ArticlePDF Available

Abstract and Figures

Background Meta-analytical methods are frequently used to combine dose-response findings expressed in terms of relative risks. However, no methodology has been established when results are summarized in terms of differences in means of quantitative outcomes. Methods We proposed a two-stage approach. A flexible dose-response model is estimated within each study (first stage) taking into account the covariance of the data points (mean differences, standardized mean differences). Parameters describing the study-specific curves are then combined using a multivariate random-effects model (second stage) to address heterogeneity across studies. Results The method is fairly general and can accommodate a variety of parametric functions. Compared to traditional non-linear models (e.g. Emax, logistic), spline models do not assume any pre-specified dose-response curve. Spline models allow inclusion of studies with a small number of dose levels, and almost any shape, even non monotonic ones, can be estimated using only two parameters. We illustrated the method using dose-response data arising from five clinical trials on an antipsychotic drug, aripiprazole, and improvement in symptoms in shizoaffective patients. Using the Positive and Negative Syndrome Scale (PANSS), pooled results indicated a non-linear association with the maximum change in mean PANSS score equal to 10.40 (95 % confidence interval 7.48, 13.30) observed for 19.32 mg/day of aripiprazole. No substantial change in PANSS score was observed above this value. An estimated dose of 10.43 mg/day was found to produce 80 % of the maximum predicted response. Conclusion The described approach should be adopted to combine correlated differences in means of quantitative outcomes arising from multiple studies. Sensitivity analysis can be a useful tool to assess the robustness of the overall dose-response curve to different modelling strategies. A user-friendly R package has been developed to facilitate applications by practitioners.
Content may be subject to copyright.
Crippa and Orsini BMC Medical Research Methodology (2016) 16:91
DOI 10.1186/s12874-016-0189-0
RESEARCH ARTICLE Open Access
Dose-response meta-analysis of
differences in means
Alessio Crippa1* andNicolaOrsini
1
Abstract
Background: Meta-analytical methods are frequently used to combine dose-response findings expressed in terms
of relative risks. However, no methodology has been established when results are summarized in terms of differences
in means of quantitative outcomes.
Methods: We proposed a two-stage approach. A flexible dose-response model is estimated within each study (first
stage) taking into account the covariance of the data points (mean differences, standardized mean differences).
Parameters describing the study-specific curves are then combined using a multivariate random-effects model
(second stage) to address heterogeneity across studies.
Results: The method is fairly general and can accommodate a variety of parametric functions. Compared to
traditional non-linear models (e.g. Emax, logistic), spline models do not assume any pre-specified dose-response curve.
Spline models allow inclusion of studies with a small number of dose levels, and almost any shape, even non
monotonic ones, can be estimated using only two parameters. We illustrated the method using dose-response data
arising from five clinical trials on an antipsychotic drug, aripiprazole, and improvement in symptoms in shizoaffective
patients. Using the Positive and Negative Syndrome Scale (PANSS), pooled results indicated a non-linear association
with the maximum change in mean PANSS score equal to 10.40 (95 % confidence interval 7.48, 13.30) observed for
19.32 mg/day of aripiprazole. No substantial change in PANSS score was observed above this value. An estimated
dose of 10.43 mg/day was found to produce 80 % of the maximum predicted response.
Conclusion: The described approach should be adopted to combine correlated differences in means of quantitative
outcomes arising from multiple studies. Sensitivity analysis can be a useful tool to assess the robustness of the overall
dose-response curve to different modelling strategies. A user-friendly R package has been developed to facilitate
applications by practitioners.
Keywords: Meta-analysis, Dose-response, Mean differences, Random-effects
Background
The identification and characterization of dose-response
relationships is an essential part of the analysis in many
scientific disciplines such as toxicology, pharmacology,
and epidemiology. This is particularly important in the
development and testing of new compounds (e.g. a new
drug, pharmaceutical treatment) where trials at different
stages aim to evaluate the efficacy of increasing levels of
dosage (Phase II-III trials) or to derive a dose-response
curve for selection of optimal doses (Phase IV trials) [1, 2].
*Correspondence: alessio.crippa@ki.se
1Department of Public Health Sciences, Karolinska Institutet, Stockholm,
Sweden
Randomized clinical trials often investigate a continu-
ous outcome variable, such as the efficacy or safety of
a drug, reporting the change from baseline of a medical
score, or the final value of a clinical measurement. The
dose-response results are typically summarized by dose-
specific means and standard deviations [3]. Measures of
effect are expressed in terms of mean or standardized
mean differences using a dose level, usually the placebo
group, as referent [1]. Over the last few years method-
ological research focused on developing and improving
methods for performing dose-response analysis in a sin-
gle study [4, 5]. A conclusive result is hardly obtained by a
single investigation and there is often the need to synthe-
size information collected from multiple studies. In such a
© 2016 The Author(s). Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and
reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver
(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Crippa and Orsini BMC Medical Research Methodology (2016) 16:91 Page 2 of 10
case meta-analytic methods can be used to define an over-
all relation or to investigate heterogeneity across study
findings.
A method for pooling aggregated dose-response data
where the outcome is a log relative risk was originally pre-
sented by Greenland and Longnecker in 1992 [6]. Since
then, several papers have refined and covered specific
aspects of the methodology such as model specification
[7, 8], modeling strategies [9, 10], and software implemen-
tation [11, 12]. Other methodological articles extended
the approach for continuous outcome but in the case
where individual patient data are available, mainly in the
context of time-series environmental studies [13–15].
Only a few alternatives have been proposed to pool
aggregated dose-response data where the findings are
summarized by differences in means. Davis and Chen
[16] in 2004 described a methodology for summariz-
ing dose-response curves of first and second generation
antipsychotics in schizoaffective patients. The authors
reconstructed drug-specific dose-response curves and
conducted a meta-analysis to compare the effectiveness of
medium vs high dosages. A common alternative to analyze
the drug effect consists of fitting a random-intercept Emax
model, where the random component accounts for het-
erogeneity in placebo effect across trials [17]. Heterogene-
ity, however, may be related to other study characteristics
rather than differences in placebo response such as imple-
mentation, participants, intervention, and outcome defi-
nition. Thomas et al. [18] adopted hierarchical Bayesian
models to summarize and describe, independently, the
distribution of study-specific model parameters derived
from an Emax model.
The mentioned strategies assumed pre-specified mod-
els that do not allow for non-monotonic curves which
may occur in practice [19], as in case of dose-response
data of antipsychotics. In addition, fitting study-specific
sigmoidal curves such as the Emax model requires
that the single studies have assessed at least three
dose levels in order to estimate model parameters.
Discarding studies not providing enough data points
represents a loss of information and may introduce
bias.
The aim of this paper is to formalize and propose a
general and flexible methodology to pool dose-response
relations from aggregated data where the changes in the
distribution of the quantitative outcome are expressed
in terms of differences in means. We first present the
data necessary for a dose-response meta-analysis and
derive formulas for obtaining effect sizes and their vari-
ance/covariance structure. We describe flexible dose-
response models with particular emphasis on regression
splines. The method is then applied to dose-response data
from clinical trials on use of aripiprazole and symptoms
improvement in schizoaffective patients.
