

∝∫

Y

{∫θ

η2p(η2,θ)L(Y|θ)dθ

}L(Y|θtr )dY

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Commensurate Prior: One Historical Study

I commensurate prior for µ (HCMS, 2011)

µ ∼ N(µ| µ0, 1/τ)

I µ is a “non-systematically biased” representation of µ0

I initial prior, p(µ0), characterizes info. before observing hist. data

I one-to-one relationship between τ and η2: τ = 1/(2η2)

I joint posterior: q(θ|τ, y, y0) ∝

N(µ| µ0, 1/τ)p(µ0)p(σ, σ0)n0∏j=1

N(y0j | µ0, σ20)

n∏i=1

N(yi | µ+ diλ, σ2)

I Pocock (1976) repeated analysis under several fixed values of 1/τ

I HCMS (2011) consider fully Bayesian approaches as well as powerprior formulations

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Commensurate Prior: One Historical Study (cont’d)

I HSC (2011) proceed with parametric empirical Bayesian inferenceby replacing hyperparameter τ in with its marginal MLE,

τ∗ = arg maxτ∈[lτ ,uτ ]

{m(Y|τ)} ,

restricted to the effective range of borrowing of strength

I Gaussian data, τ∗ ∈ [0.5/102, 0.5/0.052], or η ∈ [0.05, 10]

Our proposed empirical Bayes (EB) procedure:

I “underestimates” variability in θ given that posterior uncertainty inτ∗ is unacknowledged

I offers alternative, (perhaps more desirable) trade-off betweenborrowing of strength and bias

I reduce dimensionality in the numerical marginalization of θ with anormal approx. for non-Gaussian data

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Commensurate Prior: Multiple Historical Studies

I assume homogeneity among the hist. studies (or fixed degree ofheterogeneity)

I commensurate prior for µ cond. on the hist. population mean:

N(µ| ξ0, 1/τ)

Relationship between τ and η2 is more complicated

I denote v0,h = σ20,h/n0,h, and let v = σ2/(n −

∑di )

I τ−1 characterizes the meta-analytic between-study variability, plusthe diff. between the summed variability among the sample means,and the pop. mean when heterogeneity is estimated η2 versus whenfull homogeneity is assumed,

τ−1 = η2+

{1/(v + η2) +

H∑h=1

1/(v0,h + η2)

}−1−

(1/v +

H∑h=1

1/v0,h

)−1

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Ratios of the post. variances of λ versus under no borrowing

∆tr = 0 ∆tr = 0.5 ∆tr = 1H = 1 3 5 H = 1 3 5 H = 1 3 5

unif. var. 1.00 0.99 0.99unif. shrink 1.01 0.99 0.99

unif. sd 1.00 0.97 0.98half-Cauchy 1.03 0.97 0.97inv. gamma 1.03 0.92 0.95commens.

homog. 1.25 1.18 1.00

Absolute Bias: |E(λ|∆tr )− λtr |unif. var. 0.00 0.00 0.01

unif. shrink 0.00 0.00 0.01unif. sd 0.00 0.00 0.01

half-Cauchy 0.00 0.00 0.01inv. gamma 0.00 0.02 0.01commens.

homog. 0.00 0.18 0.32

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Ratios of the post. variances of λ versus under no borrowing

∆tr = 0 ∆tr = 0.5 ∆tr = 1H = 1 3 5 H = 1 3 5 H = 1 3 5

unif. var. 1.00 0.99 0.99unif. shrink 1.01 0.99 0.99

unif. sd 1.00 0.97 0.98half-Cauchy 1.03 0.97 0.97inv. gamma 1.03 0.92 0.95commens. 1.23 1.14 1.03

homog. 1.25 1.18 1.00

Absolute Bias: |E(λ|∆tr )− λtr |unif. var. 0.00 0.00 0.01

unif. shrink 0.00 0.00 0.01unif. sd 0.00 0.00 0.01

half-Cauchy 0.00 0.00 0.01inv. gamma 0.00 0.02 0.01commens. 0.00 0.09 0.03

homog. 0.00 0.18 0.32

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Ratios of the post. variances of λ versus under no borrowing

∆tr = 0 ∆tr = 0.5 ∆tr = 1H = 1 3 5 H = 1 3 5 H = 1 3 5

unif. var. 1.00 1.05 1.15 0.99 0.99unif. shrink 1.01 1.07 1.14 0.99 0.99

unif. sd 1.00 1.16 1.28 0.97 0.98half-Cauchy 1.03 1.17 1.28 0.97 0.97inv. gamma 1.03 1.24 1.33 0.92 0.95commens. 1.23 1.37 1.44 1.14 1.03

homog. 1.25 1.49 1.60 1.18 1.00

Absolute Bias: |E(λ|∆tr )− λtr |unif. var. 0.00 0.00 0.00 0.00 0.01

unif. shrink 0.00 0.00 0.00 0.00 0.01unif. sd 0.00 0.00 0.00 0.00 0.01

half-Cauchy 0.00 0.00 0.00 0.00 0.01inv. gamma 0.00 0.00 0.00 0.02 0.01commens. 0.00 0.00 0.00 0.09 0.03

homog. 0.00 0.00 0.00 0.18 0.32

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Ratios of the post. variances of λ versus under no borrowing

