Bayesian Clinical Trials in Action
Bayesian Clinical Trials in Action
Author Manuscript
Stat Med. Author manuscript; available in PMC 2013 November 10.
Published in final edited form as:
Stat Med. 2012 November 10; 31(25): 2955–2972. doi:10.1002/sim.5404.
Abstract
Although the frequentist paradigm has been the predominant approach to clinical trial design since
the 1940s, it has several notable limitations. The alternative Bayesian paradigm has been greatly
enhanced by advancements in computational algorithms and computer hardware. Compared to its
frequentist counterpart, the Bayesian framework has several unique advantages, and its
incorporation into clinical trial design is occurring more frequently. Using an extensive literature
review to assess how Bayesian methods are used in clinical trials, we find them most commonly
used for dose finding, efficacy monitoring, toxicity monitoring, diagnosis/decision making, and
for studying pharmacokinetics/pharmacodynamics. The additional infrastructure required for
implementing Bayesian methods in clinical trials may include specialized software programs to
$watermark-text
run the study design, simulation, and analysis, and Web-based applications, which are particularly
useful for timely data entry and analysis. Trial success requires not only the development of
proper tools but also timely and accurate execution of data entry, quality control, adaptive
randomization, and Bayesian computation. The relative merit of the Bayesian and frequentist
approaches continues to be the subject of debate in statistics. However, more evidence can be
found showing the convergence of the two camps, at least at the practical level. Ultimately, better
clinical trial methods lead to more efficient designs, lower sample sizes, more accurate
conclusions, and better outcomes for patients enrolled in the trials. Bayesian methods offer
attractive alternatives for better trials. More such trials should be designed and conducted to refine
the approach and demonstrate its real benefit in action.
Keywords
$watermark-text
adaptive trial design; Bayesian paradigm; clinical trial conduct; frequentist paradigm; trial
efficiency; trial ethics
1. Introduction
A clinical trial is a prospective study that evaluates the effect of interventions in humans
under prespecified conditions. Clinical trials provide the most definitive mechanism for
assessing the outcome of interventions and form the foundation for evidence-based medicine
through reliable data. Clinical trials also represent key components in research, with the
potential to change the standard of care, improve quality of health, and control costs through
careful comparison of alternative treatments. The results of the first modern clinical trial,
which involved the use of streptomycin to treat pulmonary tuberculosis, were published in
1948 in the United Kingdom [1]. That trial involved randomizing patients into treatment and
control groups and assessing the outcome without knowledge of the treatment assignment.
Correspondence to: J. Jack Lee, Ph.D., Department of Biostatistics, The University of Texas MD, Anderson Cancer Center, 1400
Pressler Street, Unit 1411, Houston, Texas 77030, U.S.A. [email protected]; Tel: 1-713-794-4158; Fax: 1-713-563-4243.
Lee and Chu Page 2
Since then, clinical trials have been widely applied in medicine for the advancement of
science and the search for better treatments to improve health.
In the United States, the National Institutes of Health, particularly the National Heart, Lung,
and Blood Institute (NHLBI) and the National Cancer Institute (NCI), have led the effort to
develop and conduct clinical trials [2, 3]. Clinical Trials: Past, Present and Future, an
NHLBI-sponsored workshop held in 2010, explored the significance of clinical trials by
examining their historical development, surveying their present use and impact on medicine,
and discussing the future direction of clinical trials. This paper examines the emerging use
of Bayesian methods in clinical trials, focusing on their expanding implementation and
impact on medicine.
$watermark-text
From a statistical framework point of view, the frequentist paradigm has dominated the field
of clinical trials over the past 60 years. Considering the treatment effect, θ, which is the
unknown parameter of interest, the frequentist framework assumes that θ is fixed, yet
unknown. Through clinical trials, we can collect data to inform θ. Hence, the inference on
the treatment effect can be made by evaluating the probability: Prob(data|θ), where the data
are considered to be random and the parameter θ is fixed. Conversely, the Bayesian
framework assumes that the data are fixed and the unknown parameter θ is random.
Bayesian inference is made by computing Prob(θ|data). Thanks to the work of R.A. Fisher,
J. Neyman, and K. Pearson, among others, the frequentist theory was well developed in the
early 1900s [4]. Compared to the Bayesian methods, frequentist probability calculation is
simpler and less computationally intensive. As a result, the frequentist framework became
the mainstream of statistics and was quickly adopted into clinical trials as they evolved.
$watermark-text
Despite its usefulness and proven success in clinical trials, the frequentist framework suffers
from some major deficiencies. Most notably, frequentist inference on the parameter of
interest, θ, is made indirectly as it calculates Prob(data|θ) and not Prob(θ|data), as Bayesian
inference does. In-depth comparisons between the frequentist and Bayesian approaches can
be found in the literature [5, 6]. In this paper, our focus is on the use of Bayesian methods in
clinical trials; in particular, on their implementation and impact on medicine. The rest of the
paper is organized as follows. Section 2 describes the unique strength of the Bayesian
paradigm. Section 3 discusses the barriers for Bayesian clinical trials and efforts to
overcome them. Section 4 gives a brief overview of the various schools of Bayesian
methods. Section 5 shows the results of our literature review, through which we sought to
determine how Bayesian methods are used in clinical trials. Section 6 presents the MD
Anderson Cancer Center experience in the design and conduct of Bayesian clinical trials.
$watermark-text
Section 7 concludes with further discussion and a glimpse into the future.
pharmaceutical industry [13]. The Bayesian framework has several unique advantages over
its frequentist counterpart. We describe the key strengths of the Bayesian method in this
section.
inference depends on the unobserved data. In contrast, the Bayesian approach conditions on
the data and conforms to the likelihood principle. As a result, frequentist inference is valid
only when the prespecified clinical trial design is followed. When the study conduct deviates
from the original design, frequentist inference suffers and adjustments are difficult. On the
other hand, Bayesian inference is conditioned on the data and not on the design, so it can
still maintain validity as long as the prior distribution and the probability model are correctly
specified.