Methods
Dose-response data
The notation and data required for a dose-response meta-
analysis for a generic study are displayed in Table 1. We
consider Istudies indexed by i=1, ...,Ireporting the
results of a common treatment at different dose levels
xij,j=1, ...,Ji,wherex0i=0 indicates the control or
placebo group in the i-th study. The study-specific results
typically consist of dose-specific means of an outcome
variable, Yij, that measures the efficacy of the j-th dose
in the i-th study [3]. Additional information about the
number of patients allocated in each treatment, nij ,and
the sample standard deviations of Yij,sdij , is generally
reported or obtained from the study-specific results.
Effect sizes and their variance/covariance
A common way to reduce heterogeneity in placebo
response is to compute the effect size (or treatment effect)
as difference between dose-specific means and placebo
mean. In case all studies measure the outcome on a com-
mon and interpretable scale, the difference can be based
on the absolute scale
dij =¯
Yij ¯
Yi0,j=1, ...,Ji,i=1, ...,I(1)
Assuming common study-specific population standard
deviations, the variance of dij is defined as
Var dij =nij +ni0
nijni0
s2
pi,j=1, ...,Ji,i=1, ...,I
(2)
where s2
pi=Ji
j=0nij 1sd2
ij/Ji
j=0nij 1is the
square of the pooled standard deviation for the i-th study.
Since the study-specific mean differences dij use the same
referent values, ¯
Yi0, they cannot be regarded as indepen-
dent. The covariance term is defined as
Cov dij,dij=Va r ¯
Yj0=s2
i0
ni0
,j= j,i=1, ...,I
(3)
Table 1 Notation for aggregated data in the i-th study used in
dose-response meta-analysis of differences in meas
dose mean(Y)asd(Y) nbdcVar (d)ddVar d
0¯
Yi0sdi0ni00– 0
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
xij ¯
Yij sdij nij dij Var dijd
ij Var d
ij
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
.
xiJi¯
YiJisdiJiniJidiJiVar diJid
iJiVar d
iJi
aY is the continuous outcome
bNumber of patients
cMean difference
dStandardized mean difference
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Crippa and Orsini BMC Medical Research Methodology (2016) 16:91 Page 3 of 10
In case the outcome is measured on different scales the
effect sizes can be based on standardized mean differences
d
ij =¯
Yij ¯
Yi0
spi
,j=1, ...,Ji,i=1, ...,I(4)
with
Var d
ij=1
nij
+1
ni0
+d
ij
2
2Ji
j=0nij
,j=1, ...,Ji,
i=1, ...,I
Cov d
ij,d
ij=1
ni0
+
d
ijd
ij
2Ji
j=0nij
,j= j,i=1, ...,I
(5)
Dose-response analysis
The chosen effect sizes and the corresponding
(co)variances are used to estimate the study-specific dose-
response curves. The dose-response curves characterize
the relative efficacy of the dose under investigation using
the placebo effect as referent (i.e. the relative efficacy
for the placebo is zero by definition). The dose-response
models are expressed through the parametric model f,
which specifies how the effect size varies according to the
dose values. The functional relationship fis parametrized
in terms of θi,thep×1 vector of dose-response coef-
ficients. We consider the case of mean differences, dij,
but the same principles apply for standardized mean
differences, d
ij. The study-specific curves can be written
as
di=f(xi,θi)+εi,εiN0,
ˆ
i,i=1, ...,I
(6)
ˆ
iis the covariance matrix of the residual error term,
with Var dijalong the diagonal and Cov dij,dijoff-
diagonal.
Several alternatives are available to model the dose-
response curve (i.e. for the choice of f). Table 2 shows
the definition of 4 types of models ordered according to
the number of parameters, ranging from 1 for a linear
modelupto3foralogisticmodel.SeeBretzetal.fora
comprehensive description [2].
The most common choice in dose findings [2] is the use
of the Emax model which is expressed in terms of three
Table 2 Frequently used dose-response models
Model Equation No. of parameters
Linear E di|xi=θ1ixi1
Quadratic E di|xi=θ1ixi+θ2ix2
i2
Emax Edi|xi=θ1ixθ3i
i/θ2i+xθ3i
i3
Logistic E di|xi=θ1i/{1+exp [2ixi)]3i}3
parameters: the maximum effect (θ1i),thedosetopro-
duce half of the maximum effect (θ2i) and the steepness
of the curve (θ3i) [20]. As other non-linear models, the
Emax model assumes a specific shape that does not allow
for non-monotonic curve and its estimation requires at
least three non reference dose levels. Quadratic models
are defined by only p=2 coefficients butmay poorly fit at
extreme dose values [9]. Other non-linear models such as
logistic and sigmoidal models, are commonly defined by
p3 coefficients so that study-specific aggregated data
may not be sufficient to estimate the parameters.
We propose the use of regression splines to flexibly
model the dose of interest. Splines represent a family of
smooth functions that can describe a wide range of curves
(i.e. U-shaped, J-shaped, S-shaped, threshold) [21]. The
curves consist of piecewise polynomials over consecu-
tive intervals defined by kknots. Their use may facilitate
curve fitting since many non-linear curves can be exam-
ined by estimating only a small number of coefficients.
For instance, a restricted cubic spline model with three
knots k=(k1,k2,k3)is defined only in terms of p=2
coefficients [22]
E[di|xi]=θ1ix1i+θ2ix2i(7)
with two transformations [23] defined as
x1=x
x2=(xk1)3
+k3k1
k3k2(xk2)3
++k2k1
k3k2(xk3)3
+
(k3k1)2
(8)
wherethe‘+’notation,withu+=uif u0andu+=0
otherwise, has been used.
An alternative flexible approach to model the dose-
response association is represented by fractional poly-
nomials. In particular, a dose-response model based on
fractional polynomial of order two can be written as in
Eq. 6 with the two transformations defined as
x1=xp1and x2=xp2if p1= p2
x1=xp1and x2=xp1log(x)if p1=p2(9)
for each combination of p1and p2in the predefined set of
values {2, 1, 0.5, 0, 0.5, 1, 2, 3};forp=0, xpbecomes
log(x). The best fitting fractional polynomial is typically
chosen based on the Akaike’s Information Criterion [24].
Once the functional relation fhas been selected, gen-
eralized least square estimation can be performed to effi-
ciently estimates the dose-response coefficients
ˆ
θjand the
corresponding (co)variance matrix
ˆ
Vj, by minimizing
dif(xi,θi)Tˆ
1
idif(xi,θi)(10)
that generally requires numerical optimization algo-
rithms. If fis a linear combination of the parameters
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Crippa and Orsini BMC Medical Research Methodology (2016) 16:91 Page 4 of 10
θi, as in the case of regression splines and fractional
polynomials, the close solution can be written as
ˆ
θi=XT
i
ˆ
1
iXi1
XT
i
ˆ
1
idi
ˆ
Vi=Var ˆ
θi=XT
i
ˆ
1
iXi1(11)
where Xiindicates the Ji×pdesign matrix in the i-th study.
