scispace - formally typeset
Search or ask a question
Journal ArticleDOI

A Bayesian dose-finding design for drug combination clinical trials based on the logistic model.

TL;DR: Under the proposed design, the posterior estimates of the model parameters continuously update to make the decisions of dose assignment and early stopping, and the design is competitive and outperforms some existing designs.
Abstract: In early phase dose-finding cancer studies, the objective is to determine the maximum tolerated dose, defined as the highest dose with an acceptable dose-limiting toxicity rate. Finding this dose for drug-combination trials is complicated because of drug–drug interactions, and many trial designs have been proposed to address this issue. These designs rely on complicated statistical models that typically are not familiar to clinicians, and are rarely used in practice. The aim of this paper is to propose a Bayesian dose-finding design for drug combination trials based on standard logistic regression. Under the proposed design, we continuously update the posterior estimates of the model parameters to make the decisions of dose assignment and early stopping. Simulation studies show that the proposed design is competitive and outperforms some existing designs. We also extend our design to handle delayed toxicities. Copyright © 2014 John Wiley & Sons, Ltd.

Summary (3 min read)

1 Introduction

  • For oncologists, the objective of phase I dose-finding studies is to determine the maximum tolerated dose (MTD), defined as the highest dose with a relatively acceptable dose-limiting toxicity (DLT) [1, 2].
  • Mandrekar et al. proposed an approach incorporating the toxicity and efficacy of each agent into the identification of an optimal dosing region for the combination by using a continuation ratio model to separate each agent’s toxicity and efficacy curves [8, 9].
  • Most of these existing designs rely on complicated statistical models that typically are not familiar to clinicians, which hinder their acceptance and application in practice.
  • In addition, the performance of these designs seems comparable and there is no consensus which design should be used [13].

Dose-combination model

  • Let there be a two-drug combination used in a phase I dose-finding clinical trial for which the dose-toxicity relationship is monotonic and increases with the dose levels.
  • Before two agents can be combined, each of them typically have been thoroughly investigated individually.
  • Therefore, there is often rich prior information on pj’s and qk’s and their values can be readily elicited from physicians.
  • Using the prior information to define standardized dose has been widely used in dose-finding trial designs, and the most well known example perhaps is the skeleton of the continuous reassessment method (CRM) [21] with a logistic model.
  • Research has shown that this approach improves the estimation stability and trial performance [2, 22].

Likelihood and posterior inference

  • Under the proposed model, the likelihood is simply a product of the Bernoulli density, given by L(β0, β1, β2, β3|data) ∝ (2) We sample this posterior distribution using Gibbs sampler, which sequentially draws each of the parameters from their full conditional distributions .the authors.the authors.
  • Dose finding algorithm and determination of the MTD During the trial conduct, the authors use the dose-finding algorithm proposed by Yin and Yuan [11, 10] to determine dose escalation and deescaltion, and propose a different criterion for MTD selection at the end of the trial.
  • Shown in dark gray is the AUC for a toxicity probability greater than 0.4, which is equal to the probability of overdosing.

BCOPULA and BGUMBEL methods

  • Yin and Yuan proposed two Bayesian methods that use copula regression for combinations.
  • The parameter γ characterizes the drug interactive effect, and α and β the uncertainty of the initial guesses.
  • The combination allocation algorithm is the same as that presented in Section 2.1.
  • The final MTD is the combination with a toxicity probability closest to the target among the combinations already administered in the trial.

LOGODDS

  • By backsolving this equation, an explicit expression for the probability of toxicity is obtained.
  • A normal prior centered in 0 and with variance of 100 was chosen for the interaction parameter.
  • The rest of the dose allocation process, estimation and MTD determination was the same as their proposed design in order to compare their method involving a simple interaction logistic model with other logistic models.

One-dimensional CRM

  • In practice, one-dimensional CRM sometimes is used to conduct dose-combination trials [13].
  • Under this method, the authors first preselect a subset of combinations, for which the toxicity probability order is known, and then apply the standard CRM to find the MTD.
  • The drawback of such an approach is that the authors only investigate a subset of the whole two-dimensional dose space and may miss the true MTD.

3 Simulations

  • The authors simulated 2000 independent replications of phase I trials that evaluate two agents in drug combinations, with five dose levels for Agent 1 and three for Agent 2, giving 15 possible combinations.
  • The authors fixed the toxicity target at 0.3 and used an overall sample size of 60.
  • The design parameters of all the designs (e.g., working model) have been calibrated via simulation before used for the comparison.
  • The authors selected these features in order to employ typical trial set-ups in their simulation study.
  • The authors set the length around the targeted interval, δ, at 0.1.