2.2. Bayesian methods model the unknown parameter with a distribution and properly
address various levels of uncertainty. The Bayesian approach is ideal for hierarchical
models
Under the Bayesian framework, all unknown parameters are random and follow certain
$watermark-text
probability distributions. The distribution parameters, themselves, are also unknown and can
be modeled with hyper priors. Thus, the Bayesian method is intrinsically hierarchical. Under
such a model, the uncertainty of the random variable can be captured by the variance of the
distribution. Hierarchical models are commonly used in clinical trials. For example, patients
undergo operations conducted by surgeons, and surgeons work within hospitals. Although
the frequentist method can apply to mixed models by modeling the treatment effect as fixed
and the surgeon and hospital effects as random, the construct is somewhat arbitrary in
designating a certain effect as fixed or random. The frequentist approach requires a stronger
assumption that observations are identical and independent. By assuming the parameter is
fixed, frequentist methods often underestimate the variability of the parameter of interest.
Conversely, the Bayesian method appropriates different levels of variability naturally under
the hierarchical model assumption. The de Finetti theorem states that subjects enrolled in the
$watermark-text
clinical trial are exchangeable if and only if the probability of the observed data can be
expressed as the data likelihood given the parameter that is integrated over the prior
distribution of the parameter. Exchangeability implies conditional independence of the data
given the parameter, which nicely fits the clinical trial setting [15].
2.3. Bayesian methods formally incorporate prior information gathered before, during and
outside of the trial
Typically, the concept for initiating a clinical trial does not arise from an information
vacuum, but is developed because of intriguing information found before the trial. To design
a trial, frequentists use the prior information in an ad hoc way to make assumptions on the
parameter of interest; whereas Bayesians elicit the prior distribution for θ and formally
incorporate it to make an inference. Although the prior distribution assumption may be
subject to criticism, it is spelled out explicitly and its impact can be evaluated by the
sensitivity analysis. In addition, the Bayesian framework allows for the incorporation of
information of two types: that which accumulated in the trial and that which was obtained
outside of the trial. Incorporating both types of information into the analysis strengthens the
evidence for making an inference. This provides an ideal tool for monitoring a clinical trial,
as well as for synthesizing information across multiple trials as a meta-analysis [16, 17].
2.4. Bayesian methods allow for more frequent monitoring and interim decision making
during the trial
By definition, Bayesian methods provide a platform for sequential learning. The prior
distribution is updated by the data to form the posterior distribution. The formed posterior
distribution then becomes the prior distribution for a future evaluation. Many clinical trials
are conducted over an extended period of time, and it is desirable to frequently monitor the
interim results of such trials so decisions can be made early when sufficient evidence has
accumulated. Although group sequential methods have been well developed under the
$watermark-text
frequentist paradigm [18]; frequentist properties are directly affected by the number and
timing of interim analyses. In contrast, Bayesian methods do not impose a penalty on
sequential learning. Another main difference between the two approaches is that the
frequentist approach makes interim decisions based on the conditional power, which is
calculated by fixing the parameter of interest at a certain value. The Bayesian approach
calculates the predictive probability by integrating the conditional power over the
distribution of θ because the parameter θ is random. The predictive probability factors in the
uncertainty of θ, whereas the conditional power does not [19].
2.5. Bayesian methods give direct answers to the questions that most people want to ask
By addressing the question directly, Bayesian methods calculate the probability of θ given
the data, and can answer a question such as, “For the new treatment, what is the probability
$watermark-text
that the success rate is more than 80%?” or “What is the probability that the true success rate
lies between the interval of (0.76, 0.92)?” Frequentists calculate the probability of the
observed data given a certain hypothesis, but they cannot answer the question, “What is the
probability that the null or alternative hypothesis is true?” Similarly, the frequentist
confidence interval is random because the data are random. The frequentist approach can be
used to calculate the probability that such an interval covers the true parameter if the process
is repeated many times, i.e., the long-range frequentist property; however, it cannot be used
to determine the coverage rate containing the true parameter for a given confidence interval.
Frequentists have to constantly explain to non-statisticians that the P value is not the
probability of the null hypothesis being true and that the 95% confidence interval does not
contain the true parameter 95% of the time. In contrast, Bayesian methods deal with the
problem head-on and give direct answers to the questions that most people want to ask.
$watermark-text
2.7. Bayesian methods can incorporate the utility function for informed decision making
In the Bayesian theoretic approach, clinical trial investigators can specify the “utility” or
“loss” of various events. For example, “what is the utility (or importance) of curing cancer
and what is the negative utility (loss) of developing a long-term toxicity due to the
treatment?” The optimal decision of the best treatment for a given patient can be made by
maximizing the utility function or minimizing the loss function. Bayesian methods allow
subjective opinions to be incorporated into the specification of the prior distribution and the
utility function. Different people can have different levels of prior belief and different
preferences as they rate the relative importance of events, such as being cured or suffering
treatment-related toxicity. Bayesian methods formulate these components explicitly and
quantitatively to aid investigators in making an informed decision.
2.8. Bayesian methods use a “learn as we go” approach. This real-time learning feature
forms the basis of adaptive clinical trial designs
As previously stated, the Bayesian method is a sequential learning method and takes a “learn
as we go” approach. It naturally adapts to the data and to all relevant information at hand.
$watermark-text
Traditional clinical trial designs and conduct are less adaptive, often lead to large trials over
an extended period of time, and are extremely expensive. Adaptive designs have been
proposed with the aim of creating more efficient, more flexible, and more ethical designs by
making design changes based on the interim data. The Bayesian framework naturally and
ideally fits into the development of adaptive designs [20]. Bayesian adaptive approaches are
especially useful in the following three areas: adaptive randomization, interim monitoring
for early stopping, and sample size adjustments. (1) Outcome adaptive randomization is
used to assign more patients into more effective treatment arms as data accumulate in the
trial. (2) Interim monitoring of the study endpoint uses predictive probability. If there is
convincing evidence that the new treatment is superior to the standard treatment, the trial
can be stopped early for efficacy. On the other hand, if the data strongly suggest that the new
treatment is worse or not better than the standard treatment, the trial can be stopped early for
$watermark-text
futility. (3) Adaptive sample size estimation can be achieved by calculating the probability
for a successful trial given the current result and the proposed sample size. If the current
result is promising but the sample size is not adequate, the study can be expanded by
increasing the sample size to have a high probability of reaching a definitive conclusion at
the end of study. In 2010, the U.S. FDA issued a guidance document for adaptive clinical
trials. [21] Although much of its content is in the frequentist framework, it also points out
the usefulness of the Bayesian approach in adaptive designs.
methods from becoming popular: the inherent computational demands and the use of
subjective information. We discuss these two barriers and the efforts to overcome them.