∆tr = 0 ∆tr = 0.5 ∆tr = 11 3 5 1 3 5 1 3 5

unif. var. 1.00 1.05 1.15 0.99 0.99 0.96 0.99 0.99 0.97unif. shrink 1.01 1.07 1.14 0.99 0.98 0.98 0.99 0.98 0.98

unif. sd 1.00 1.16 1.28 0.97 0.95 0.95 0.98 0.97 0.98half-Cauchy 1.03 1.17 1.28 0.97 0.97 0.97 0.97 0.98 1.00inv. gamma 1.03 1.24 1.33 0.92 0.94 0.95 0.95 0.98 0.99commens. 1.23 1.37 1.44 1.14 1.09 1.06 1.03 1.01 1.00

homog. 1.25 1.49 1.60 1.18 1.39 1.49 1.00 1.14 1.21

Absolute Bias: |E(λ|∆tr )− λtr |unif. var. 0.00 0.00 0.00 0.00 0.01 0.05 0.01 0.01 0.02

unif. shrink 0.00 0.00 0.00 0.00 0.03 0.06 0.01 0.02 0.03unif. sd 0.00 0.00 0.00 0.00 0.04 0.08 0.01 0.02 0.03

half-Cauchy 0.00 0.00 0.00 0.00 0.04 0.08 0.01 0.03 0.03inv. gamma 0.00 0.00 0.00 0.02 0.06 0.10 0.01 0.03 0.04commens. 0.00 0.00 0.00 0.09 0.13 0.11 0.03 0.04 0.02

homog. 0.00 0.00 0.00 0.18 0.33 0.38 0.32 0.69 0.81

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionLogistic Generalized Linear Time-to-Event Mixed Simulations

Extension to Binary RegressionFormulate commensurate power prior for binary outcomes

I Historical data y0i ∼ Ber [π0(X0i )]; Current data yi ∼ Ber [π(Xi , di )]where di is the treatment indicator

I Location commensurate prior for binary regression followsproportional to

n0∏i=1

Ber (y0i | π(x0i ))× N

(β |β0,

1

τ

)× p(τ) ,

where we use p(τ) to bound τ away from 0 or ∞.

I Can’t integrate out β0 analytically, so instead just multiply bycurrent data likelihood and normalize via MCMC...

I Can choose probit link, π0(X0) = Φ(X0β0) andπ(X , d) = Φ(Xβ + dλ) ⇒ closed form full conditionals!

I Can instead pick logit link, π(x0) =(1 + e−x0β0

)−1 ⇒ requiresMetropolis-Hastings

I Example: Adaptively incorporating nonrandomized HIV trial arms!

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionLogistic Generalized Linear Time-to-Event Mixed Simulations

Extension to Generalized Linear Models

I Assume an initial flat prior on β0, and use the “Bayesian CentralLimit Theorem” with the historical likelihood to obtain

β0·∼ N

(β0, Σ0

),

where β0 is the historical MLE for β0, and Σ0 is the inverse of thehistorical observed Fisher information matrix

I Specifying a flat prior for λ, a N(β0,1τ Ip) prior for β, and integrating

out β0 again leads to joint location commensurate prior of

p(β, λ, τ |y0) ∝ Np

(V−1M,

1

τV−1

)p(τ)

I The joint posterior follows by multiplying by the current datalikelihood, which yields intractable full conditionals. Here we useMetropolis-Hastings (NOT the BCLT again, which would requirefixed β and λ in the Fisher information matrix)

I Primary target application: formulation for survival outcomes...

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionLogistic Generalized Linear Time-to-Event Mixed Simulations

Time-to-Event ResponseHistorical, concurrent data are triples (t0j , δ0j ,X0j) for j = 1, ..., n0 and

(ti , δi ,Xi ) for i = 1, ..., n; where ts are the observed (possibly censored)failure times, δs are noncensoring indicators, and X0j and Xi are rowvectors of p covariates associated with historical subject j and concurrentsubject i .

I Let f (t0j) and f (ti ) denote survival time densities with survivalfunctions F (t) and F (t0)

I Adopt a log-linear model: y0 = log(t0) = X0β0 + σ0e0 andy = log(t) = Xβ + dλ+ σe, where e0 = (y0 − X0β0)/σ0 ande = (y − Xβ − dλ)/σ. The likelihoods follow as

L0 (β0, σ0|y0) ∝n0∏j=1

[1

σ0f (e0j)

]δ0jF (e0j)

1−δ0j

and

L (β, σ|y) ∝n∏

i=1

[1

σf (ei )

]δiF (ei )

1−δi

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionLogistic Generalized Linear Time-to-Event Mixed Simulations

Time-to-Event Response (cont’d)

I Weibull regression occurs when e0 and e follow the extreme valuedistribution, f (u) = exp [u − exp (u)].

I We consider a location-scale commensurate approach, so borrowingdepends upon commensurability among σ0 and σ, as well as β0 andβ.

I LSCP follows from general theory (2 slides back) as

p(θ, λ, τ |y0) ∝ Np+1

(Λ−1Q,

1

τΛ−1

)p(τ) ,

where Q =(

Ψ0 + τ Ip+1

)−1Ψ0θ0, Λ = Ip+1 − τ

(Ψ0 + τ Ip+1

)−1,

and Ψ0 = Ψ0(θ0) is the observed Fisher information matrix. Theposterior is then proportional to the product of this LCPP and theconcurrent data likelihood, as usual.

I Note that the exponential model is a special case where σ = 1.

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionLogistic Generalized Linear Time-to-Event Mixed Simulations

Extension to General/Generalized Mixed ModelsConsider the Gaussian case for now (non-Gaussian also feasible):

I historical data: y0ij , i = 1, . . . , n0j , j = 1, . . . ,m0; arrange in avector y0 and model as y0 = X0β0 + Z0u0 + ε0, where

E

(u0

ε0

)=

(0

0

)and Cov

(u0

ε0

)=

(G0 00 R0

)I current data: uses similar notation: y = Xβ + dλ+ Zu + ε, where

d is a N × 1 indicator of treatment.