4 Results

  • For each scenario, the authors present the correct MTD selection rate, or percentages of correct selection (PCS), in Table 2.
  • For these scenarios, all model-based designs gave high PCS, whereas LOGISTIC seemed to perform better than the other methods.
  • The addition of the stopping rule for unacceptable toxicity resulted in PCS that were similar to those presented above (Table 5), except in scenario 4 in which the first dose combination, (1, 1), was the MTD.

4.1 Sensitivity analysis

  • The authors conducted a sensitivity analysis in order to study the performance of their design using different prior distributions and parameters values.
  • According to Table 5, the authors can see that the PCS for all scenarios were very similar under these different prior distributions.

4.2 Time-to-event outcome

  • In practice, a longer follow-up time may be required to assess the toxicity outcome.
  • Before combination assignment, the likelihood is defined as L(β0, β1, β2, β3|data) =.
  • The authors simulated the time-to-toxicity outcomes using an exponential distribution such that the toxicity probabilities at the end of follow-up matched those given in Table 1.
  • Table 6 shows the results of the extended LOGISTIC for all 15 scenarios.
  • The authors observe that the performance of the design decreases only slightly by 2%, and the PCS for all scenarios are still very high.

5 Discussion

  • The authors have proposed a statistical method for clinical trial designs that evaluate various dose combinations for two agents.
  • One benefit of their method compared with the other proposed designs is that it is also efficient when the MTDs are not necessarily located on the same diagonal.
  • When combining several agents, designs developed for single-agent dose-finding trials cannot be applied to combination studies.
  • This approach performs well if the target dose combinations happen to be included in the subset.
  • These files are freely available upon request.

Did you find this useful? Give us your feedback

Content maybe subject to copyright    Report

HAL Id: hal-01298657
https://hal.sorbonne-universite.fr/hal-01298657
Submitted on 6 Apr 2016
HAL is a multi-disciplinary open access
archive for the deposit and dissemination of sci-
entic research documents, whether they are pub-
lished or not. The documents may come from
teaching and research institutions in France or
abroad, or from public or private research centers.
L’archive ouverte pluridisciplinaire HAL, est
destinée au dépôt et à la diusion de documents
scientiques de niveau recherche, publiés ou non,
émanant des établissements d’enseignement et de
recherche français ou étrangers, des laboratoires
publics ou privés.
A Bayesian dose-nding design for drug combination
clinical trials based on the logistic model
Marie-Karelle Riviere, Ying Yuan, Frédéric Dubois, Sarah Zohar
To cite this version:
Marie-Karelle Riviere, Ying Yuan, Frédéric Dubois, Sarah Zohar. A Bayesian dose-nding design for
drug combination clinical trials based on the logistic model. Pharmaceutical Statistics, Wiley, 2014,
13 (4), pp.247-257. �10.1002/pst.1621�. �hal-01298657�

A Bayesian dose-finding design for drug combination
clinical trials based on the logistic model
Marie-Karelle Riviere
(a,b)
, Ying Yuan
(c)
, Fr
´
ed
´
eric Dubois
(b)
and Sarah Zohar
(a)
(a) INSERM, UMRS 1138, Equipe 22, Centre de Recherche des Cordeliers, Universit
´
e Paris 5, Universit
´
e Paris 6, Paris, France
(b) IRIS (Institut de Recherches Internationales Servier), Suresnes, France
(c) Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, U.S.A.
Abstract
In early phase dose-finding cancer studies, the objective is to determine the maximum tolerated
dose, defined as the highest dose with an acceptable dose-limiting toxicity rate. Finding this dose
for drug-combination trials is complicated due to drug-drug interactions, and many trial designs
have been proposed to address this issue. These designs rely on complicated statistical models that
typically are not familiar to clinicians, and are rarely used in practice. The aim of this paper is
to propose a Bayesian dose-finding design for drug combination trials based on standard logistic
regression. Under the proposed design, we continuously update the posterior estimates of the model
parameters to make the decisions of dose assignment and early stopping. Simulation studies show
that the proposed design is competitive and outperforms some existing designs. We also extend our
design to handle delayed toxicities.
Keywords: Bayesian inference; Dose-finding; Drug combination; Oncology; Phase I trial.
1