Meta-analysis
The estimated study-specific dose-response coefficients
ˆ
θiand the accompanying (co)variance matrices
ˆ
Viare
combined by means of multivariate meta-analysis
ˆ
θiNθ,
ˆ
Vi+(12)
A fixed-effects model assumes no statistical hetero-
geneity among study results, i.e. differences in the dose-
response coefficients are only related to sampling error.
The assumption of homogeneity may not hold in prac-
tice, unless it is known that the studies are performed
in a similar way and are sampled from the same popula-
tion [25]. The Cochran’s Q test [26] is typically used to
test statistical heterogeneity across studies (H0:=0)
[27]. Selected studies, however, will typically differ with
respect to study design and implementation, selection of
participants, and type of analyses. A certain degree of het-
erogeneity is expected and should be taken into account
in the analysis. A random-effects model allows the dose-
response coefficients, θi, to vary across studies. Statistical
heterogeneity is captured by the between-studies variance
while θrepresents the mean of the distribution of dose-
response coefficients and an estimate,
ˆ
θ, can be obtained
using (restricted) maximum likelihood estimation [15].
As a final result, the pooled dose-response curve can be
presented in either a graphical or tabular form by predict-
ing the mean differences of the outcome for a set of xdose
values
E[
ˆ
d|x]=fX,
ˆ
θ(13)
with an approximate (1α/2)% confidence interval (CI),
that in case fis a linear combination of θcan be expressed
as
E[
ˆ
d|x]zα
2diag fX,
ˆ
θTCov ˆ
θfX,
ˆ
θ(14)
where zα/2is the α/2-th quantile of a standard normal
distribution.
Dose findings
Once the pooled dose-response curve has been estimated,
it may be of interest to determine a set of target doses,
i.e. doses associated with prespecified outcome effects.
In development of new compounds it is often important
to select an optimal dose which is almost as effective as
the maximum effective dose but has less undesired side
effects, which often occur at high dosages. Suppose one
wants to determine which is the lowest dose (EDγ)topro-
duce an almost complete effect, e.g. γ%oftheobserved
maximum predicted response.
The EDγcan be determined as
EDγ=argmax
x(0,xmax]E[ ˆ
d|x]
E[ ˆ
d|xmax]γ(15)
where xmax is the dose corresponding to the maximum
predicted outcome.
An important step when presenting results from dose
findings analysis is to accompany the previous estimates
with a measure of precision, typically confidence intervals.
Pinheiro et al. [3] proposed the use a parametric bootstrap
approach based on the asymptotic normal distribution of
ˆ
θ, the pooled estimate of the dose-response coefficients.
The approach consists in re-sampling the dose-response
coefficients θfrom its approximate normal distribution
and derive the distribution of
EDγbased on the sam-
ples. Approximated confidence intervals for
EDγcan be
constructed using percentiles of the sampling distribution.
Results
To illustrate the methodology we examined the
dose-response relation between aripiprazole, a second-
generation antipsychotic, and symptoms improvement in
schizoaffective patients. We updated the search strategy
presented in a previous review by Davis and Chen [16]
by searching the Medline, International Pharmaceutical
Abstracts,CINAHL,andtheCochraneDatabaseofSys-
tematic Reviews. To reduce the exclusion of unpublished
papers, additional sources including Food and Drug
administration website, data from Cochrane reviews,
poster presentations and conference abstracts were also
searched. All random-assignment, double-blind, con-
trolled clinical trials of schizoaffective patients providing
dose-response results for at least two non-zero dosages of
aripiprazole were eligible.
Five studies [28–32] met the inclusion criteria and were
included in the analysis. All the studies reported mean
changes from baseline as main outcome variable, using
the Positive and Negative Syndrome Scale (PANSS). The
PANSS scale is an ordinal score derived from 30 items
ranging from 1 to 7. Computations of ratios such as per-
centage changes are not directly applicable and may lead
to erroneous results [33, 34]. To address this issue, the the-
oretical minimum (i.e. 30) needs to be subtracted from
the original score [35]. Information about the means, the
number of patients assigned to each treatment, and the
standard deviation was available from the published data.
Because all the studies measured the outcome variable on
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Crippa and Orsini BMC Medical Research Methodology (2016) 16:91 Page 5 of 10
the same scale, we computed PANSS mean differences as
effect sizes. Data are reported in Table 3.
We used the trial by Cutler et al. [28] to illustrate the
steps required for estimating the dose-response curve for
a single study. For example, the difference in mean PANSS
comparing the dose of 2 mg/day relative to 0 mg/day
is d11 =8.23 5.3 =2.93 mg/day. Its variance is
Var (d11 )=(85 +92)/(85 ×92)×s2
p1= 7.59, where
s2
p1=119, 419/356 =335.4. The covariance of this
difference in means PANSS is 18.312/85 =3.94. The vari-
ance/covariance structure associated with the vector of
differences in means for this trial d1can be presented in a
matrix form
ˆ
1=
7.59
3.94 7.72
3.94 3.94 7.52
Toestimatethedose-responsecurveweneedfirstto
specify the model f. We characterized the dose-response
relation using a restricted cubic spline model with three
knots located at the 10th, 50th, and 90th percentiles
(0, 10, and 30 mg/day) of the overall dose distribu-
tion (p= 2). The restricted cubic spline dose-response
model is defined as in Eq. 7. Efficient estimates of the
dose-response coefficients and (co)variance matrix were
obtained by generalized least square estimation
ˆ
θ1=(1.215, 5.738)T
ˆ
V1=Var ˆ
θ1=0.49
3.65 31.64 (16)
We applied the same procedure to the other stud-
ies included in the meta-analysis in order to obtain the
study-specific
ˆ
θiand
ˆ
Vi,i=1, ..., 5 (Table 4). The study-
specific predicted curves are presented in Fig. 1. Under
a random-effects model, restricted maximum likelihood
estimates were
ˆ
θ=(0.937, 1.156)T
Cov ˆ
θ=0.03
0.05 0.10 (17)
Ap-value <0.001 for the multivariate Wald-type test
H0:θ=0provided strong evidence against the null
hypothesis of no relation between different doses of arip-
iprazole and mean change PANSS score. The Q test (Q=
3.5, p-value = 0.899) did not detect substantial statistical
heterogeneity across studies.