I The location commensurate prior emerges as proportional to

Np

(β V−1M,

1

τV−1

)× Nm

(u 0, σ2

uIm)× p(λ, σε, σu, τ),

where M =(

XT0 Σ−1X0 + τ Ip

)−1XT0 Σ−1X0β0,

V = Ip − τ(

XT0 Σ−1X0 + τ Ip

)−1, and β0 is the BLUE.

I As usual, multiply by current data likelihood and normalize; fullconditionals are normal and inverse Wishart; non-Gaussian caserequires Metropolis-Hastings

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionLogistic Generalized Linear Time-to-Event Mixed Simulations

Regression Model Simulations

Check frequentist OCs of the linear, exponential, & Weibull models:

E [y0] = µ0 and E [y ] = µ0 + dλ,

where d is treatment indicator and µ0 & µ are intercepts for controls

I Let ∆ = µ0 − µ, & spz λ > 0 indicates positive treatment effect

I ∆ > 0 implies hist. performed better than current controlsI ∆ < 0 implies hist. performed worse than current controls

I Type I error (λ = 0), power, & 95% post. CI interval coverage werecomputed by sampling y0 and y for true fixed values of ∆ and λ

I Use 95% posterior CI to test the null hypothesis H0 : λ = 0

I Below, Weibull sims compare commensurability among shapeparameters, through ratio ω = σ0/σ, when we set ∆ = 0

I Compare commensurate priors to models that ignore (no borrowing)& fully incorporate (full borrowing) the historical data

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionLogistic Generalized Linear Time-to-Event Mixed Simulations

∆ = −1 ∆ = 0 ∆ = 1

0.0 0.2 0.4 0.6

0.0

0.2

0.4

0.6

0.8

1.0

λ0.0 0.2 0.4 0.6

0.0

0.2

0.4

0.6

0.8

1.0

λ0.0 0.2 0.4 0.6

0.0

0.2

0.4

0.6

0.8

1.0

λ

0.0 0.2 0.4 0.6

0.0

0.2

0.4

0.6

0.8

1.0

λ0.0 0.2 0.4 0.6

0.0

0.2

0.4

0.6

0.8

1.0

λ0.0 0.2 0.4 0.6

0.0

0.2

0.4

0.6

0.8

1.0

λ

Power curves for the full borrowing, commensurate prior, and “no borrowing”models based on the 95% posterior CIs for λ. The top row shows results forthe Gaussian linear model, where µ0 = 1, σ2

0 = σ2 = 1, n0 = 100, and n = 50;bottom row the exponential model, for µ0 = 2, n0 = 200, and n = 100.

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionLogistic Generalized Linear Time-to-Event Mixed Simulations

Gaussian Exponential Weibull

−1.5 −0.5 0.5 1.5

0.0

0.2

0.4

0.6

0.8

1.0

∆−1.0 −0.5 0.0 0.5 1.0

0.0

0.2

0.4

0.6

0.8

1.0

∆0.5 1.0 1.5 2.0 2.5 3.0

0.0

0.2

0.4

0.6

0.8

1.0

ω

−1.5 −0.5 0.5 1.5

0.90

1.00

1.10

1.20

∆−1.0 −0.5 0.0 0.5 1.0

0.70

0.80

0.90

∆0.5 1.0 1.5 2.0 2.5 3.0

0.4

0.8

1.2

1.6

ω

Sim. results for full borrowing, commensurate prior, and “no borrowing” models.Top row plots average 95% posterior coverage probabilities for λ. Bottom rowplots average length of 95% posterior CIs for λ. X-axes correspond to intercepts,∆ = µ0 − µ (left & center), or the scale ratio, ω = σ0/σ where ∆ = 0 (right).

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Adaptive Randomization

Friedman, Furberg, and DeMets (1998, p.69) broadly refer torandomization procedures that adjust the allocation ratio as the studyprogresses as adaptive. Commonly used to

I balance prognostic factors among the intervention arms

I assign patients to the better performing regimen in early phase trials

Commensurate prior models for controlled trials naturally advocate arandomization scheme that is “optimal” with respect to the amountstrength borrowed from the historical controls

I Use more of the new patients to learn about the efficacy and safetyprofile of the new intervention

I Adjust prob. of allocating next patient to new intervention accordingto the commensurability of the historical and current controls

I Evaluate sequentially using repeated fitting

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Optimal-Balanced Randomization

I Define “allocation probability balance” as a function of the effectivesample size of the historical controls

I Morita, Thall, and Muller (2008) define prior effective sample size(ESS) of a parametric prior for non-adaptive models

I We expand their method for computing the prior effective samplesize of the intercepts & identically measured covariates (previouslyβ)

I Referred to as the effective number of historical controls, EHC

I First find the posterior distribution of the commensurabilityparameter, τ ; then EHC is essentially ESS is computed for

p(β, λ, τ |y0) ∝ Np

(V−1M,

1

τV−1

)p(τ),

where τ replaced with its posterior median, τ

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Optimal-Balanced Randomization (cont’d)

I The new treatment allocation probability, or balance function, forthe jth+1 new patient follows as

δj =Cj + EHCj

Tj + Cj + EHCj,

where Tj and Cj denote the number of subjects randomized to newtreatment and control following the jth enrollment

I This imposes information balance by encouraging optimal use ofnew patients relative to amount of incorporated prior information