1 Introduction
For oncologists, the objective of phase I dose-finding studies is to determine the maximum tolerated
dose (MTD), defined as the highest dose with a relatively acceptable dose-limiting toxicity (DLT)
[1, 2]. DLT is usually defined as a toxicity of grade 3 or higher according to the U.S. National
Cancer Institute toxicity criteria [3]. In practice, patients included in phase I clinical cancer trials
have already been heavily pre-treated and in many cases no alternative therapeutic options are
available to them. For cytotoxic anti-cancer drugs, a dose-toxicity effect is assumed whereby the
higher the dose, the greater the risk of DLT and the greater the efficacy.
Dose finding for drug combination trials is more difficult than that for conventional single-
agent trials due to complicated drug-drug interactions. Moreover, when combining several agents,
the order of the toxicity probabilities is not fully known. Should investigators wish to gradually
increase the acceptable level of toxicity during the trial, the appropriate order in which the doses for
the various drugs in the combination should be increased would be of great interest. For instance,
when combining two cytotoxic agents, it remains difficult to decide how to escalate or de-escalate
the dose combination, even when a partial ordering is known [4, 5].
Recently, many phase I dose-finding designs have been proposed for drug combination trials.
Thall et al. proposed a Bayesian dose-finding method based on a six-parameter model [6]. Wang
and Ivanova developed a 3-parameter model-based method in which the parameters are estimated
using Bayesian inference [7]. Mandrekar et al. proposed an approach incorporating the toxicity and
efficacy of each agent into the identification of an optimal dosing region for the combination by us-
ing a continuation ratio model to separate each agent’s toxicity and efficacy curves [8, 9]. Yin and
Yuan developed a Bayesian adaptive design based on latent 2x2 tables in which the combination’s
toxicity probabilities in the two-dimensional space are estimated using a Gumbel-type model [10].
Yin and Yuan extended their method by changing to a copula-type model to simulate the effect of
two or more drugs in combination [11]. Bailey et al. introduced a second agent as a covariate in
a logistic model [12]. Wages et al. considered an approach based on the continual reassessment
method and taking into account different orderings with partial order between combinations. In this
case, the MTD is estimated for the order associated with the highest model-selection criterion [5].
Most of these existing designs rely on complicated statistical models that typically are not familiar
to clinicians, which hinder their acceptance and application in practice. In addition, the perfor-
mance of these designs seems comparable and there is no consensus which design should be used
[13]. As a result, many of the dose-finding clinical trials conducted to evaluate drug combinations
still use the conventional “3+3” approach, which was developed for single agents and was shown
to be inefficient in terms of dose identification [14, 15, 16, 17, 18].
In this paper, we propose an Bayesian dose-finding design for drug combination trials based
on standard logistic regression. Under the Bayesian paradigm, data monitoring, early stopping
and dose assignment occur continuously throughout the trial by updating the posterior estimates of
the model parameters. Simulation studies show that in general the proposed design provide better
performance than some existing designs. To facilitate the use of the proposed design by clinicians,
R package will be developed for implementing the new design.
This manuscript is laid out as follows: in Section 2, we propose the simple statistical method
for modeling the toxicity probabilities of the drug combination under evaluation. Moreover, we
present the likelihood function and the prior specifications for the unknown parameters. In this
section, we also describe our allocation and dose-finding method, as well as propose stopping rules.
2