Another significant step forward in Bayesian computing was the development of the BUGS
(Bayesian inference using Gibbs sampling) software [24]. The BUGS project was started in
1989 by a group at the Medical Research Council (MRC) Biostatistics Unit and Imperial
College School of Medicine in the United Kingdom. BUGS was the first general purpose
software available for Bayesian computing. Users could specify the model and the
probability distributions of data from a rich set of commonly used distributions. By
supplying the prior distribution and the data, the posterior distribution could be computed.
Subsequently, an open source version called OpenBUGS was developed, which could run
on different operating systems. WinBUGS was then developed by adding useful GUIs
(graphics user interfaces) to facilitate its use in the Microsoft Windows environment. In
addition, R2WinBUGS and BRugs were developed for users to run WinBUGS within R
$watermark-text
such that WinBUGS code could be integrated within the R environment to ease the
generation, analysis, and reporting.
Another similar development was JAGS (just another Gibbs sampler). This program
analyzed Bayesian hierarchical models using MCMC simulation. The unique features of
JAGS include (1) a cross-platform engine for the BUGS language; (2) the ability for users to
write their own functions, distributions and samplers; and (3) a platform for experimenting
with Bayesian modeling.
To address the increasing use of Bayesian methods, SAS (SAS Institute, Cary, NC) added
the BAYES statement in the GENMOD, LIFEREG, and PHREG procedures. In addition,
starting from version 9.2, SAS introduced a new MCMC procedure. PROC MCMC is a
flexible simulation-based procedure suitable for fitting a wide range of Bayesian models.
$watermark-text
Upon specifying a likelihood function for the data and a prior distribution for the
parameters, PROC MCMC obtains samples from the corresponding posterior distributions.
It also produces summary and diagnostic statistics.
Although general computation tools such as BUGS or WinBUGS are available, specialized
computer programs are often needed to run a Bayesian study design, simulation, and
analysis. Web-based applications are particularly useful for timely data entry and analysis.
Web services are useful tools for exchanging information between the database module and
the computing module. The success of a Bayesian clinical trial requires not only the
development of proper tools, but also timely and accurate execution of data entry, quality
control, adaptive randomization, outcome assessment, and Bayesian computation. With
advancements in both computational algorithms and computer hardware, Bayesian
computation in the 21st century is no longer formidable.
$watermark-text
accordingly [25]. Others stress the importance of being objective and propose an objective
Bayesian approach by specifying objective priors [26]. Although the debate continues; the
goal is one shared by both communities — to efficiently and accurately infer conclusions
based on the data [27].
1. The empirical Bayes approach derives the prior distribution from the data; whereas
the standard Bayesian approach sets the prior before any data are observed. The
empirical approach can be viewed as a hierarchical Bayes model in which the
parameters at the highest level of the hierarchy are set to their most likely values,
instead of being integrated out.
2. The reference Bayes approach uses an “objective” or a “reference” prior such that
the inference is more objective. Some criticize this approach as “an attempt to
make the Bayesian omelets without breaking the Bayesian eggs.”
3. The proper Bayes approach uses informative prior distributions based on the
available evidence, but summarizes conclusions by posterior distributions without
explicit incorporation of the utility function. Some have called this a “stylist Bayes”
$watermark-text
approach.
4. The decision-theoretic or “full” Bayes approach uses explicit utility (or loss)
functions and makes decisions based on maximizing the expected utility (or
minimizing loss). One can argue that the decision-theoretic approach provides the
ultimate answer to the research question. For example, in drug development, not
only does the toxicity and efficacy of the drug need to be assessed, but the relative
risk and benefit of the drug also need to be specified explicitly in the utility
function. Furthermore, the cost of making a false positive decision (accepting a bad
drug) and the cost of making a false negative decision (rejecting a good drug) need
to be specified, as well. In complex settings with conflicting goals, the decision-
theoretic approach can provide the best (optimal) answer after considering all the
loss and gain of each decision. However, it is not easy to come up with a generally
$watermark-text
search to “clinical trial, all or clinical trial, phase I or clinical trial, phase II or clinical trial,
phase III or clinical trial, phase IV or clinical trial or controlled clinical trial or multicenter
study randomized controlled trial.” The search was further limited to “review articles and
meta analysis or review,” producing results that were subsequently removed from the
search. We also performed a similar search in Ovid Embase (Table I.B) using the terms
“Bayes or Bayesian.” We then searched “clinical trial*” under subject heading and
combined it with the previous “Bayes or Bayesian” search line. The search was limited to
“Cochrane library and meta analysis or systematic review” and the results were removed
from the final search. The publications obtained in the two literature searches were
combined (a total of 2012 articles) and the references were imported into Endnote X4.
$watermark-text
not been identified by the search algorithm [33–35]. The final number of articles reviewed
was 122.
Articles for inclusion were placed into three categories: The first category included studies
that used a Bayesian design and analysis (BDA; n = 21), where the clinical trial was
prospectively based upon a Bayesian design and the data were analyzed using Bayesian
methods. The second included prospective studies that used a frequentist design with a
Bayesian analysis (FDBA; n = 83). Bayesian reanalysis (BR) studies (n = 18) comprised the
final category, which involved the use of Bayesian methods to retrospectively analyze data
from a previously run clinical trial.
endpoint category, Bayesian method category, type of prior distribution, number of interim
analyses and whether the trial was terminated early. The results are summarized in Tables II
through IV.