I We suggest adjusting the allocation probability in blocks after aninitial period where δj is fixed at 1/2

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Adaptive Randomization Example

Flexible Initial Retrovirus Suppressive Therapies (FIRST) trial:

I This was a large, long-term, randomized, prospective comparison ofthree different antiretroviral strategies in highly active antiretroviraltherapy-naive, HIV-1-infected persons (MacArthur et al., 2001)

I Patients within all three strategies were also assigned one or twonucleoside reverse transcriptase inhibitors (NRTIs)

I The three strategies for initial treatment were compared forlong-term virological and immunological durability and safety, for thedevelopment of drug resistance, and for clinical disease progression

I Before randomization, patients within the two NNRTIs arms weregiven the option of preselecting the NNRTI drug, either nevirapine(NVP) or efavirenz (EFV), or allowing an additional randomizationto NVP or EFV

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Randomization Design

Randomization to FIRST

PI Strategy(PI + NRTIs)

(data not used)

NNRTI Strategy(NNRTI + NRTIs)

N=409

NNRTI

substudy

Randomization

Prescribed

NVP

(N=100)

Prescribed

EFV

(N=211)

NVP

(N=50)

EFV

(N=48)

3-Class Strategy(PI + NNRTI + NRTIs)

N=403

NNRTI

substudy

Randomization

Prescribed

NVP

(N=137)

Prescribed

EFV

(N=161)

NVP

(N=51)

EFV

(N=54)

Outline of FIRST design and randomization for eligible subjects (Berg-Wolf et

al., 2006).

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Adaptive Randomization Example (cont’d)

I Current trial consists of data from the randomized arms of FIRST,

1. EFV (n = 99) is the novel intervention and2. NVP (n = 104) the control therapy

I Historical controls consist of the NVP obs. arm (n = 237)

I Compare the prob. of virological suppression (HIV RNA < 50copies/mL) under EFV and NVP at 32 weeks

I Use the commensurate prior probit regression model

Technical details:

Φ−1[π0] = µ0 and Φ−1[π(d)] = µ+ dλ

,

W0jj =φ(X0β0)2jj

Φ(X0β0)jj(

1− Φ(X0β0)jj) ,

where Φ() is the standard normal c.d.f. & φ(.) is the standard normal p.d.f.

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Adaptive Randomization Example (cont’d)

Simulating the adaptive randomization method proceeds iteratively:

I assign the jth+1 new patient to NVP or EFV with probability, δj

I generate response for several fixed values of ∆ = µ0 − µI λ is fixed at 0.23 (MLE obtained from fitting the real current data

from FIRST)

I equal allocation to NVP & EFV for first 80 assignments

I after randomizations 80, 100, 120, and 160, EHC is computed andδj is updated

I record EHC and δj after each randomization block, & total assignedto each group after all n = 203 patients are randomized

I ∆ = −0.221 is MLE obtained from fitting the real current datafrom FIRST

Intro Power Priors Alternatives Metaanalysis GLMs Rando Discussion

Optimal-Balanced Randomization Simulation

Simulation Averages for adaptive randomization to EFV, for n = 203, wheren0 = 237, Ratio=δ/(1− δ):1.

Rz ∆ = −2.25 ∆ = −1.5 ∆ = −0.75 ∆ = −0.221 ∆ = 0EHC ,Ratio EHC ,Ratio EHC ,Ratio EHC ,Ratio EHC ,Ratio

80 1, 1.03:1 18, 1.42:1 177, 5.43:1 235, 6.86:1 237, 6.92:1

100 1, 1.02:1 15, 1.23:1 168, 3.74:1 235, 4.82:1 237, 4.85:1

120 1, 1.01:1 11, 1.11:1 164, 2.95:1 235, 3.80:1 237, 3.82:1

160 1, 1.01:1 5, 1.01:1 144, 2.02:1 235, 2.74:1 237, 2.75:1

NVP, EFV NVP, EFV NVP, EFV NVP, EFV NVP, EFV203 101, 102 99, 104 77, 126 66, 137 65, 138

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionConclusion

Is all of this “legal”?

I X-L Meng: NO: Agreement between the historical andconcurrent data does not necessarily imply they are studyingthe same thing! External validation/information is alwaysrequired!

I (Fairly hardcore) frequentist referee: Not yet: One needs to“evaluate theoretical properties, such as Bernstein-von Misestype results [showing asymptotic equivalence between Bayesand frequentist results] to provide some kind of universalassurances and/or insight beyond simulations into when gainscan be expected.”

I BP Carlin: Yes: Point taken, but our goal here was never to“match” frequentist results, and moreover the frequentistsimulations we have carried out speak for themsselves and are“Bayesianly justified” in the sense of Rubin (1984)!

Intro Power Priors Alternatives Metaanalysis GLMs Rando DiscussionConclusion

ConclusionI Adaptive borrowing is consistent with recent arguments on

behalf of the ease and desirability of adaptivity in Bayesianclinical trials generally; c.f. new Chapman and Hall textbookby Berry, Carlin, Lee, and Muller (2010)!

Papers related to this work:

I Hobbs, B.P., Carlin, B.P., Mandrekar, S., and Sargent, D.J.(2011). Hierarchical commensurate and power prior modelsfor adaptive incorporation of historical information in clinicaltrials. Biometrics, 67, 1047–1056.

I Hobbs, B.P., Sargent, D.J., and Carlin, B.P. (2010).Commensurate priors for incorporating historical informationin clinical trials using general and generalized linear models.Under revision for Bayesian Analysis.