We conduct extensive simulation studies to examine the operating characteristics of our design in
Sections 3 and 4, and conclude with a discussion in Section 5.
2 Methods
2.1 Statistical method for combination evaluation (LOGISTIC)
Dose-combination model
Let there be a two-drug combination used in a phase I dose-finding clinical trial for which the
dose-toxicity relationship is monotonic and increases with the dose levels. Let (j, k) denote the
dose level of a combination in which j refers to Agent 1 (j = 1, . . . , J), and k refers to Agent 2
(k = 1, . . . , K). Y
i
is a Bernoulli random variable, denoting the toxicity that is equal to 1 if DLT
occurs in patient i and 0 otherwise (i = 1, . . . , N). We assume that n
j,k
patients are allocated at
combination (j, k) and that a total of t
j,k
DLTs are observed for that combination. We define π
j,k
as
the toxicity probabilities of combination (j, k), θ as the target probability of toxicity, and p
1
, ..., p
J
and q
1
, ..., q
K
as the respective prior toxicity probabilities of Agent 1 and Agent 2 taken alone. For
simplicity purposes, we refer to the selected combination as the MTD in order to maintain the same
designation as in single-agent trials. In this manuscript, we focus on finding one MTD at the end
of the trial. We note that in some cases, it is of interest to find multiple MTDs that can be further
tested in phase II trials, see Yuan and Yin [19] and Ivanova and Wang [20] for related designs.
Let u
j
and v
k
denote the “effective” or standardized doses ascribed to the jth level of Agent 1
and k level of Agent 2, respectively. We model the drug combination-toxicity relationship using a
4-parameter logistic model, as follows:
logit(π
j,k
) = β
0
+ β
1
u
j
+ β
2
v
k
+ β
3
u
j
v
k
, (1)
where β
0
, β
1
, β
2
and β
3
are unknown parameters that represent the toxicity effect of Agent 1 (β
1
),
that of Agent 2 (β
2
) and that of the interaction between the two agents (β
3
). These parameters are
defined such that β
1
> 0 and β
2
> 0, ensuring that the toxicity probability is increasing with the
increasing dose level of each agent alone, k, β
1
+ β
3
v
k
> 0 and j, β
2
+ β
3
u
j
> 0, ensuring that
the toxicity probability is increasing with the increasing dose levels of both agents together, and
intercept −∞ < β
0
< . The standardized dose of two agents are defined as u
j
= log
(
p
j
1p
j
)
and v
k
= log
(
q
k
1q
k
)
, where p
j
and q
k
are the prior estimates of the toxicity probabilities of the jth
dose level of Agent 1 and the kth dose level of Agent 2, respectively, when they are administered
individually as a single agent. Before two agents can be combined, each of them typically have
been thoroughly investigated individually. Therefore, there is often rich prior information on p
j
s
and q
k
s and their values can be readily elicited from physicians. Using the prior information to
define standardized dose has been widely used in dose-finding trial designs, and the most well
known example perhaps is the skeleton of the continuous reassessment method (CRM) [21] with a
logistic model. Research has shown that this approach improves the estimation stability and trial
performance [2, 22]. Our definition of u
j
and v
k
can be viewed as an extension of the skeleton of
the CRM (with a logistic model) to drug-combination trials.
3

Likelihood and posterior inference
Under the proposed model, the likelihood is simply a product of the Bernoulli density, given by
L(β
0
, β
1
, β
2
, β
3
|data)
J
j=1
K
k=1
π
t
j,k
j,k
(1 π
j,k
)
n
j,k
t
j,k
.
We assume the prior distributions of the model parameters are independent. For β
0
and β
3
, we
assign a vague normal prior N(0, 10) centered at 0 to indicate that a priori we do not favor either
positive or negative values for these parameters and let the observed data speak for themselves
through posteriors. For β
1
and β
2
, we assume an informative prior distribution Exp(1) centered
at 1, as those parameters should not be too far from 1. Then, the joint posterior distribution of
parameters β
0
, β
1
, β
2
and β
3
is given by
f(β
0
, β
1
, β
2
, β
3
|data) L(β
0
, β
1
, β
2
, β
3
|data)f(β
0
)f(β
1
)f(β
2
)f(β
3
). (2)
We sample this posterior distribution using Gibbs sampler, which sequentially draws each of the
parameters from their full conditional distributions (see Appendix). These full conditional dis-
tributions do not have closed forms, and we use the Adaptive Rejection Metropolis Sampling
(ARMS) method [23] to sample them. In order to impose the constrain that k, β
1
+ β
3
v
k
> 0
and j, β
2
+ β
3
u
j
> 0, at each iteration of Gibbs sampling, if sampled β
1
, β
2
and β
3
fail to satisfy
the constraint, we re-sample β
1
, β
2
and β
3
. Let (β
()
0
, β
()
1
, β
()
2
, β
()
3
)
=1,...,L
denote the L posterior
samples obtained from Gibbs sampler, the posterior toxicity probabilities can be estimated using
Monte Carlo by:
˜π
j,k
=
1
L
L
=1
exp(β
()
0
+ β
()
1
u
j
+ β
()
2
v
k
+ β
()
3
u
j
v
k
)
1 + exp(β
()
0
+ β
()
1
u
j
+ β
()
2
v
k
+ β
()
3
u
j
v
k
)
.
Dose finding algorithm and determination of the MTD
During the trial conduct, we use the dose-finding algorithm proposed by Yin and Yuan [11, 10] to
determine dose escalation and deescaltion, and propose a different criterion for MTD selection at
the end of the trial. In our design, similar to Yin and Yuan [11], we restrict dose escalation and de-
escalation one level at a time (i.e, we do not allow dose escalate or de-escalate along the diagonal)
based on the practical consideration that physicians are conservative and typically do not allow
two agent to escalate at the same time for patient safety. Nevertheless, we note that Sweeting and
Mander [24] showed that diagonal escalation strategy may be more efficient in reaching the target
toxicity level quicker with fewer patients treated at sub-optimal doses and have a higher percentage
of correct selection at the end of the trial. We take this strategy in our start-up phase described later.
Let c
e
be the probability threshold for dose escalation and c
d
the probability threshold for dose
de-escalation. We require c
e
+c
d
> 1 to avoid that the decisions of dose escalation and deescalation
occur at the same time. Our dose-finding algorithm can be described as follows:
If the current combination is (j, k) and P (π
j,k
< θ|data) > c
e
,
4