As seen in Table II, publications prior to 1990 included only three clinical trials that used
Bayesian methods. That number quickly jumped to 20 in the 1990s, and to 99 in the period
since 2000. Most trials (67%) applied Bayesian methods for testing treatment efficacy; 8%
of the trials applied them for testing treatment safety; 16% of the trials applied them in the
areas of diagnostics and decision making. In terms of the medical fields, oncology led the
pack (25%), followed by cardiovascular research (16%), and CNS research (11%). These
122 papers were published dispersedly in 92 journals, with nine in the Journal of Clinical
$watermark-text
Oncology, four in PLoS One, and three each in The New England Journal of Medicine,
JAMA, Cancer, and Complementary Therapies in Medicine (data not shown).
From Table III we see that the vast majority of the trials are 2-arm (59%) or 1-arm (30%)
studies. About 50% of the trials had a control group. Almost 60% of the studies were
randomized trials, 48% of which applied equal randomization, and 6% of which applied
fixed, but unequal randomization. Only 5% of the trials applied adaptive randomization. In
terms of sample size, 23% were very small (n ≤ 30) and 26% had sample sizes between 31
and 99. Sample sizes were between 100 to 499 and 500 to 999, respectively, for 37% and
9% of the trials. Only 6% of the trials had sample sizes of 1,000 or more patients. About
51% of the trials enrolled patients during 2 or fewer years; whereas 35% of trials spent 2 to
5 years enrolling patients. The remaining 14% of the trials had an accrual period of 6 to 8
years.
$watermark-text
Table IV shows that continuous, binary, ordinal, and time-to-event variables, respectively,
were used as the primary endpoints in 43%, 33%, 12%, and 12% of the trials. A number of
Bayesian methods were applied in clinical trials, including for hypothesis testing (23%),
estimation (19%), model selection (12%), prediction (12%), regression (10%), hierarchical
modeling (10%), a decision-theoretic approach (5%), sensitivity analysis (5%), classification
(3%), and a Bayesian network (2%). Informative priors were used in 43% of the trials;
noninformative priors were used in 24% of the trials. The remaining 34% of the trials did
not provide sufficient information regarding the priors that were used. A vast majority of the
trials (87%) did not specify an interim analysis. Only 7%, 3%, and 4% of the trials had 1, 2,
or 3–7 interim analyses, respectively. Twelve percent of the trials were stopped early; 6 due
to futility, 4 due to efficacy, 1 for equivalence, and 1 for toxicity.
$watermark-text
5.4. Limitations
Although efforts were made to best identify the use of Bayesian methods in clinical trials,
our search has several limitations. First, we included the words “Bayes or Bayesian” and
“clinical trials” in our search criteria. Therefore, articles without these words were excluded.
The most prominent inadvertent exclusions were trials that used the continual reassessment
method (CRM) or the escalation with over-dose control (EWOC) method that did not
mention the words “Bayes or Bayesian.” We performed a separate literature search and
identified 81 CRM trials and 9 EWOC trials (see Appendix 1 for the search algorithm and
results). Only three of such trials were included in the 122 articles we reported. Most of the
studies were dose-finding cancer studies, with a goal of determining the maximum tolerated
dose. These were typically single-arm, open-label studies without a control group and with a
total sample size of less than 60. Most studies applied Bayesian design and analysis.
However, we decided not to include these studies not identified in the main search in the
tabulation because, similar to the Bayesian PK/PD studies, they form a distinct subgroup.
clinical trial were affiliated with MD Anderson. A review published in 2009 described 964
clinical protocols registered at MD Anderson between 2000 and early 2005 [37]. Bayesian
designs and/or analyses had been used in about 20% of the total protocols reviewed, and in
about 30% of the MD Anderson trials, but in only 7% of the multicenter protocols. Bayesian
methods had been applied in 34% of the phase I or phase II trials. The majority of the
Bayesian design and analysis features were found in non-mutually exclusive categories,
which included efficacy monitoring (62%), toxicity monitoring (27%), adaptive
randomization (10%), dose finding (9%), hierarchical modeling (7%), and determinations of
predictive probability (6%).
To facilitate the conduct of Bayesian clinical trials, a proper infrastructure must be set up for
registering patients, assigning patients to treatments, recording the outcomes, and providing
$watermark-text
interim and final analyses. At MD Anderson, we have developed a Clinical Trial Conduct
(CTC) platform. This secure, web-based application allows users to register a new trial and
select the type of design so that proper treatment assignment or randomization and
monitoring can be implemented. In this “role-based” system, each user has different
privileges, depending on his/her role in the trial. For example, the research nurse can verify
patients’ eligibility criteria, register patients, and enter patients’ toxicity and efficacy
outcomes. The statistician can read the treatment assignment and access the details of the
statistical computations, such as the randomization probability, but cannot alter the data. As
of August 2011, there were 133 trials and over 4,300 patients enrolled in the system. The
most commonly used designs were outcome adaptive randomization (n = 44), Pocock-
Simon baseline adaptive randomization (n = 42), the continual reassessment method (n =
29), and trials with time-to-event interim monitoring (n = 11).
$watermark-text
In addition to the CTC platform, we have also built custom-made applications for certain
specialized trials. For example, Figure 2 shows the schematic diagram of the web-based
application for running the BATTLE trial [34, 38]. The top panel shows the study flow
chart. Eligible patients are registered to the trial and have a biopsy taken for molecular
marker analysis. The research molecular pathology laboratory analyzes the sample for
mutations and uses the results of fluorescence in situ hybridization (FISH) and
immunohistochemistry (IHC) expression analyses to determine the biomarker group for the
patient. This process, from registration to reporting the biomarker results, is completed
within two weeks. Based on the patient’s biomarker group and the cumulative outcome
results, the patient is then adaptively randomized into one of the four treatment groups.