To communicate results of the pooled dose-response
analysis, we can estimate the pooled mean differences in
PANSS scores using 0 mg/day as referent as 0.937x1
1.156x2, together with the corresponding 95 % confidence
interval for a generic dose xof interest as following
(0.937x11.156x2)1.960.03x2
1+0.1x2
20.1x1x2
Table 3 Aggregated dose-response data of five clinical trials investigating effectiveness of different dosages of aripiprazole in
schizoaffective patients. The continuous outcome is measured on the Positive and Negative Syndrome Scale and summarized by
mean values (mean(Y)) and standard deviations (sd(Y))
ID Author, Year dose mean(Y) sd(Y) n d Var(d)
1 Cutler, 2006 [28] 0 5.300 18.310 85 0.000 0.000
2 8.230 18.320 92 2.930 7.593
5 10.600 18.310 89 5.300 7.715
10 11.300 18.320 94 6.000 7.515
2 McEvoy, 2007 [29] 0 2.330 26.100 107 0.000 0.000
10 15.040 27.600 103 12.710 13.344
15 11.730 26.200 103 9.400 13.344
20 14.440 25.900 97 12.110 13.764
3 Kane, 2002 [30] 0 2.900 24.280 102 0.000 0.000
15 15.500 26.490 99 12.600 12.038
30 11.400 22.900 100 8.500 11.977
4 Potkin, 2003 [31] 0 5.000 21.140 103 0.000 0.000
20 14.500 20.160 98 9.500 8.563
30 13.900 20.880 96 8.900 8.654
5 Study 94202 [32] 0 1.400 25.730 57 0.000 0.000
2 11.000 25.000 51 9.600 25.447
10 11.500 25.200 51 10.100 25.447
30 15.800 28.510 54 14.400 24.701
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Crippa and Orsini BMC Medical Research Methodology (2016) 16:91 Page 6 of 10
Table 4 Study-specific dose-response coefficients and corresponding covariances for different dose-response models considered in
the analysis
Model id ˆ
θ1ˆ
θ2Var ˆ
θ1Cov ˆ
θ1,ˆ
θ2Var ˆ
θ2
Restricted cubic splines 1 0.55 0.55 0.065 0.065 0.065
2 0.59 0.59 0.031 0.031 0.031
30.841.69 0.054 0.12 0.38
40.470.54 0.021 0.043 0.15
50.781.25 0.23 0.62 1.8
Fractional Polynomials 1 17.47 7.84 2.9e+02 2.2e+02 1.8e+02
2 29.59 12.42 2.5e+02 1.6e+02 1e+02
3 32.12 13.47 1.4e+02 70 37
4 18.48 6.42 1.8e+02 93 49
5 21.97 7.00 2.3e+02 1.1e+02 55
Emax 1 8.13 3.13 27 24 36
2 11.39 0.00 38 35 42
3 10.54 0.00 37 53 1e+02
4 9.20 0.00 60 1.4e+02 3.6e+02
5 13.28 0.94 23 2.7 2.7
Quadratic 1 1.54 0.09 0.96 0.086 0.0083
21.540.05 0.35 0.017 0.00089
31.400.04 0.18 0.0055 0.00018
40.830.02 0.14 0.0047 0.00017
51.080.02 0.51 0.016 0.00051
Piecewise linear 1 0.55 0.065
2 0.59 0.031
30.841.69 0.054 0.12 0.38
40.470.54 0.021 0.043 0.15
50.781.25 0.23 0.62 1.8
where x1and x2are defined as in Eq. 8. For instance,
the model-based predicted mean changes in PANSS score
compared to placebo were 4.52 (95 % CI: 2.96, 6.08) for
5 mg/day, 8.08 (95 % CI: 5.43, 10.73) for 10 mg/day, 9.95
(95 % CI: 6.97, 12.94) for 15 mg/day, 10.38 (95 % CI: 7.49,
13.27) for 20 mg/day, 9.84 (95 % CI: 6.86, 12.83) for 25
mg/day, and 8.83 (95 % CI: 5.11, 12.54) for 30 mg/day. The
pooled predicted dose-response curve together with the
confidence intervals and the model mean differences is
provided in Fig. 2.
The results indicated a statistically significant positive
association between increasing doses of aripiprazole and
the mean change in PANSS score with the maximum value
of 10.39 (95 % CI: 7.48, 13.30) observed at xmax = 19.32
mg/day. The model suggested a slight decrease in the pre-
dicted mean PANSS score for dosages greater than 20
mg/day. The estimated dose to produce 50 % and 80 % of
the predicted maximum effect were
ED50 =5.82 mg/day
(95 % CI: 5.10, 8.58) and
ED80 =10.43 mg/day (95 % CI:
9.02, 16.73).
Sensitivity analysis
A sensitivity analysis is often required to evaluate the
robustness of the pooled dose-response curve. In the
spline model, for example, the location of the knots may
affect the shape of the dose-response curve. Therefore
we considered alternative knots locations including differ-
ent combinations of the 10th, 25th, 50th, 75th and 90th
percentiles of the overall dose distribution (0, 0.5, 10,
18.75, and 30 mg/day). A graphical comparison is pre-
sented in the left panel of Fig. 3. The alternative curves
roughly described the same dose-response shape with
no substantial variation, all indicating an increase in the
mean change PANSS score up to 20 mg/day of arip-
iprazole. We can assess whether there is an increasing
trend above 20 mg/day by simply re-defining x2equal
to (x20)+in Eq. 8; this approach is known as piece-
wise linear model. The rate of change in the PANSS
mean differences was negative and not statistically sig-
nificant (θ1+θ2=-0.284, p= 0.18) after 20 mg/day of
aripiprazole.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Crippa and Orsini BMC Medical Research Methodology (2016) 16:91 Page 7 of 10
0 5 10 15 20 25 30
0
5
10
15
20
25
Aripiprazole (mg/day)
Mean Difference
Cutler 2006
0 5 10 15 20 25 30
0
5
10
15
20
25
Aripiprazole (mg/day)
Mean Difference
McEvoy 2007
0 5 10 15 20 25 30
0
5
10
15
20
25
Aripiprazole (mg/day)
Mean Difference
Kane 2002
0 5 10 15 20 25 30
0
5
10
15
20
25
Aripiprazole (mg/day)
Mean Difference
Potkin 2003
0 5 10 15 20 25 30
0
5
10
15
20
25
Aripiprazole (mg/day)
Mean Difference
Study 94202
Fig. 1 Study-specific mean differences in Positive and Negative Syndrome Scale score for increasing dosages of aripiprazole. The first author and
year of publication of the subjects included in the original analyses are reported. Black squares indicate the mean differences and whiskers their
95 % confidence interval. Ariprazole dosage was modeled with restricted cubic splines. Solid lines represent the estimated dose-response curves,
dashed lines the corresponding 95 % confidence intervals. The placebo group (dose = 0) served as the referent group
Fig. 2 Pooled dose-response association between aripiprazole and mean change in Positive and Negative Syndrome Scale score (solid line).
Aripiprazole dosage was modeled with restricted cubic splines in a random-effects model. Dash lines represent the 95 % confidence intervals for the
spline model. The placebo group (dose = 0) served as the referent group. Circles indicate observed mean differences in individual studies; size of
bubbles is proportional to precision (inverse of variance) of the mean differences. Right axis represents percentage of the maximum predicted effect
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Crippa and Orsini BMC Medical Research Methodology (2016) 16:91 Page 8 of 10
Fig. 3 Graphical sensitivity analysis for the pooled dose-response curves between aripiprazole and mean change in Positive and Negative Syndrome
Scale score. The placebo group (dose = 0) served as the referent group. Right axis represents percentage of the maximum predicted effect. Left
panel: different location of the three knots in a restricted cubic spline model. Right panel: different models, restricted cubic splines (solid line),
fractional polynomials (dashed line), quadratic polynomial (dotted line), and Emax model (dot-dashed line). Circles indicate observed mean differences
in individual studies; size of bubbles is proportional to precision (inverse of variance) of the mean differences
To evaluate the sensitivity of the dose-response curve
to the choice of the parametric model fadopted, instead,
we considered three alternatives: fractional polynomials;
quadratic; and Emax. Since two studies only had two non-
referent doses, the study-specific (sigmoidal) Emax models
as described in Table 2 cannot be estimated. A common
solution is to fix the steepness of the curve θ3to be 1, also
referred to as hyperbolic Emax [20].