Citations
More filters
Journal ArticleDOI
TL;DR: In this article, the authors proposed a novel Bayesian adaptive design for drug-combination trials based on a robust dimension-reduction method, which continuously update the order of dose combinations and reduce the two-dimensional searching space to a one-dimensional line based on the estimated order.
Abstract: Phase I drug-combination trials are becoming commonplace in oncology. Most of the current dose-finding designs aim to quantify the toxicity probability space using certain prespecified yet complicated models. These models need to characterize not only each individual drug’s toxicity profile, but also their interaction effects, which often leads to multi-parameter models. We propose a novel Bayesian adaptive design for drug-combination trials based on a robust dimension-reduction method. We continuously update the order of dose combinations and reduce the two-dimensional searching space to a one-dimensional line based on the estimated order. As a result, the common approaches to single-agent dose finding, such as the continual reassessment method (CRM), can be applied to drug-combination trials. We further utilize the ensemble technique in machine learning, the so-called bootstrap aggregating (bagging) in conjunction with Bayesian model averaging, to enhance the efficiency and reduce the variability of the proposed method. We conduct extensive simulation studies to examine the operating characteristics of the proposed method under various scenarios. Compared with existing competitive designs, the bagging CRM demonstrates its precision and robustness in terms of pinning down the correct dose combination. We apply the proposed bagging CRM to two recent cancer clinical trials with combined drugs for dose finding.

9 citations


Cites background or methods from "A Bayesian dose-finding design for ..."

  • ...…CRM in comparison with four existing methods: the partial ordering CRM (POCRM) [Wages et al. (2011)], the Clayton copula-type regression [Yin and Yuan (2009a)], the logistic method [Riviere et al. (2014)] and the two-dimensional Bayesian optimal interval design (2d-BOIN) [Lin and Yin (2016)]....

    [...]

  • ...Hirakawa et al. (2013) developed a shrinkage logistic regression model to estimate the joint toxicity probabilities, and the logistic model is also considered in Riviere et al. (2014)....

    [...]

  • ...Such an escalation rule may be aggressive at the very beginning of the trial, but it can be more efficient in locating the target toxicity level [Riviere et al. (2014)]....

    [...]

Journal ArticleDOI
TL;DR: A “zone-finding stage” is proposed that determines the most admissible toxicity zone on the dose combination matrix and subsequently select the doses allocated to the next patient from that zone in Phase I, which would eventually improve the accuracy of optimal dose combination identification in Phase II.
Abstract: In Phase I/II trials for a combination therapy of two agents, we ideally want to explore as many dose combinations as possible with limited sample size in Phase I and to reduce the number of untrie...

9 citations


Cites methods from "A Bayesian dose-finding design for ..."

  • ...We modeled the true probability of toxicity outcome for djk, πTðdjkÞ, using an ordinary four-parameter logistic model (Riviere et al., 2014) as follows:...

    [...]

  • ...Following Riviere et al. (2014), the standardized dose of two agents is defined as sj 1⁄4 logðpj=ð1 pjÞÞ; tk 1⁄4 logðpk=ð1 pkÞÞ; uj 1⁄4 logðqj=ð1 qjÞÞ and vk 1⁄4 logðqk=ð1 qkÞÞ, where pj and pk are the prior probabilities of toxicity for agents A and B, respectively, and qj and qk are the prior probabilities of an efficacy response for agents A and B, respectively....

    [...]

  • ...Following Riviere et al. (2014), we assume a normal prior with mean 0 and variance 10 on the parameters of intercept (i.e., β0 and γ0) and interaction term (i.e., β3 and γ3) in the logistic models for toxicity and efficacy outcomes and also assume an exponential prior with a mean of 1 for β1, β2,…...

    [...]