Additional clinical visits are scheduled for the patient and the disease control status (primary
endpoint of the study) is evaluated eight weeks after randomization. Patients are continually
followed for secondary endpoints, such as progression-free survival and overall survival,
until they are off the study. The middle panel of Figure 2 shows different modules in the
application. Through a web-interface, data are entered into different modules. For example,
research nurses enter the medical history, physical examination, adverse events, efficacy
assessment, etc. The laboratory technician enters the results of the marker analysis. To
perform adaptive randomization, the patient’s marker information, eligibility status, and up-
to-date outcome information are passed to an R code through web services. The R code
performs Bayesian computation to determine the randomization probability and randomize
patients to eligible treatments accordingly. All the data are stored in the institutional
database CORe and/or the study-specific SQL Server 2005 database. The application also
has a report generation module that can automatically generate several reports for
monitoring and quality assurance purposes. For example, an accrual report is generated to
check the accrual rate. An outcome timeliness report can check whether the 8-week disease
control status has been timely entered. If the assessment of the primary endpoint of a patient
remains past due for two weeks, an automatically generated email will be sent to the study
coordinator/research nurse. The toxicity and drug compliance reports can also be generated
$watermark-text
Bayesian methods are increasingly being used in clinical trials. This trend will likely
continue [36, 40]. We have also begun to see the impact of Bayesian applications among
policymakers in regulatory agencies regarding the approval of new medical devices or drugs
evaluated in studies that use Bayesian designs or methods. The Center for Devices and
Radiological Health (CDRH) at the FDA issued a “Guidance for the Use of Bayesian
Statistics in Medical Device Clinical Trials” in 2010 [41]. The CDRH has approved more
than 20 original Pre-Market Approvals (PMAs) and PMA supplements with a Bayesian
analysis as the primary method. Many Investigational Device Exemptions (IDEs) and
applications for “substantial equivalence” (510(k)s) that used Bayesian methods have also
been approved [42]. On the drug side, the Center for Drugs and Experimental Research of
the FDA approved Pravigard Pac (Bristol-Myers Squibb) based on Bayesian analyses of
efficacy in 2003 [17]. As many clinical trials using Bayesian methods are underway, it is
$watermark-text
expected that more drugs and devices will be approved by the FDA based on Bayesian
methods.
The development of newer and better clinical trial designs under the Bayesian paradigm
continues to be an active area of statistical methodology research. The availability of
accompanying software for the implementation of Bayesian methods is crucial for the use of
these methods in clinical trials. Altman indicated that there is a delay of 4 to 6 years between
the date when a statistical method is published and when that publication is cited 25 times in
medical journals [43]. The time gap between the publication of a new trial design and its
adoption still exists, but is closing rapidly. This is evident in the Bayesian dose-finding
studies and adaptive designs [36, 40]. (We list some useful information/tools for learning
Bayesian clinical trial methods and designs in Appendix 2.)
Bayesian methods hold great promise for improving the efficiency and flexibility of
conducting clinical trials and are ideal for learning and adaptation. Bayesian methods
provide excellent tools when searching for effective treatments and predictive markers in the
quest for biomarker-based personalized medicine — with a goal of treating more patients
with more effective therapies. Good examples for such trials include the BATTLE trial [34],
the currently ongoing BATTLE-2 trial, and the I-SPY 2 trial [44]. Successful
implementations of Bayesian methods have been demonstrated in a wide range of clinical
trial applications.
Bradley Efron, in his 2004 address as the president of the American Statistical Association,
stated that the field of statistics was dominated by the Bayesian view in the 19th century and
by the frequentist view in the 20th century. He suggested that statistics in the 21st century,
challenged by greater magnitudes of data and complexity, will require a combination of both
Bayesian and frequentist methods [45]. The following year, Roderick J. Little, in his
presidential address, proposed the “calibrated Bayes” approach [46]. The calibrated Bayes
$watermark-text
approach uses frequentist methods for model development and assessment, and Bayesian
methods for inference under a model. This capitalizes on the strengths of both paradigms
and provides a useful roadmap for many problems of statistical modeling and inference.
The relative merit of the Bayesian and frequentist approaches continues to be the subject of
debate in statistics and other scientific fields. Regarding the two paradigms, the past was
combative, the present is competitive, and the future will be cooperative. After all, Bayesian
and frequentist approaches offer complementary views and can learn from each other.
Recently, more evidence can be found showing the convergence of the two camps, at least
on a practical level [47]. Ultimately, better clinical trial methods lead to more efficient
designs, lower sample sizes, more accurate conclusions, and better outcomes for patients
enrolled in the trials. Bayesian methods offer an attractive alternative for better trials. More
such trials should be conducted to refine the approach and demonstrate the real benefit of the
$watermark-text
Acknowledgments
The authors thank Donald A. Berry for helpful discussions, Richard Herrick for providing information on the MD
Anderson Clinical Trial Conduct web-based software application, and Ms. LeeAnn Chastain for editorial assistance.
References
1. Streptomycin-in-Tuberculosis-Trials-Committee. Streptomycin treatment of pulmonary
tuberculosis: a Medical Research Council investigation. British Medical Journal. 1948; 2:769–
782.10.1136/bmj.2.4582.769 [PubMed: 18890300]
2. Halperin M, DeMets DL, Ware JH. Early methodological developments for clinical trials at the
$watermark-text
National Heart, Lung and Blood Institute. Statistics in Medicine. 1990; 9(8):881–892.10.1002/sim.
4780090804 [PubMed: 2218191]
3. Gehan EA, Schneiderman MA. Historical and methodological developments in clinical trials at the
National Cancer Institute. Statistics in Medicine. 1990; 9:871–880.10.1002/sim.4780090803
[PubMed: 2218190]
4. Stigler SM. The history of statistics in 1933. Statistical Science. 1996; 11:244–252.10.1214/ss/
1032280216
5. Gehan EA. Methodological issues in cancer clinical trials: the comparison of therapies. Biomedicine
and Pharmacotherapy. 1988; 42 (3):161–165.