The “best” fractional polynomials (p1=0.5, p2=1)
provided overall a similar dose-response curve when com-
pared to the spline model, with slightly higher value for the
maximum predicted response (right panel of Fig. 3). The
hyperbolic Emax had substantially higher predicted mean
differences for low values of the dose. The non-linear
model assumes a specific hyperbolic dose-response curve
that did not seem to fit the data and may be dependent
from the choice of fixing θ3to be 1. The dose-response
curve described by the quadratic model fall in between the
spline and the hyperbolic Emax curves.
Discussion
In this paper we proposed a statistical method to com-
bine differences in means of quantitative outcomes.
The method consists of dose-response models estimated
within each study (first stage) and an overall curve
obtained by pooling study-specific dose-response coef-
ficients (second stage). The covariance among study-
specific mean differences is taken into account in the first
stage analysis using generalized least square estimators,
while statistical heterogeneity across studies is allowed by
multivariate random-effects model in the second stage.
One major strength of the proposed method is that
it is fairly general and can accommodate different mod-
eling strategies, including non-linear ones described by
Pinheiro et al. [3]. Non-linear models, however, are
defined by at least three or four parameters, and hence
require an equal number of dose levels for each single
study included in the analysis. Given that some stud-
ies may have investigated a lower number of dose lev-
els, exclusion of these studies may result in substantial
loss of information. In addition, many non-linear models
assume a specific behaviour (e.g. monotonicity) requir-
ing a strong a priori information about the dose-response
curve. The choice of the parametric model is critical,
since it highly influences the final results [3]. Indeed, the
selection of the dose-response model should be informed
by subject-matter knowledge as well as understanding of
the research questions at hand. We presented the use of
regression splines as a flexible tool for modeling any quan-
titative exposure. The major advantage is that a variety
of curves, even non monotonic ones, can be estimated
using only two parameters. It is considered to be closed
to non-parametric regression, since no major assump-
tions about the shape of the curve are needed [9]. A
possible alternative is the use of fractional polynomials.
In comparing the two strategies, we did not find impor-
tant differences between the two strategies and concluded
that both are useful tools to characterize a (non-linear)
dose-response curve. Nonetheless a sensitivity analysis
is generally required to evaluate the robustness of the
combined results.
A possible limitation of the proposed methodology is
that it requires information about dose-specific means
and standard deviations. Studies providing other sum-
mary measures, such as dose-specific medians, would
not be included the analysis. The dose-response analysis
presented in Eq. 6 is based on the asymptotic normal dis-
tribution of the conditional mean effect size. Extension of
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Crippa and Orsini BMC Medical Research Methodology (2016) 16:91 Page 9 of 10
the introduced methodology to percentiles is not straight-
forward and may represent an interesting topic of future
research.
An additional limitation of aggregated dose-response
data is that supplementary information for approximat-
ing the covariance terms may not be available. Articles
may report directly mean or standardized mean differ-
ences and standard errors for non-referent dose groups.
Whenever the standard deviation for the outcome vari-
able in the control group (s2
i0) cannot be obtained, it
may be approximated using the pooled standard deviation
based on the non-referent dose levels (s2
pij ). Alternatively
a specific value may be imputed and a sensitivity anal-
ysis can be performed to evaluate how the results of
the meta-analysis vary for different values of s2
0j.Fur-
ther limitations relate to the general application of meta-
analysis based on aggregated data. These include restric-
tions in subgroup analyses, the impossibility of assessing
the appropriateness of individual analyses, and to har-
monize variable definitions and analyses for reducing the
extent of heterogeneity, as well as specific biases such as
aggregation (or ecological) bias in meta-regression mod-
els. Meta-analysis of individual patient data, however, are
often difficult to undertake especially for the availabil-
ity of individual data, so that usage of aggregated data
may represent the only alternative [36]. Specific to aggre-
gated dose-response data, different dose references and
exposure range may complicate the analysis. The pre-
sented methodology assume that all the selected studies
share a common dose-response model. Important depar-
ture from this assumption may limit and/or impact the
pooling of individual dose-response coefficients. An alter-
native methods has been proposed based on a series of
univariate meta-analyses of effect sizes for a pre-specified
grid of dose-levels [37]. Further work is needed to analyze
this possibility and potential advantages. Depending on
the extent of heterogeneity of the dose-response curves,
however, it may not be opportune to pool study-specific
results, and meta-regression or stratified analyses should
be performed [38].
In our application, we considered the effectiveness
of increasing dosages of aripiprazole in shizoaffective
patients. We described the steps needed to obtain the
overall dose-response curve and to present it in a graph-
ical form. We observed a non-linear association with the
maximum efficacy corresponding to aripiprazole 19.32
mg/day. An estimated dose of 10.46 mg/day, however,
may be sufficient to obtain 80 % of the maximum effect,
which may be relevant for avoiding possible undesired
side effects. Sensitivity analysis showed similar results as
compared to fractional polynomials. The Emax model pre-
sented higher drug efficacy for low dosages. Compared to
the previous models, the Emax model did not seem to fit
properly the data at low dosages.
Conclusions
We described an approach to combine differences in
means of a quantitative outcome contrasting different
dose levels relative to a placebo in randomized trials.
The general framework of the proposed methodology can
include a variety of flexible models. Sensitivity analysis
can be a useful tool to assess the stability of the over-
all dose-response curve to different modelling strategies.
Although the method was presented for the analysis of
randomized trials, it may be extended to observational
studies where mean differences are further adjusted for
potential confounders. Future work is needed to evalu-
ate the properties of the statistical model and validity of
the underlying assumptions. A user friendly procedure is
implemented in the dosresmeta R package [39] with
worked examples available on GitHub.
Abbreviations
PANSS, positive and negative syndrome scale
Acknowledgements
We are grateful to Dr. Stefan Leucht and John M. Davis for providing the data
and raising the methodological question under study.
Funding
This work was supported by Karolinska Institutet’s funding for doctoral
students (KID-funding) (AC) and by a Young Scholar Award from the
Karolinska Institutet’s Strategic Program in Epidemiology (SfoEpi) (NO).
Availability of data and materials
The data on the effectiveness of aripiprazole are publicly available and also
contained in dosresmeta R package on github [39] (https://github.com/
alecri/dosresmeta/blob/master/data/ari.rda).
Authors’ contributions
AC developed the methods and prepared a draft. NO provided critical reviews,
corrections and revisions. Both authors read and approved the final version of
the manuscript.
Authors’ information
AC is a PhD student in Epidemiology and Biostatistics. Dr. NO is Associate
Professor of Medical Statistics.
Competing interests
The authors declare that they have no competing interest.
Consent for publication
Not applicable.
Ethics approval and consent to participate
Not applicable.
Received: 27 October 2015 Accepted: 13 July 2016
References
1. Bretz F, Pinheiro JC, Branson M. Combining multiple comparisons and
modeling techniques in dose-response studies. Biometrics. 2005;61(3):
738–48. doi:10.1111/j.1541-0420.2005.00344.x. Accessed 24 Feb 2015.