  • ...Simulation study 2 demonstrated that the proposed and conventional methods that use the logistic model proposed by Riviere et al. (2014) were no worse than the WC method....

    [...]

  • ...We modeled the true probability of toxicity outcome for djk, πTðdjkÞ, using an ordinary four-parameter logistic model (Riviere et al., 2014) as follows: logitðπTðdjkÞÞ ¼ β0 þ β1sj þ β2tk þ β3sjtk (4) where sj and tk are the standardized doses for the jth level of agent A and k level of agent B, β0;…...

    [...]

Journal ArticleDOI
TL;DR: Three methods for Phase I combination studies that are easy to understand and straightforward to implement are reviewed and demonstrate the operating characteristics of the designs through illustration in a single trial, as well as through extensive simulation studies.
Abstract: Phase I trials evaluating the safety of multidrug combinations are becoming more common in oncology. Despite the emergence of novel methodology in the area, it is rare that innovative approaches are used in practice. In this article, we review three methods for Phase I combination studies that are easy to understand and straightforward to implement. We demonstrate the operating characteristics of the designs through illustration in a single trial, as well as through extensive simulation studies, with the aim of increasing the use of novel approaches in Phase I combination studies. Design specifications and software capabilities are also discussed.

9 citations

Journal ArticleDOI
TL;DR: The product of independent beta probabilities escalation design for dual agent phase I dose escalation trials is extended to use censored time‐to‐event toxicity outcomes for making dose escalation decisions and it is shown that trial duration can be reduced by up to 35%, particularly when recruitment is faster than expected, without compromising on other operating characteristics.
Abstract: The product of independent beta probabilities escalation (PIPE) design for dual-agent phase I dose-escalation trials is a Bayesian model-free approach for identifying multiple maximum tolerated dose combinations of novel combination therapies. Despite only being published in 2015, the PIPE design has been implemented in at least two oncology trials. However, these trials require patients to have completed follow-up before clinicians can make dose-escalation decisions. For trials of radiotherapy or advanced therapeutics, this may lead to impractically long trial durations due to late-onset treatment-related toxicities. In this paper, we extend the PIPE design to use censored time-to-event (TITE) toxicity outcomes for making dose escalation decisions. We show via comprehensive simulation studies and sensitivity analyses that trial duration can be reduced by up to 35%, particularly when recruitment is faster than expected, without compromising on other operating characteristics.

8 citations


Cites methods from "A Bayesian dose-finding design for ..."

  • ...Riviere et al. (2014) proposed a TITE extension of their Bayesian dose finding design for combination therapies that used a four-parameter logistic model, which they compared with a non-TITE version of the same design....

    [...]

Journal ArticleDOI
TL;DR: It is found that AAA enhances the chance of finding the optimal dose combinations and shortens the trial duration, and is the first design that allows for all three adaptations at the same time.
Abstract: We propose a flexible design for the identification of optimal dose combinations in dual-agent dose finding clinical trials. The design is called AAA, standing for three adaptations: adaptive model selection, adaptive dose insertion and adaptive cohort division. The adaptations highlight the need and opportunity for innovation for dual-agent dose finding and are supported by the numerical results presented in the proposed simulation studies. To our knowledge, this is the first design that allows for all three adaptations at the same time. We find that AAA enhances the chance of finding the optimal dose combinations and shortens the trial duration. A clinical trial is being planned to apply the AAA design and a Web tool is being developed for both statisticians and non-statisticians.

7 citations


Cites methods from "A Bayesian dose-finding design for ..."

  • ...In addition, on the basis of a four-parameter logistic model, Riviere et al. (2014) proposed a Bayesian adaptive design to find the MTDC, Shi and Yin (2013) applied a twodimensional escalation with overdose control design by searching the MTDC along the rows and columns of the dose matrix, and…...

    [...]

References
More filters
Journal ArticleDOI
TL;DR: A new approach to the design and analysis of Phase 1 clinical trials in cancer and a particularly simple model is looked at that enables the use of models whose only requirements are that locally they reasonably well approximate the true probability of toxic response.
Abstract: This paper looks at a new approach to the design and analysis of Phase 1 clinical trials in cancer. The basic idea and motivation behind the approach stem from an attempt to reconcile the needs of dose-finding experimentation with the ethical demands of established medical practice. It is argued that for these trials the particular shape of the dose toxicity curve is of little interest. Attention focuses rather on identifying a dose with a given targeted toxicity level and on concentrating experimentation at that which all current available evidence indicates to be the best estimate of this level. Such an approach not only makes an explicit attempt to meet ethical requirements but also enables the use of models whose only requirements are that locally (i.e., around the dose corresponding to the targeted toxicity level) they reasonably well approximate the true probability of toxic response. Although a large number of models could be contemplated, we look at a particularly simple one. Extensive simulations show the model to have real promise.