6. Bayarri MJ, Berger JO. The interplay of Bayesian and frequentist analysis. Statistical Science. 2004;
19:58–80.10.1214/088342304000000116
7. Bayes T. An essay towards solving a problem in the doctrine of chances. 1763. M D Computing:
Computers in Medical Practice. 1991; 8:157–171. [PubMed: 1857193]
8. Stigler SM. Thomas Bayes’ Bayesian inference. Journal of the Royal Statistical Society, Series A.
1982; 145:250–258.10.2307/2981538
9. Bernardo, JM.; Smith, AFM. Bayesian Theory. Wiley; West Sussex: 1994.
10. Meier P. Jerome Cornfield and the methodology of clinical trials. Controlled Clinical Trials. 1981;
1(4):339–345. [PubMed: 7021066]
11. Ederer F. Jerome Cornfield’s contributions to the conduct of clinical trials. Biometrics. 1982; 38
(Suppl):25–32. [PubMed: 7046822]
12. Ashby D. Bayesian statistics in medicine: a 25 year review. Statistics in Medicine. 2006; 25(21):
3589–3631.10.1002/sim.2672 [PubMed: 16947924]
13. Grieve AP. 25 years of Bayesian methods in the pharmaceutical industry: a personal, statistical
bummel. Pharmaceutical Statistics. 2007; 6:261–281.10.1002/pst.315 [PubMed: 17955514]
14. Berger, JO.; Wolpert, RL. The Likelihood Principle. Gupta, SS., editor. Institute of Mathematical
Statistics; Hayward, CA: 1984. http://projecteuclid.org/euclid.lnms/1215466210
15. Bernardo JM. The concept of exchangeability and its applications. Far East Journal of
$watermark-text
21. Center for Drug Evaluation and Research, Food and Drug Administration. Guidance for industry:
adaptive design clinical trials for drugs and biologics. U.S. Department of Health and Human
Services; Rockville, MD: 2010. available at http://www.fda.gov/downloads/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances/UCM201790.pdf
22. Geman S, Geman D. Stochastic relaxation, Gibbs distributions, and the Bayesian restoration of
images. IEEE Transactions on Pattern Analysis and Machine Intelligence. 1984; PAMI-6:721–
741.10.1109/TPAMI.1984.4767596 [PubMed: 22499653]
23. Gelfand AE, Smith AFM. Sampling-based approaches to calculating marginal densities. Journal of
the American Statistical Association. 1990; 85:398–409.10.2307/2289776
24. Lunn D, Spiegelhalter D, Thomas A, Best N. The BUGS project: evolution, critique and future
directions. Statistics in Medicine. 2009; 28:3049–3067.10.1002/sim.3680 [PubMed: 19630097]
25. Goldstein M. Subjective Bayesian analysis: principles and practice. Bayesian Analysis. 2006;
1:403–420.10.1214/06-BA116
$watermark-text
26. Berger J. The case for objective Bayesian analysis. Bayesian Analysis. 2006; 1:385–
402.10.1214/06-BA115
27. Fienberg SE. Does it make sense to be an “objective Bayesian”? (Comment on articles by Berger
and by Goldstein). Bayesian Analysis. 2006; 1:429–432.10.1214/06-BA116C
28. Spiegelhalter, DJ.; Abrams, KR.; Myles, JP. Bayesian Approaches to Clinical Trials and Health-
Care Evaluation. Wiley; West Sussex: 2004.
29. Sheiner LB, Beal SL. Evaluation of methods for estimating population pharmacokinetic
parameters. I. Michaelis-Menten model: routine clinical pharmacokinetic data. Journal of
Pharmacokinetics and Biopharmaceutics. 1980; 8:553–571.10.1007/BF01060053 [PubMed:
7229908]
30. Sheiner LB, Ludden TM. Population pharmacokinetics/dynamics. Annual Review of
Pharmacology and Toxicology. 1992; 32:185–209.10.1146/annurev.pa.32.040192.001153
31. Whiting B, Kelman AW, Grevel J. Population pharmacokinetics. Theory and clinical application.
Clinical Pharmacokinetics. 1986; 11:387–401.10.2165/00003088-198611050-00004 [PubMed:
3536257]
32. Bauer RJ, Guzy S, Ng C. A survey of population analysis methods and software for complex
pharmacokinetic and pharmacodynamic models with examples. The AAPS Journal. 2007;
910.1208/aapsj0901007
33. Buzdar AU, Ibrahim NK, Francis D, Booser DJ, Thomas ES, Theriault RL, Pusztai L, Green MC,
Arun BK, Giordano SH, Cristofanilli M, Frye DK, Smith TL, Hunt KK, Singletary SE, Sahin AA,
Ewer MS, Buchholz TA, Berry D, Hortobagyi GN. Significantly higher pathologic complete
remission rate after neoadjuvant therapy with trastuzumab, paclitaxel, and epirubicin
chemotherapy: results of a randomized trial in human epidermal growth factor receptor 2-positive
operable breast cancer. Journal of Clinical Oncology. 2005; 23:3676–3685.10.1200/JCO.
2005.07.032 [PubMed: 15738535]
34. Kim ES, Herbst1 RS, Wistuba II, Lee JJ Jr, GRB, Tsao A, Stewart DJ, Hicks ME Jr, JE, Gupta S,
$watermark-text
Alden CM, Liu S, Tang X, Khuri FR, Tran HT, Johnson BE, Heymach JV, Mao L, Fossella F,
Kies MS, Papadimitrakopoulou V, Davis SE, Lippman SM, Hong WK. The BATTLE trial:
personalizing therapy for lung cancer. Cancer Discovery. 2011; 1:44–
53.10.1158/2159-8274.CD-10-0010 [PubMed: 22586319]
35. Julian TB, Blumencranz P, Deck K, Whitworth P, Berry DA, Berry SM, Rosenberg A, Chagpar
AB, Reintgen D, Beitsch P, Simmons R, Saha S, Mamounas EP, Giuliano A. Novel intraoperative
molecular test for sentinel lymph node metastases in patients with early-stage breast cancer.