I Hobbs, B.P. and Carlin, B.P. (2012). Optimal-balancedrandomization for controlled clinical trials that incorporatehistorical controls. Manuscript in process.

Bayesian Hierarchical Modeling

for Detecting Safety Signals

in Clinical Trials

Amy Xia, Amgen, Inc.

Haijun Ma, Amgen, Inc.

Brad Carlin, University of Minnesota

Medtronic Statistics Conference

Minneapolis, MN, October 4, 2011

Outline

• Introduction

• Why Are Bayesian Models Helpful?

• Three Stage Bayesian Hierarchical Models

• An Example

• Simulation Study

• Closing Remarks

Three-Tier System for Analyzing

Adverse Events in Clinical Trials

• Tier 1 AEs -- events for which a hypothesis has

been defined

• Tier 2 AEs -- events that are not pre-specified

and “common”

• Tier 3 AEs – events that are not pre-specified

and infrequent

Gould 2002 & Mehrotra and Heyes 2004

SPERT White Paper -- Crowe et al 2009

Multiplicity Issue in Detecting

Signals Is Challenging

• Detection of safety signals from routinely collected, not pre-specified AE data in clinical trials is a critical task in drug development

• Multiplicity issue in such a setting is a challenging statistical problem – Without multiplicity considerations, there is a potential

for an excess of false positive signals

– Traditional ways of adjusting for multiplicity such as Bonferroni may lead to an excessive rate of false negatives

– The challenge is to develop a procedure for flagging safety signals which provides a proper balance between „no adjustment‟ versus „too much adjustment‟

Bayesian Work in Signal Detection

• Spontaneous adverse drug reaction reports

– Gamma Poisson Shrinker (GPS) on FDA AERS database (DuMouchel,1999)

– Bayesian Confidence Propagation Neural Network (BCPNN) on WHO database (Bate, et al. 1998)

• Clinical trial safety (AE) data

– Bayesian hierarchical mixture modeling (Berry and Berry, 2004)

“Safety assessment is one area

where frequentist strategies have

been less applicable. Perhaps

Bayesian approaches in this area

have more promise.”

-- Chi, Hung, and O‟Neill;

Pharmaceutical Report, 2002

Proposed Bayesian Approach

1. A three-level hierarchical mixture model for binary responses was constructed following the work by Berry & Berry (2004)

2. Further extended to a hierarchical Poisson mixture model, to account for different exposure/follow-up times between patients

3. Provided guidance on how to choose a signal detection threshold to achieve acceptable false discovery rate (FDR) and power for meaningful risk sizes

4. Extended the work to the setting of meta-analysis and incorporating individual patient-level data.

5. Implemented the above models with available software – WinBUGS for model implementation

– S-Plus graphics for inference

Considerations Regarding Whether

to Flag an Event

• Actual significance levels

• Total number of types of AEs

• Rates for those AEs not considered for flagging

• Biologic relationships among various AEs

1st two are standard considerations in the

frequentist approach. The 2nd two are not,

but relevant in the Bayesian approach

-- Berry and Berry, 2004

Example of the Medical Dictionary for

Regulatory Activities (MedDRA) Hierarchy

System Organ Class (SOC)

Infections and Infestations

High Level Group Term (HLGT)

Viral infectious disorders

High Level Term (HLT)

Influenza Viral Infections

Preferred Term (PT)

Influenza

Lowest Level Term (LLT)

Flu Syndrome

Reported AE verbatim

Flu Syndrome

Why Are Bayesian Models Helpful?

• Bayesian hierarchical models allow for explicitly modeling AEs with the existing coding structure (eg, SOC and PT in MedDRA) – AEs in the same SOC more likely to be similar within than across

SOCs.

– Allow for this possibility, but does not impose it, depending on the actual data

– Flexible in modeling SOC/PT, HLT/PT or even the full hierarchy

• In fact, clinical and safety people would (informally) consider the similarity of the AEs, say, within SOCs when they review AE tables – For example, if differences in several CV events were observed,

then each would be more likely to be causal than if differences came from medically unrelated areas (eg, skin, neurological, thrombosis, cancer)

• Bayesian hierarchical modeling allows a scientific, explicit, and more formal way to take it into consideration

An Example

• Pooled data from four double-blind placebo-controlled studies on drug X

• Sample size: Nt = 1245 and Nc = 720

• Reported AEs are coded to 465 PTs under 24 SOCs

Notations

• SOC b=1,…B and PT j=1,…kb

• Data: For AEbj – Treatment group: Ybj incident events in Nt patients

– Control group: Xbj incident events in Nc patients

r = subject incidence (%)

n/E = exposure-adjusted event rate (per subject-year)

* Fisher‟s 2-sided exact test unadjusted p-value <=0.05 with higher risk on treatment arm.