1,402 citations


"A Bayesian dose-finding design for ..." refers methods in this paper

  • ...In practice, one-dimensional CRM sometimes is used to conduct dose-combination trials [13]....

    [...]

  • ...We also compared the performance of this multidimensional designs with a one-dimensional CRM, CRM anti-diag, as described previously....

    [...]

  • ...As the CRM focused on only a subset of doses, it had more patients per dose to find the target doses than the multidimensional designs, given the same total number of patients....

    [...]

  • ...Under this method, we first preselect a subset of combinations, for which the toxicity probability order is known, and then apply the standard CRM to find the MTD....

    [...]

  • ...Using the prior information to define standardized dose has been widely used in dose-finding trial designs, and the most well-known example perhaps is the skeleton of the continuous reassessment method (CRM) [21] with a logistic model....

    [...]

Journal ArticleDOI
TL;DR: In Monte Carlo simulations, two two-stage designs are found to provide reduced bias in maximum likelihood estimation of the MTD in less than ideal dose-response settings and several designs to be nearly as conservative as the standard design in terms of the proportion of patients entered at higher dose levels.
Abstract: The Phase I clinical trial is a study intended to estimate the so-called maximum tolerable dose (MTD) of a new drug. Although there exists more or less a standard type of design for such trials, its development has been largely ad hoc. As usually implemented, the trial design has no intrinsic property that provides a generally satisfactory basis for estimation of the MTD. In this paper, the standard design and several simple alternatives are compared with regard to the conservativeness of the design and with regard to point and interval estimation of an MTD (33rd percentile) with small sample sizes. Using a Markov chain representation, we found several designs to be nearly as conservative as the standard design in terms of the proportion of patients entered at higher dose levels. In Monte Carlo simulations, two two-stage designs are found to provide reduced bias in maximum likelihood estimation of the MTD in less than ideal dose-response settings. Of the three methods considered for determining confidence intervals--the delta method, a method based on Fieller's theorem, and a likelihood ratio method--none was able to provide both usefully narrow intervals and coverage probabilities close to nominal.

816 citations


"A Bayesian dose-finding design for ..." refers methods in this paper

  • ...Therefore, as suggested by other authors [10,16,20,26,27], we implement an algorithm-based start-up phase in order to gather enough information to estimate the j,k ....

    [...]

Journal ArticleDOI
TL;DR: A robust nonlinear full probability model for population pharmacokinetic data is proposed and it is demonstrated that the method enables Bayesian inference for this model, through an analysis of antibiotic administration in new‐born babies.
Abstract: Gibbs sampling is a powerful technique for statistical inference. It involves little more than sampling from full conditional distributions, which can be both complex and computationally expensive to evaluate. Gilks and Wild have shown that in practice full conditionals are often log‐concave, and they proposed a method of adaptive rejection sampling for efficiently sampling from univariate log‐concave distributions. In this paper, to deal with non‐log‐concave full conditional distributions, we generalize adaptive rejection sampling to include a Hastings‐Metropolis algorithm step. One important field of application in which statistical models may lead to non‐log‐concave full conditionals is population pharmacokinetics. Here, the relationship between drug dose and blood or plasma concentration in a group of patients typically is modelled by using nonlinear mixed effects models. Often, the data used for analysis are routinely collected hospital measurements, which tend to be noisy and irregular. Consequently, a robust (t‐distributed) error structure is appropriate to account for outlying observations and/or patients. We propose a robust nonlinear full probability model for population pharmacokinetic data. We demonstrate that our method enables Bayesian inference for this model, through an analysis of antibiotic administration in new‐born babies.

687 citations


"A Bayesian dose-finding design for ..." refers methods in this paper

  • ...These full conditional distributions do not have closed forms, and we use the adaptive rejection Metropolis sampling (ARMS) method [23] to sample them....

    [...]