Journal of Clinical Oncology. 2008; 26:3338–3345.10.1200/JCO.2007.14.0665 [PubMed:
18612150]
36. Chevret S. Bayesian adaptive clinical trials: a dream for statisticians only? Statistics in Medicine.
2011 Sep 9. Epub ahead of print. 10.1002/sim.4363
37. Biswas S, Liu DD, Lee JJ, Berry DA. Bayesian clinical trials at the University of Texas M. D.
Anderson Cancer Center. Clinical Trials. 2009; 6:205–216.10.1177/1740774509104992 [PubMed:
19528130]
$watermark-text
38. Zhou X, Liu S, Kim ES, Herbst RS, Lee JJ. Bayesian adaptive design for targeted therapy
development in lung cancer — a step toward personalized medicine. Clinical Trials. 2008; 5:181–
193.10.1177/1740774508091815 [PubMed: 18559407]
39. Tighiouart M, Rogatko A. Dose finding with escalation with overdose control (EWOC) in cancer
clinical trials. Statistical Science. 2010; 25:217–226.10.1214/10-STS333
40. Rogatko A, Schoeneck D, Jonas W, Tighiouart M, Khuri FR, Porter A. Translation of innovative
designs into phase I trials. Journal of Clinical Oncology. 2007; 25:4982–4986.10.1200/JCO.
2007.12.1012 [PubMed: 17971597]
41. Center for Devices and Radiological Health, Food and Drug Administration. Guidance for the use
of Bayesian statistics in medical device clinical trials. U.S. Department of Health and Human
Services; Rockville, MD: 2010. available at http://www.fda.gov/downloads/MedicalDevices/
DeviceRegulationandGuidance/GuidanceDocuments/ucm071121.pdf
$watermark-text
42. Campbell, G. Bayesian statistics at the FDA: the trailblazing experience with medical devices.
Presentation at Rutgers, the State University of New Jersey; April 3, 2009; available at http://
www.stat.rutgers.edu/iob/bioconf09/slides/Campbell.pdf
43. Altman DG, Goodman SN. Transfer of technology from statistical journals to the biomedical
literature—past trends and future predictions. JAMA. 1994; 272:129–132.10.1001/jama.272.2.129
[PubMed: 8015123]
44. Barker AD, Sigman CC, Kelloff GJ, Hylton NM, Berry DA, Esserman LJ. I-SPY 2: An adaptive
breast cancer trial design in the setting of neoadjuvant chemotherapy. Clinical Pharmacology and
Therapeutics. 2009; 86:97–100.10.1038/clpt.2009.68 [PubMed: 19440188]
45. Efron B. Bayesians, frequentists, and scientists. Journal of the American Statistical Association.
2005; 100:1–5.10.1198/016214505000000033
46. Little RJ. Calibrated Bayes: A Bayes/frequentist roadmap. American Statistician. 2006; 60:213–
223.10.1198/000313006X117837
47. Arjas E. On future directions in statistical methodologies — some speculations. Scandinavian
Journal of Statistics. 2011; 38:185–194.10.1111/j.1467-9469.2011.00737.x
48. O’Quigley J, Pepe M, Fisher L. Continual reassessment method: a practical design for phase 1
clinical trials in cancer. Biometrics. 1990; 46:33–48.10.2307/2531628 [PubMed: 2350571]
49. Babb J, Rogatko A, Zacks S. Cancer phase I clinical trials: efficient dose escalation with overdose
control. Statistics in Medicine. 1998; 17:1103–1120.10.1002/
(SICI)1097-0258(19980530)17:10<1103::AID-SIM793>3.0.CO;2-9 [PubMed: 9618772]
Appendix 1. Search algorithm and results for identifying clinical trials using
continual reassessment method (CRM) and escalation with overdose
control (EWOC)
Appendix Table I
Schema for literature search for continual reassessment method
$watermark-text
A. Ovid Medline ® In-process and other non-indexed citations and Ovid MEDLINE ® 1948 to Search result
present
continual reassessment method.mp. [mp=protocol supplementary concept, rare disease supplementary 148
concept, title, original title, abstract, name of substance word, subject heading word, unique identifier]
limit 1 to (clinical trial, all or clinical trial, phase i or clinical trial, phase ii or clinical trial, phase iii or 46
clinical trial, phase iv or clinical trial or controlled clinical trial or multicenter study randomized
controlled trial)
Continual reassessment method.mp. [mp=title, abstract, subject headings, heading word, drug trade 169
name, original title, device manufacturer, drug manufacturer, device trade name, keyword
Appendix Table II
Schema for literature search for escalation with overdose control method
A. Ovid Medline ® In-process & Other Non-Indexed Citations and Ovid MEDLINE ® 1948 to Search result
present
escalation with overdose control.mp. [mp=protocol supplementary concept, rare disease supplementary 9
concept, title, original title, abstract, name of substance word, subject heading word, unique identifier]
limit 1 to (clinical trial, all or clinical trial, phase i or clinical trial, phase ii or clinical trial, phase iii or 2
clinical trial, phase iv or clinical trial or controlled clinical trial or multicenter study randomized
controlled trial)
escalation with overdose control.mp. [mp=title, abstract, subject headings, heading word, drug trade 16
name, original title, device manufacturer, drug manufacturer, device trade name, keyword
In addition, we performed reverse citation searches on two original articles for the CRM and
EWOC methods, respectively [48, 49]. The results from all our searches were placed into
EndNote X4 for duplicate removal and categorization.
Our reverse citation search netted 356 articles from J. O’Quigley and 109 from J. Babb.
There were 95 duplicates, 119 statistical journals, 4 computer/engineering journals, 75
review/opinion papers, 100 method papers, 3 meta-analysis papers, 3 observational/database
papers, and 5 non-CRM papers. From the resulting journals, we found 59 CRM and 9
EWOC papers.
The Ovid Medline/Embase search was combined with the reverse citation search and 21
duplicates were removed. Our final cohort consisted of 81 CRM and 9 EWOC papers.
A. Articles
1. Berry DA. Bayesian clinical trials. Nature Reviews Drug Discovery 2006; 5(1):27–
36.