Data from the Example

b j SOC Preferred Term

Ctrl

n

Trt

n

Ctrl

r (%)

Trt

r (%)

Ctrl

n/E

Trt

n/E

1 1

Blood and Lymphatic System

Disorders Lymphadenopathy 6 10 0.83 0.80 0.04 0.03

… … … …

8 8

General Disorders and

Administration Site

Conditions Fatigue* 9 37 1.25 2.97 0.06 0.13

8 9 Feeling Cold 1 0 0.14 0.00 0.01 0.00

8 10 Feeling Hot 1 3 0.14 0.24 0.01 0.01

… … … … …

11 34 Infections and Infestations Herpes Simplex* 3 19 0.42 1.53 0.02 0.07

… … …

11 64 Sinusitis* 12 46 1.67 3.69 0.08 0.16

… … … …

12 12

Injury, Poisoning and

Procedural Complications Excoriation* 0 8 0.00 0.64 0.00 0.03

… … … … …

22 15

Skin and Subcutaneous

Tissue Disorders Ecchymosis* 0 12 0.00 0.96 0.00 0.04

… … … … …

Logistic Regression Models for

Subject Incidence (Berry & Berry 2004)

Ybj~Binom(Nt , tbj ); Xbj~Binom(Nc , cbj )

where tbj and cbj are event rates for AEbj in the

treatment and control groups, respectively

logit(cbj )=λbj; logit(tbj )=λbj + θbj ,

Note that θbj = log(ORbj)

Model 1a: Logistic Regression Model

with Normal Prior on log-OR

• Stage 1 priors λbj ~N(μλb , σ2

λb ); θbj ~N(μθb , σ2

θb );

• Stage 2 priors μλb~N(μλ0, σ2

λ0 ); σ2

λb ~IG(αλ, βλ);

μθb ~N(μθ0, σ2

θ0 ); σ2

θb ~IG(αθ, βθ)

• Stage 3 priors μλ0~N(μλ00 , σ2

λ00 ); σ2

λ0 ~IG(αλ00, βλ00)

μθ0~N(μθ00 , σ2

θ00 ); σ2

θ0 ~IG(αθ00, βθ00)

Hyperparameters μλ00, σ2

λ00, αλ00, βλ00, μθ00, σ2

θ00, αθ00, βθ00, αλ, βλ ,

αθ, βθ are fixed constants

Model 1b: Logistic Regression Model

with Mixture Prior on log-OR

• Same likelihood and mean structure

• Same priors for control group parameters

• Mixture prior for log(OR): point mass at 0 + normal

distribution

• Stage 1 prior: θbj ~pb δ(0) + (1-pb )N(μθb , σ2

θb );

• Stage 2 prior: pb ~Beta(αpb, βpb);

• Stage 3 prior: αpb~exp( ξα )I(αpb >1);

βpb~exp( ξβ )I(βpb >1)

This mixture allows a point mass on equality of the

treatment and the control rates because many AEs may

be completely unaffected by treatment

Model 1c: Non-hierarchical One-stage

Logistic Regression Model with

Mixture Prior on log-OR

• Simply the 1st stage of three-level hierarchical mixture

model

• Same likelihood and mean structure

• No information is borrowed across AEs within the same

SOC , i.e. the PTs are treated independently:

– λbj ~N(0 , 102);

– Mixture prior for log(OR): point mass at 0 + normal distribution

• Prior: θbj ~0.5 δ(0) + 0.5N(0 , 102 );

• With vague prior information, this model delivers similar

results as frequentist approaches

Model Selection - DIC (Spiegelhalter 2002)

• Hierarchical models with mixture priors are

preferred based on their smaller DIC values

Dbar pD DIC

Model 1a 2604.86 382.09 2986.95

Model 1b 2644.86 324.03 2968.89

Model 1c 2574.86 602.38 3177.24

• Deviance Information Criterion (DIC)

DIC = Dbar (fit) + pD (effective # of parameters)

Inference

• AEbj is flagged if

– Pr( θbj > c | Data) > p, where θbj is OR in Binomial

models

– Pr( (tbj - cbj ) > d | Data) > p, where tbj - cbj is risk

difference (RD)

– c, d and p are all prespecified constants

Examples

– c=1 and p = 0.8 means that the given AE type is flagged if the

posterior exceedance probability of the true OR being > 1 is more

than 80%

– d=0.02 and p = 0.8 means that the given AE type is flagged if the

posterior exceedance probability of the true RD being > 2% is

more than 80%

Inferences of Binomial Hierarchical Model

with Mixture Prior (Model 1b)

Posterior Exceedance Probability for:

SOC PT Unadjusted

p-value OR>1.0 OR>1.2 OR>2 RD>2% RD>5%

General Disorders & Administration Site Conditions

Fatigue .019 .56 .55 .32 .10 .00

Infections and Infestations

Herpes Simplex .025 .53 .51 .36 .00 .00

Sinusitis .012 .70 .69 .42 .28 .00

Injury, Poisoning & Procedural Complications

Excoriation .030 .30 .28 .18 .00 .00

Skin & Subcutaneous Tissue Disorders

Ecchymosis .005 .54 .52 .44 .00 .00

OR = odds ratio (drug:placebo), RD = risk difference (drug - placebo)

Fisher's Exact Test 2-sided P-values

Raw Risk Difference(%), Treatment - Placebo

- lo

g10

(F

ish

er

exa

ct

p-v

alu

e)

-0.01 0.0 0.01 0.02

0.0

0.5

1.0

1.5

2.0

2.5

Fatigue

Herpes Simplex

Sinusitis

Excoriation

Ecchymosis

Blood and Lymphatic Syste...Cardiac DisordersCongenital, Familial and ...Ear and Labyrinth Disorde...Endocrine DisordersEye DisordersGastrointestinal Disorder...General Disorders and Adm...Hepatobiliary DisordersImmune System DisordersInfections and Infestatio...Injury, Poisoning and Pro...InvestigationsMetabolism and Nutrition ...Musculoskeletal and Conne...Neoplasms Benign, Maligna...Nervous System DisordersPsychiatric DisordersRenal and Urinary Disorde...Reproductive System and B...Respiratory, Thoracic and...Skin and Subcutaneous Tis...Surgical and Medical Proc...Vascular Disorders

Binomial Hierarchical Model with Mixture Prior

OR posterior means

P(O

R >

1|D

ata

)