2. Bretz F, Hsu J, Pinheiro J, Liu Y. Dose finding - a challenge in statistics.
Biometrical Journal. Biometrische Zeitschrift. 2008;50(4):480–504.
doi:10.1002/bimj.200810438.
3. Pinheiro JC, Bretz F, Branson M. Analysis of dose–response studies.
modeling approaches. In: Dose Finding in Drug Development. New York
City: Springer; 2006. p. 146–71.
4. Ting N. Dose finding in drug development. New York City: Springer; 2006.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Crippa and Orsini BMC Medical Research Methodology (2016) 16:91 Page 10 of 10
5. Chevret S. Statistical methods for dose-finding experiments. New York
City: Wiley; 2006.
6. Greenland S, Longnecker MP. Methods for trend estimation from
summarized dose-response data, with applications to meta-analysis. Am J
Epidemiol. 1992;135(11):1301–1309.
7. Berlin JA, Longnecker MP, Greenland S. Meta-analysis of epidemiologic
dose-response data. Epidemiology. 1993;4(3):218–28.
8. Dumouchel W. Meta-analysis for dose–response models. Stat Med.
1995;14(5-7):679–85.
9. Bagnardi V, Zambon A, Quatto P, Corrao G. Flexible meta-regression
functions for modeling aggregate dose-response data, with an
application to alcohol and mortality. Am J Epidemiol. 2004;159(11):
1077–1086. doi:10.1093/aje/kwh142. Accessed 25 Nov 2013.
10. Liu Q, Cook NR, Bergström A, Hsieh CC. A two-stage hierarchical
regression model for meta-analysis of epidemiologic nonlinear
dose–response data. Comput Stat Data Anal. 2009;53(12):4157–167.
11. Orsini N, Bellocco R, Greenland S, et al. Generalized least squares for
trend estimation of summarized dose-response data. Stata J. 2006;6(1):40.
12. Orsini N, Li R, Wolk A, Khudyakov P, Spiegelman D. Meta-analysis for
linear and nonlinear dose-response relations: examples, an evaluation of
approximations, and software. Am J Epidemiol. 2012;175(1):66–73.
13. Dominici F, Samet JM, Zeger SL. Combining evidence on air pollution
and daily mortality from the 20 largest us cities: a hierarchical modelling
strategy. J Royal Stat Soc Ser A (Stat Soc). 2000;163(3):263–302.
14. Simmonds MC, Higginsa JP, Stewartb LA, Tierneyb JF, Clarke MJ,
Thompson SG. Meta-analysis of individual patient data from randomized
trials: a review of methods used in practice. Clin Trials. 2005;2(3):209–17.
15. Gasparrini A, Armstrong B, Kenward MG. Multivariate meta-analysis for
non-linear and other multi-parameter associations. Stat Med. 2012;31(29):
3821–839. doi:10.1002/sim.5471. Accessed 11 Dec 2013.
16. Davis JM, Chen N. Dose response and dose equivalence of
antipsychotics. J Clin Psychopharmacol. 2004;24(2):192–208.
17. Holford NH, Sheiner LB. Understanding the dose-effect relationship:
clinical application of pharmacokinetic-pharmacodynamic models. Clin
Pharmacokinet. 1981;6(6):429–53.
18. Thomas N, Sweeney K, Somayaji V. Meta-analysis of clinical
dose-response in a large drug development portfolio. Stat Biopharm Res.
2014;6(4):302–17. doi:10.1080/19466315.2014.924876. Accessed 11 Feb
2015.
19. Geddes J, Freemantle N, Harrison P, Bebbington P. Atypical
antipsychotics in the treatment of schizophrenia: systematic overview
and meta-regression analysis. BMJ : British Medical Journal.
2000;321(7273):1371–1376. Accessed 24 Mar 2015.
20. Macdougall J. Analysis of dose–response studies. emax model. In: Dose
Finding in Drug Development. New York City: Springer; 2006. p. 127–45.
21. Durrleman S, Simon R. Flexible regression models with cubic splines. Stat
Med. 1989;8(5):551–61. doi:10.1002/sim.4780080504. Accessed30 Mar 2015.
22. Desquilbet L, Mariotti F. Dose-response analyses using restricted cubic
spline functions in public health research. Stat Med. 2010;29(9):
1037–1057. doi:10.1002/sim.3841. Accessed 25 Nov 2013.
23. Harrell F.E. Jr, Lee KL, Pollock BG. Regression models in clinical studies:
determining relationships between predictors and response. J Natl
Cancer Inst. 1988;80(15):1198–1202.
24. Royston P. A strategy for modelling the effect of a continuous covariate in
medicine and epidemiology. Stat Med. 2000;19(14):1831–1847.
25. Jackson D, White IR, Riley RD. Quantifying the impact of between-study
heterogeneity in multivariate meta-analyses. Stat Med. 2012;31(29):
3805–820.
26. Cochran WG. The combination of estimates from different experiments.
Biometrics. 1954;10(1):101–29. doi:10.2307/3001666. Accessed 09 Jan
2015.
27. Ritz J, Demidenko E, Spiegelman D. Multivariate meta-analysis for data
consortia, individual patient meta-analysis, and pooling projects. J Stat
Plann Infer. 2008;138(7):1919–1933.
28. Cutler AJ, Marcus RN, Hardy SA, O’Donnell A, Carson WH, McQuade RD.
The efficacy and safety of lower soses of aripiprazole for the treatment of
patients with acute exacerbation of schizophrenia. CNS Spectrums.
2006;11(09):691–702. doi:10.1017/S1092852900014784. Accessed 06 Feb
2015.
29. McEvoy JP, Daniel DG, Carson WH, McQuade RD, Marcus RN. A
randomized, double-blind, placebo-controlled, study of the efficacy and
safety of aripiprazole 10, 15 or 20 mg/day for the treatment of patients
with acute exacerbations of schizophrenia. J Psychiatr Res. 2007;41(11):
895–905. doi:10.1016/j.jpsychires.2007.05.002.
30. Kane JM, Carson WH, Saha AR, McQuade RD, Ingenito GG, Zimbroff DL,
Ali MW. Efficacy and safety of aripiprazole and haloperidol versus placebo
in patients with schizophrenia and schizoaffective disorder. J Clin
Psychiatry. 2002;63(9):763–71.
31. Potkin SG, Saha AR, Kujawa MJ, Carson WH, Ali M, Stock E, Stringfellow
J, Ingenito G, Marder SR. Aripiprazole, an antipsychotic with a novel
mechanism of action, and risperidone vs placebo in patients with
schizophrenia and schizoaffective disorder. Arch Gen Psychiatr.
2003;60(7):681–90. doi:10.1001/archpsyc.60.7.681.
32. Turner EH, Knoepflmacher D, Shapley L. Publication bias in antipsychotic
trials: an analysis of efficacy comparing the published literature to the US
Food and Drug Administration Database. PLoS Med. 2012;9(3).
doi:10.1371/journal.pmed.1001189. Accessed 30 Mar 2015.
33. Obermeier M, Schennach-Wolff R, Meyer S, Möller HJ, Riedel M, Krause
D, Seemüller F. Is the panss used correctly? a systematic review. BMC
psychiatry. 2011;11(1):1.