2. Goodman SN. Introduction to Bayesian methods I: measuring the strength of
evidence. Clinical Trials 2005; 2:282–290.
3. Louis TA. Introduction to Bayesian methods II: fundamental concepts. Clinical
Trials 2005; 2:291–294.
4. Berry DA. Introduction to Bayesian methods III: use and interpretation of Bayesian
tools in design and analysis. Clinical Trials 2005; 2:295–300.
5. Berry DA. Bayesian statistics and the efficiency and ethics of clinical trials.
$watermark-text
B. Books
1. Berry DA, Stangl D. Bayesian Biostatistics. CRC Press: Boca Raton, FL, 1996.
2. Spiegelhalter DJ, Abrams KR, Myles JP. Bayesian Approaches to Clinical Trials
and Health-Care Evaluation. Wiley: West Sussex, 2004.
$watermark-text
3. Carlin BP, Louis TA. Bayesian Methods for Data Analysis. 3rd edition. Chapman
& Hall/CRC: Boca Raton, FL, 2008.
4. Hoff PD. A First Course in Bayesian Statistical Methods. Springer: New York,
2009.
5. Albert J. Bayesian Computation with R (Use R). 2nd edition. Springer: New York,
2009.
6. Gelman A, Carlin JB, Stern HS, Rubin DB. Bayesian Data Analysis. 2nd edition.
Chapman & Hall/CRC: Boca Raton, FL, 2009.
7. Berry SM, Carlin BP, Lee JJ, Mueller P. Bayesian Adaptive Methods for Clinical
Trials. Chapman & Hall/CRC: Boca Raton, FL, 2010.
C. Video tutorials
1. FDA and the Johns Hopkins University Workshop: Can Bayesian approaches to
studying new treatments improve regulatory decision-making? http://
webcasts.prous.com/bayesian2004/
D. Computer programs
1. General Bayesian computation tools
a. BUGS, OpenBUGS, and WinBUGS: http://www.mrc-bsu.cam.ac.uk/bugs/
$watermark-text
b. JAGS: http://mcmc-jags.sourceforge.net/
2. Running WinBUGS from R
a. BRugs: http://www.biostat.umn.edu/~brad/software/BRugs/
b. R2WinBUGS: http://cran.r-project.org/web/packages/R2WinBUGS/
index.html
3. Running WinBUGS from Stata - The winbugsfromstata package: http://
www2.le.ac.uk/departments/health-sciences/research/ships/gen-epi/Progs/winbugs-
from-stata
4. A collections of useful tools for Bayesian clinical trials, including CRM, BMA-
CRM, EFF-TOX, Multc99, adaptive randomization, predictive probability, etc., can
$watermark-text
FACTS_introduction.pdf
8. Cytel’s Compass: software for adaptive dose-finding trials http://www.cytel.com/
Software/Compass.aspx
Figure 1.
Study selection algorithm
§ comp = computers
* also includes Cochrane journals and systematic reviews
** also includes commentaries, letters, replies, surveys, notes, guidelines and short articles
*** obs = observational; also includes registries and epidemiologic studies
**** methodology focused includes statistical strategies, algorithms, trial designs, method
comparisons/demonstrations, tutorials, model development/validation, and simulations.
† pharmacokinetics/pharmacodynamics
$watermark-text
Figure 2.
Schematic diagram of the web-based database application for the conduct of the BATTLE
trial
$watermark-text
$watermark-text
Table I
Schema for literature search
$watermark-text
A. Ovid Medline ® In-process and other non-indexed citations and Ovid MEDLINE ® 1948 to September 2011 Search result
1 (bayes or bayesian).mp. [mp=protocol supplementary concept, rare disease supplementary concept, title, original title, 21,310
abstract, name of substance word, subject heading word, unique identifier]
2 limit 1 to (clinical trial, all or clinical trial, phase i or clinical trial, phase ii or clinical trial, phase iii or clinical trial, phase 682
iv or clinical trial or controlled clinical trial or multicenter study randomized controlled trial)
1 (bayes or bayesian).mp. [mp=title, abstract, subject headings, heading word, drug trade name, original title, device 21,997
manufacturer, drug manufacturer, device trade name, keyword
2 clinical trial*.sh. 829,875
$watermark-text
3 1 and 2 1346
5 3 not 4 1333
$watermark-text
Table II
Characteristics of publications reviewed (n = 122): year of publication, type of clinical trial and medical area
$watermark-text
of study.
Years
1975–1989 3 2.5
1990–1994 6 4.9
1995–1999 14 11.5
2000–2004 27 22.1
2005–2011 72 59.0
Type of clinical trial
Efficacy 82 67.2
$watermark-text
Genetics 2 1.6
Genitourinary system 2 1.6
Geriatrics 2 1.6
Hematology 2 1.6
Infectious disease 10 8.2
Metabolic disorder 3 2.5
Obstetrics and gynecology 9 7.4
Oncology 30 24.6
Ophthalmology 1 0.8
Pain 2 1.6
Pediatrics 1 0.8
Pulmonary system 8 6.6
Radiology 2 1.6
Renal system 4 3.3
Transplant 4 3.3
Table III
Trial characteristics in publications reviewed (n = 122).
$watermark-text
Number of arms*
1 35 30.4
2 68 59.1
3 7 6.1
≥4 5 4.3
Yes** 57 49.6
None 58 50.4
Method of Randomization*
$watermark-text
None 47 40.9
Actual Sample Size
≤ 30 28 23.0
31–59 18 14.8
60–99 13 10.7
100–199 16 13.1
200–499 29 23.8
$watermark-text
500–999 11 9.0
1000–9999 7 5.7
*
Does not include seven papers with multiple studies
**
23 active; 19 placebo; 1 no treatment; 14 unspecified
***
includes: forty-seven 2-arm trials; five 3-arm trials; two 4-arm trials; one 5-arm trial
****
all 2-arm trials (one 1:1.5, one 2.8:1, four 2:1, one 3:1)
*****
79 studies did not specify
Table IV
Statistical attributes in publications reviewed (n = 122).
$watermark-text