Journal ArticleDOI
TL;DR: Accelerated titration (i.e., rapid intrapatient drug dose escalation) designs appear to effectively reduce the number of patients who are under-treated, speed the completion of phase I trials, and provide a substantial increase in the information obtained.
Abstract: Background: Many cancer patients in phase I clinical trials are treated at doses of chemotherapeutic agents that are below the biologically active level, thus reducing their chances for therapeutic benefit. Current phase I trials often take a long time to complete and provide little information about interpatient variability or cumulative toxicity. Purpose: Our objective was to develop alternative designs for phase I trials so that fewer patients are treated at subtherapeutic dose levels, trials are of reduced duration, and important information (i.e., cumulative toxicity and maximum tolerated dose) needed to plan phase II trials is obtained. Methods: We fit a stochastic model to data from 20 phase I trials involving the study of nine different drugs. We then simulated new data from the model with the parameters estimated from the actual trials and evaluated the performance of alternative phase I designs on this simulated data. Four designs were evaluated. Design 1 was a conventional design (similar to the commonly used modified Fibonacci method) using cohorts of three to six patients, with 40% dose-step increments and no intrapatient dose escalation. Designs 2 through 4 included only one patient per cohort until one patient experienced dose-limiting toxic effects or two patients experienced grade 2 toxic effects (during their first course of treatment for designs 2 and 3 or during any course of treatment for design 4). Designs 3 and 4 used 100% dose steps during this initial accelerated phase. After the initial accelerated phase, designs 2 through 4 resorted to standard cohorts of three to six patients, with 40% dose-step increments. Designs 2 through 4 used intrapatient dose escalation if the worst toxicity is grade 0-1 in the previous course for that patient. Results: Only three of the actual trials demonstrated cumulative toxic effects of the chemotherapeutic agents in patients. The average number of patients required for a phase I trial was reduced from 39.9 for design 1 to 24.4, 20.7, and 21.2 for designs 2, 3, and 4, respectively. The average number of patients who would be expected to have grade 0-1 toxicity as their worst toxicity over three cycles of treatment is 23.3 for design 1, but only 7.9, 3.9, and 4.8 for designs 2, 3, and 4, respectively. The average number of patients with grade 3 toxicity as their worst toxicity increases from 5.5 for design 1 to 6.2, 6.8, and 6.2 for designs 2, 3, and 4, respectively. The average number of patients with grade 4 toxicity as their worst toxicity increases from 1.9 for design 1 to 3.0, 4.3, and 3.2 for designs 2, 3, and 4, respectively. Conclusion: Accelerated titration (i.e., rapid intrapatient drug dose escalation) designs appear to effectively reduce the number of patients who are undertreated, speed the completion of phase I trials, and provide a substantial increase in the information obtained. [J Natl Cancer Inst 1997;89:1138-47]

591 citations


"A Bayesian dose-finding design for ..." refers methods in this paper

  • ...Our start-up phase shares the spirit of accelerated titration design [28] and can be described as follows: Treat the first cohort of patients at the lowest dose combination ....

    [...]

Journal ArticleDOI
TL;DR: A simulation study shows the method's accuracy and safety are comparable with CRM's while the former takes a much shorter trial duration: a trial that would take up to 12 years to complete by the CRM could be reduced to 2–4 years by the TITE‐CRM.
Abstract: Traditional designs for phase I clinical trials require each patient (or small group of patients) to be completely followed before the next patient or group is assigned. In situations such as when evaluating late-onset effects of radiation or toxicities from chemopreventive agents, this may result in trials of impractically long duration. We propose a new method, called the time-to-event continual reassessment method (TITE-CRM), that allows patients to be entered in a staggered fashion. It is an extension of the continual reassessment method (CRM; O'Quigley, Pepe, and Fisher, 1990, Biometrics 46, 33-48). We also note that this time-to-toxicity approach can be applied to extend other designs for studies of short-term toxicities. We prove that the recommended dose given by the TITE-CRM converges to the correct level under certain conditions. A simulation study shows our method's accuracy and safety are comparable with CRM's while the former takes a much shorter trial duration: a trial that would take up to 12 years to complete by the CRM could be reduced to 2-4 years by our method.

464 citations


"A Bayesian dose-finding design for ..." refers methods in this paper

  • ...Following the Time-to-event Continual Reassesment Method (TITE-CRM) [30], we considered a weighted dose–toxicity relationship OwF ....

    [...]

  • ...Following the Time-to-event Continual Reassesment Method (TITE-CRM) [30], we considered a weighted dose–toxicity relationship O wF ....

    [...]

Frequently Asked Questions (1)
Q1. What are the contributions mentioned in the paper "A bayesian dose-finding design for drug combination clinical trials based on the logistic model" ?

The aim of this paper is to propose a Bayesian dose-finding design for drug combination trials based on standard logistic regression.