scispace - formally typeset
Open AccessJournal ArticleDOI

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

Reads0
Chats0
TLDR
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.

read more

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
Book ChapterDOI

A Comparative Study of Model-Based Dose-Finding Methods for Two-Agent Combination Trials

TL;DR: It is found that the operating characteristics of the dose-finding methods varied depending on whether the dose combination matrix is square or not, whether the true MTDCs exist within the same group consisting of the diagonals of the doses combination matrix, and the number of trueMTDCs.
Journal ArticleDOI

Benefits of Monotonicity in Safe Exploration with Gaussian Processes

Arpan Losalka, +1 more
- 03 Nov 2022 - 
TL;DR: In this paper, the authors consider the problem of sequentially maximising an unknown function over a set of actions while ensuring that every sampled point has a function value below a given safety threshold.
Journal ArticleDOI

Bayesian optimization design for finding a maximum tolerated dose combination in phase I clinical trials.

TL;DR: In this article, the authors proposed a Bayesian optimization design for identifying a single maximum tolerated dose combination in phase I clinical trials using a balance of information between exploration and exploitation on the nonparametric estimated dose-toxicity function.
Journal ArticleDOI

Quasi-partial order continual reassessment method: Applying toxicity scores to cancer dose-finding drug combination trials

TL;DR: In this article , the authors proposed a method for modeling toxicity scores in a combination-trial setting, and demonstrated that utilizing toxicity scores has the potential to greatly improve correct dose-selection over a variety of trial scenarios.
Posted Content

SDF-Bayes: Cautious Optimism in Safe Dose-Finding Clinical Trials with Drug Combinations and Heterogeneous Patient Groups

TL;DR: In this article, the authors proposed a novel Bayesian design, SDF-Bayes, for finding the maximum tolerated dose (MTD) for drug combinations in the presence of safety constraints.
References
More filters
Journal ArticleDOI

Continual reassessment method: a practical design for phase 1 clinical trials in cancer.

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.
Journal ArticleDOI

Design and analysis of phase I clinical trials.

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.
Journal ArticleDOI

Adaptive Rejection Metropolis Sampling Within Gibbs Sampling

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.
Journal ArticleDOI

Accelerated Titration Designs for Phase I Clinical Trials in Oncology

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.
Journal ArticleDOI

Sequential designs for phase I clinical trials with late-onset toxicities.

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.
Related Papers (5)
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.