0.5 1.0 1.5 2.0 2.5

0.0

0.2

0.4

0.6

0.8

1.0

Lymphadenopathy Ear Pain Tinnitus

Vertigo

Vertigo Positional Cataract

Conjunctival Haemorrhage

Conjunctivitis

Conjunctivitis Allergic Diplopia

Dry Eye

Eye Haemorrhage Eye Inflammation Eye Irritation Eye Pruritus Eyelid Oedema Vision Blurred Abdominal Distension

Abdominal Pain Abdominal Pain Upper

Diarrhoea Dry Mouth

Dyspepsia

Flatulence Food Poisoning

Gastrointestinal Inflammation Gastrooesophageal Reflux Disease

Nausea Stomach Discomfort Toothache

Fatigue

Feeling Hot

Injection Site Bruising

Injection Site Haemorrhage

Malaise

Hypersensitivity

Seasonal Allergy

Abdominal Infection Acute Sinusitis Appendicitis

Bronchitis

Bronchitis Chronic

Candidiasis Cat Scratch Disease Cellulitis Staphylococcal Cervicitis Condyloma Acuminatum

Cystitis

Diverticulitis

Ear Infection

Erysipelas Eyelid Infection

Folliculitis

Fungal Infection

Furuncle

Gastroenteritis Viral

Genital Infection Female Gingival Infection

Herpes Simplex

Herpes Virus Infection Herpes Zoster

Impetigo

Infected Insect Bite

Infected Sebaceous Cyst Infectious Mononucleosis

Influenza

Labyrinthitis Laryngitis

Lower Respiratory Tract Infection Mastitis

Nasopharyngitis

Onychomycosis

Oral Candidiasis Oral Pustule

Otitis Externa Otitis Media

Pharyngitis

Pneumonia Postoperative Infection Pulpitis Dental Puncture Site Infection Rash Pustular

Rhinitis

Sialoadenitis

Sinusitis

Skin Infection

Subcutaneous Abscess Tinea Cruris Tinea Infection Tinea Pedis

Tinea Versicolour Tonsillitis

Tooth Abscess Tooth Infection

Upper Respiratory Tract Infection

Vaginal Candidiasis

Vaginal Infection

Vaginal Mycosis

Viral Infection

Viral Pharyngitis

Viral Upper Respiratory Tract Infection

Animal Bite Contusion

Excoriation

Fall Foot Fracture Hand Fracture Muscle Strain

Pain Trauma Activated

Road Traffic Accident Skin Laceration Sunburn Thermal Burn Tooth Injury Wound

Blood Cholesterol Increased

Blood Glucose Increased

Blood Pressure Increased

Body Temperature Increased Weight Decreased Weight Increased Decreased Appetite Back Pain

Myalgia

Shoulder Pain Tendonitis

Basal Cell Carcinoma

Fibrous Histiocytoma

Seborrhoeic Keratosis Skin Papilloma

Squamous Cell Carcinoma of Skin Burning Sensation

Carotid Artery Stenosis Carpal Tunnel Syndrome Cerebrovascular Accident Cerebrovascular Insufficiency Disturbance in Attention

Dizziness

Dizziness Postural

Dysgeusia Facial Palsy

Headache

Hypersomnia Hypertonia Hypoaesthesia

Lethargy Memory Impairment

Migraine

Nerve Compression

Neuropathic Pain

Paraesthesia

Restless Legs Syndrome Sciatica

Sensory Disturbance Somnolence Syncope

Syncope Vasovagal

Tension Headache

Agitation Alcoholism

Anxiety

Attention Deficit/Hyperactivity Disorder Bipolar I Disorder Depression

Dysphoria

Insomnia

Irritability Libido Decreased Nervousness Premature Ejaculation Stress Dysuria

Nephrolithiasis

Cervical Dysplasia Dysmenorrhoea

Erectile Dysfunction

Cough

Dysphonia

Dyspnoea Epistaxis

Nasal Congestion

Rhinitis Allergic

Rhinorrhoea

Sinus Congestion Sleep Apnoea Syndrome

Throat Irritation

Acne

Actinic Keratosis

Dermal Cyst Dermatitis Contact

Ecchymosis

Eczema Intertrigo Night Sweats Onychomadesis Psoriasis Rash

Skin Lesion

Urticaria

Flushing

Hot Flush

Hypertension

Poisson Regression for

Exposure-adjusted Incidence

• Poisson models

– Adjust for different exposures in treatment and control

– Assume constant hazard over time

– Subject exposure: time to AE first onset or end of

treatment, whichever comes first

Ybj~Pois(tbj Tbj ); Xbj~Pois(cbj Cbj)

where tbj and cbj are event rates, and Tbj and Cbj for AEbj are

total exposure times in the treatment and control groups,

respectively

log(cbj )=λbj; log(tbj )=λbj + θbj ,

Note that θbj =log(RRbj)

Poisson Regression for

Exposure-adjusted Incidence

• Model 2a: Poisson regression with normal prior on

log-RR

– Same priors as used in Model 1a

• Model 2b: Poisson regression with mixture prior on

log-RR

– Poisson likelihood and log linear mean structure, same as

Model 2a;

– Mixture prior on log-RR: point mass at 0 + Normal

Exact CI using the frequentist

approach (no multiplicity adjustment)

Bayesian binomial hierarchical

model with normal prior on log-OR

(Model 1a)

Bayesian binomial hierarchical

model with mixture prior on log-

OR (Model 1b)

Bayesian non-hierarchical one-

stage binomial model with

mixture prior on log-OR

(Model 1c)

CIs by Model

Odds Ratios, 95% CI

0 5 10 15 20Fa

tigue

Her

pes Sim

plexS

inus

itis

Exc

oriatio

n

Ecc

hymos

is

Fatig

ue

Her

pes Sim

plexS

inus

itis

Exc

oriatio

n

Ecc

hymos

is

Fatig

ue

Her

pes Sim

plexS

inus

itis

Exc

oriatio

n

Ecc

hymos

is

Fatig

ue

Her

pes Sim

plexS

inus

itis

Exc

oriatio

n

Ecc

hymos

is

|

|

|

|

|

|

|

|

|

|

|

|

|

o observed+ post.mean

Simulation Study

• Questions to answer: – Is modeling the SOC/PT hierarchy helpful?