34. Leucht S, Kissling W, Davis JM. The PANSS should be rescaled.
Schizophrenia bulletin. 2010;36:461–462.
35. Leucht S, Davis JM, Engel RR, Kane JM, Wagenpfeil S. Defining response
in antipsychotic drug trials: recommendations for the use of scale-derived
cutoffs. Neuropsychopharmacology. 2007;32(9):1903–1910.
36. Lyman GH, Kuderer NM. The strengths and limitations of meta-analyses
based on aggregate data. BMC Med Res Methodol. 2005;5(1):14.
37. Sauerbrei W, Royston P. A new strategy for meta-analysis of continuous
covariates in observational studies. Stat Med. 2011;30(28):3341–360.
38. Greenland S. Invited commentary: a critical look at some popular
meta-analytic methods. Am J Epidemiol. 1994;140(3):290–6.
39. Crippa A, Orsini N. Multivariate dose-response meta-analysis: the
dosresmeta r package. J Stat Softw. 2016. In Press.
We accept pre-submission inquiries
Our selector tool helps you to find the most relevant journal
We provide round the clock customer support
Convenient online submission
Thorough peer review
Inclusion in PubMed and all major indexing services
Maximum visibility for your research
Submit your manuscript at
www.biomedcentral.com/submit
Submit your next manuscript to BioMed Central
and we will help you at every step:
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Based on the above consideration, the aim of this meta-analysis, using novel meta-analytic techniques [i.e., dose-response metaanalysis (22,23)], is to assess the effect of OXT treatment relative to placebo and to address the optimal dose of OXT to improve social impairments and repetitive behaviors. The dose-response meta-analysis plays a crucial role in examining the relationship between independent variables (e.g., OXT dosages) and disorder outcomes (e.g., social impairments and repetitive behaviors), which can provide a preliminary estimation for the effect of OXT administration on social impairments and repetitive behaviors in ASD. ...
... In addition, following the one-stage approach (22), the doseresponse meta-analyses were conducted considering the correlation between the ranged OXT dosage per day, treatment duration, and a set of mean differences of core symptoms. SMD and its 95% CI for each 1 IU/day increase in OXT administration in each RCT were calculated with the dose-response meta-analysis using the method introduced by Crippa and Orsini (2016) (22). This method requires the number of participants in each research arm, the administration dose, and the mean and standard deviation of change across the study in each research. ...
Article
Full-text available
Introduction Social impairments and repetitive behaviors are at the core symptoms of autism spectrum disorder (ASD). Intranasal administration of the neuropeptide oxytocin (OXT) is a promising treatment. However, there have been inconsistencies in the effects of OXT on social impairments and repetitive behaviors. Methods A comprehensive search in PubMed, the Cochrane Library, Embase, and Web of Science was conducted to gather randomized controlled trials (RCTs) on the efficacy of OXT in patients diagnosed with ASD up to 11/06/2024. The core outcomes were social impairments measured by total Social Responsiveness Scale (SRS) scores and repetitive behaviors measured by the Repetitive Behavior Scale (RBS). Results This meta-analysis ultimately included 12 RCTs with 498 ASD patients. In an initial analysis, intranasal OXT showed no significant effect on social impairments. For a high dose of 48 IU per day, a beneficial effect on social impairments was found. According to the dose–response meta-analysis, the results indicated that higher doses of OXT might be more effective for social impairments. Depending on repetitive behaviors, the overall analysis showed no significant effect, while the dose over 48 IU per day revealed significant results and the dose–response meta-analysis suggested that higher doses could be more effective for repetitive behaviors. Discussion Although these findings show no consistent beneficial effects, the results of the dose–response meta-analysis suggest that high doses of intranasal OXT per day may be more effective in ASD. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42024567213.
... 61 We also performed a single-stage weighted mixed-effects metaanalysis to clarify the impact of different HS doses of HS in grams on SBP and DBP. 62 Nonlinear dose-response relationships were assessed utilizing restricted cubic splines with 3 knots at Harrell's suggested centiles (10 %, 50 %, and 90 %). The adequacy of the nonlinear model was ascertained by the significance of the Wald test. ...
... The adequacy of the nonlinear model was ascertained by the significance of the Wald test. 62,63 All statistical analyses were carried out using version 17.0 of StataCorp's statistical software. The statistical significance threshold was set at P < 0.05. ...
Article
Full-text available
Background: Conventional treatments for cardiometabolic diseases face limitations related to cost, efficacy, and side effects. Hibiscus sabdariffa (HS) is a common food product and a potential alternative. However, previous studies have shown inconsistent results and lacked assessments of result certainty, intervention safety, and subgroup analysis credibility. This study evaluated the efficacy and safety of HS on blood pressure (BP), lipid profiles, glycemic control, anthropometric parameters, inflammatory markers, oxidative stress indicators, and liver enzymes. Methods: To conduct this umbrella review, a systematic search of eligible meta-analyses was performed up to May 2024. The random-effects model was used to synthesize results from individual trials. Quality, certainty, and credibility of evidence were evaluated using the Cochrane Risk of Bias tool, AMSTAR-II, GRADE, and ICEMAN frameworks. Results: Data from 26 randomized controlled trials involving 1,797 participants revealed that HS dose-dependently reduced systolic and diastolic BP compared to placebo and other teas. Although no significant differences were found between HS and antihypertensive drugs, HS showed moderate credibility for therapeutic BP reduction (>10mmHg), especially in individuals over 50 years, in trials lasting over four weeks, and in those with a low risk of bias. HS also reduced total cholesterol, LDL-C, fasting blood glucose, and increased HDL-C. A minor, clinically insignificant increase in aspartate aminotransferase was observed without elevating adverse event risks. Conclusions: HS showed BP-lowering effects comparable to antihypertensive drugs and beneficial impacts on lipid and glycemic profiles. Although HS is generally considered safe, long-term and therapeutic dosing safety requires careful monitoring.
... A p-value greater than 0.05 suggests a linear relationship. Tis approach aids in understanding the complex relationship between dose and response and assessing the statistical signifcance of these relationships in meta-analysis [23]. ...
Article
Full-text available
Background: This systematic review aimed to assess the association of iron level with gestational diabetes mellitus (GDM) risk. Methods: The relevant articles published between January 1, 1995 and January 17, 2023 were identified through a systematic literature search. This study used random effects to summarize the relative risks (RRs) 95% confidence intervals (CIs) of GDM risk and standardized mean differences. This study investigated the association of ferritin exposure with GDM combined with dose–response analysis and explored both linear and nonlinear trends. Results: This meta-analysis selected 30 studies with serum ferritin (SF), 18 studies with serum iron (SI), 4 studies with serum transferrin receptor (sTfR), 5 studies with total iron binding capacity (TIBC), and 4 studies with transferrin (TRF). The summarized RRs comparing persons with the highest concentration categories of SF with the lowest concentration categories of SF with an unadjusted odds ratio were 2.05 (1.67–2.53; I2 = 62.8%, p<0.001, z = 6.76, p<0.001) and with an adjusted odds ratio were 1.82 (1.54–2.14; I2 = 12.9%, p=0.312, z = 7.21, p<0.001). Linear dose–response showed that an increase in SF of 5 μg/L increased the risk of GDM by 2.66% (1.026 [95% CI: 1.017, 1.036], n = 5). The nonlinear dose–response relationship also indicates that the increased SF is consistently associated with an increasing risk of GDM. Conclusion: High ferritin, high iron levels, and low TIBC are associated with an increased risk of GDM.