– What operating characteristics should be chosen to achieve acceptable false discovery rate (FDR) and power for meaningful risk sizes

• Simulation schema: – Null scenario:

• Randomly assign subjects to treatment arms

• Adverse events within subject remain unchanged to maintain the SOC/PT hierarchy

– Scenarios with elevated risks:

• Choose 2 SOCs with 3 and 21 PTs each;

– OR=2 and 5; Placebo risk = 1%, 5% and 10%;

– logOR ~ N(log2 or log5,0.25^2) within same SOC

• Other SOCs remain the same as in the null datasets.

Simulation Study (cont.)

NULL

Scenario

Scenario with

OR=2 and

Placebo risk=1%

Scenario with

OR=2 and

Placebo risk=5%

FDR/

FWER (%)

FDR

(%)

Power

(%)

FDR

(%)

Power

(%)

Non-adjusted

Fisher’s exact test

2-sided test,

p-value <=0.05

100 35 38 19 84

Non-hierarchical

Bayes Model *

(Model 1c)

c=1, p=0.6 100 50 23 25 68

c=1, p=0.7 96 38 19 15 64

c=1, p=0.8 68 24 15 7 60

Bayesian

Hierarchical

Model *

(Model 1b)

c=1, p=0.6 17 12 66 9 95

c=1, p=0.7 12 9 61 5 94

c=1, p=0.8 8 7 56 3 91

pDatacORbj )|Pr(*

FDR = False Discovery Rate = # of falsely flagged signals / # of flagged signals

Power = # of correctly flagged signals / # of signals that should be flagged

FWER = Family-wise Error Rate

Closing Remarks • Current traditional approach of flagging

routinely collected AEs based on

unadjusted p-values or CIs can result in

excessive false positive signals – As a result, it can cause undue concern for

approval/labeling/post marketing commitment

• Bayesian hierarchical mixture modeling

provides a useful tool to address

multiplicity – Allows for explicitly modeling AEs with the existing

MedDRA coding structure so that AEs within a SOC

or across SOCs can borrow strength from each other

Closing Remarks (Cont.)

(Cont.)

– Appealing in the rare event setting

• The model modulates the extremes

• Inferences based on the „exact‟ full posterior distributions,

relaxing the assumption of normality of the outcome

– Models the entire AE dataset, so the distinction

between Tier 2 and Tier 3 AEs may not be necessary

• Makes efficient use of all the data

– Straightforward and flexible to assess clinically

important difference with different scales (risk

difference, OR, or RR)

• Avoid detecting medically unimportant signals (an AE could

have high Pr(OR or RR > 1 or RD > 0 | Data), but medically

unimportant)

Closing Remarks (Cont.) • Simulation study can guide selection of a

signal detection threshold

• Graphics are effective in displaying

flagged signals when analyzing hundreds

or thousands types of AEs

• The field of clinical trial signal detection is

still in its infancy – More research and practice are needed

– Further developments await statisticians work closely

with clinicians/safety scientists to advance this field

Thank You!

References • Bate A, Lindquist M, Edwards, IR, Olsson S, Orre R, Lansner A, and De Freitas

RM (1998). A Bayesian neural network method for adverse drug reaction signal detection. Eur J Clin Pharmacol 54:315-321

• Berry S and Berry D (2004) Accounting for multiplicities in assessing drug safety: a three-level hierarchical mixture model. Biometrics, 60: 418-426

• Chi G, Hung HMJ, and O’Neill R (2002). Some comments on “Adaptive Trials and Bayesian Statistics in Drug Development” by Don Berry. In Pharmaceutical Report, Vol 9, 1-11

• Crowe B, Xia A, Berlin, J, Watson D, Shi H, Lin S, Kuebler J, et al. (2009). Recommendations for Safety Planning, Data Collection, Evaluation and Reporting During Drug, Biologic and Vaccine Development: A Report of the PhRMA Safety Planning, Evaluation and Reporting Team (SPERT). To appear in Clinical Trials

• DuMouchel W (1999). Bayesian data mining in large frequency tables, with an application to the FDA Spontaneous Reporting System (with discussion). The American Statistician 53:177-202

• Gould AL. Drug safety evaluation in and after clinical trials. Deming Conference, Atlantic City, 3 December 2002

• Mehrotra, DV and Heyse, JF (2004). Multiplicity considerations in clinical safety analysis. Statistical Methods in Medical Research 13, 227-238

• O'Connell, M. (2006). Statistical graphics for the design and analysis of clinical development studies. Insightful Webcast: http://www.insightful.com/news_events/webcasts/2006/07clinical/default.asp

• Spiegelhalter DJ, Best NG, Carlin BP and van der Linde A (2002) Bayesian measures of model complexity and fit (with discussion). J. Roy. Statist. Soc. B. 64, 583-640.