... To evaluate the dose-response relationship between BFRT dose and the clinical variables (knee extension strength and TUG), previously published methods were used [39]. A one-stage dose-response meta-analysis of mean diferences proposed by Crippa and Orsini was performed [40,41]. Te method consisted of dose-response models estimated by pooling study-specifc dose-response coefcients. ...
Article
Full-text available
Background: Previous meta‐analyses show contrasting findings regarding the effects of blood flow restriction training (BFRT) in different knee conditions. Furthermore, no previous dose‐response analysis has been conducted to determine the dose of BFRT required for maximal strength and functionality adaptations. Objective: To analyze the evidence on the effects of BFRT on strength and functionality in patients with knee osteoarthritis or rheumatoid arthritis through a systematic review with dose‐response meta‐analysis. Methods: Included studies met the following criteria: participants with knee osteoarthritis or rheumatoid arthritis; low‐load resistance BFRT as intervention; control group with traditional moderate or high intensity resistance training (MIRT and HIRT); include muscle strength and functionality as primary and secondary outcome measures, respectively; and only randomized controlled trials. A random‐effects and a dose‐response model estimated strength and functionality using estimates of the total repetitions performed. Results: We included five studies with a sample of 205 participants. No statistically significant differences were found between BFRT and MIRT or HIRT for strength (SMD = −0.06; 95% CI = −0.78–0.67; and p > 0.05) and functionality (SMD = 0.07; 95% CI = −0.23–0.37; and p > 0.05). We found an inverted U‐shaped association between the increase in total repetitions and strength gain and between the increase in total repetitions and functional improvement. Conclusions: People with knee osteoarthritis or rheumatoid arthritis can use low‐load BFRT for strength and functionality as a similarly effective alternative to MIRT and HIRT. A total of 2000 repetitions per BFRT program are necessary to maximize strength gains in these patients, while functional improvement requires 1800 total repetitions.
... mg of fluoxetine equivalents). This limitation is particularly relevant when data are sparse at higher or lower doses [2,9,36]. ...
Article
Full-text available
Pediatric major depressive disorder (MDD) often leads to recurrent depression in adulthood. The efficacy, safety and dose dependency of pharmacological effect is unclear. We conducted a systematic review and dose-response meta-analysis comprising of 22 double-blind randomized controlled trials, the majority of which had short trial durations ranging from 6 to 12 weeks. Studies were identified from PubMed, Ovid Embase, Ovid Medline, Ovid PsycInfo, Wanfang, ClinicalTrial.gov and CENTRAL until July 31, 2023. Doses of all antidepressants were converted to fluoxetine equivalents. Outcomes including treatment response, remission, suicidality, tolerability and acceptability were assessed. Sensitivity analysis, funnel plot and the trim-and fill method are used to assess and adjust for publication bias. Findings revealed that antidepressants were marginally more effective than placebos in terms of treatment response, but significantly increased the risk of adverse effects. No significant differences were observed in remission, suicidality, or overall dropout rates. Dose-response analysis indicated a relatively flat increase in response probability with higher fluoxetine equivalent doses, but also a sharp increase risk of discontinuation due to side effects. This study suggests that antidepressants for pediatric MDD may be less effective in adults, emphasizing the need to balance treatment benefits with potential adverse effects when considering interventions for this population.
Article
Full-text available
Background Hypertension and vascular dysfunction are major health concerns, and studies have suggested different interventions, including dietary nitrate (NO3), to improve it. We sought to elucidate the effects of dietary NO3 on plasma NO3 and nitrite (NO2) levels and to determine the shape of the effect of dietary NO3 on blood pressure (BP) and vascular health biomarkers. Methods PubMed, Scopus, and Web of Science were searched up to February 2024 for eligible randomized controlled trials (RCTs). The pooled results were reported as weighted mean differences (WMD) and 95% confidence intervals (CIs). Results Our analysis of 75 RCTs involving 1823 participants revealed that per each millimole (mmol) increase in the administered NO3 dose, both acute (WMD: 32.7µmol/L; 95%CI: 26.1, 39.4) and chronic-term (WMD: 19.6µmol/L; 95%CI: 9.95, 29.3) plasma NO3 levels increased. Per each mmol increase in NO3 intake, a reduction in systolic BP levels was observed in the acute (WMD: -0.28mmHg; 95%CI: -0.40, -0.17), short-term (WMD: -0.24mmHg; 95%CI: -0.40, -0.07), and medium-term (WMD: -0.48mmHg; 95%CI: -0.71, -0.25) periods. Furthermore, a decrease in diastolic BP for each mmol increase in NO3 intake (WMD: -0.12 mmHg; 95% CI: -0.21, -0.03) was shown. Moreover, a linear dose-response relationship was indicated between each mmol of NO3 intake and medium-term pulse wave velocity (WMD: -0.07 m/s; 95%CI: -0.11, -0.03), medium-term flow-mediated dilation (WMD: 0.30%; 95%CI: 0.15, 0.46), and medium-term augmentation index (WMD: -0.57%; 95%CI: -0.98, -0.15). Conclusion We observed dose-dependent increases in plasma NO3 and NO2 levels, along with consequent reductions in BP and enhancements in vascular health following dietary NO3 supplementation. Future high-quality, population-specific studies with optimized dietary NO3 dosages are needed to strengthen the certainty of the evidence. Registration The protocol for this systematic review was registered in PROSPERO under the registration number CRD42024535335.
Article
Importance The association of digital screen time with myopia has been documented, but the dose-response association and safe exposure threshold remain unclear. Objective To evaluate the dose-response association of time spent on digital screens with myopia risk. Data Sources PubMed, EMBASE, Cochrane Library databases, CINAHL, and ClinicalTrials.gov were searched for full-length articles from peer-reviewed journals without restrictions on study design, publication date, or language from inception to November 25, 2024. Study Selection Primary research articles investigating the association of exposure to digital screen devices (ie, smartphones, tablets, game consoles, computers, or television) with myopia-related outcomes (ie, prevalent or incident myopia and the rate of myopia progression) were identified by reviewers. Data Extraction and Synthesis Two independent reviewers extracted data using a standardized procedure in accordance with the Meta-Analysis of Observational Studies in Epidemiology (MOOSE) and Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) reporting guidelines. A random-effects, dose-response meta-analysis (DRMA) was utilized to examine the pattern of the association of screen time with myopia. Main Outcome and Measures Increased odds of myopia per hour of daily screen time. Results In the linear DRMA of 45 studies with 335 524 participants (mean [SD] age, 9.3 [4.3] years), an additional hour of daily screen time was associated with higher odds of myopia (odds ratio [OR], 1.21; 95% CI, 1.13-1.30). The nonlinear DRMA of 34 studies with 314 910 participants also indicated higher odds of myopia with increasing screen time, ranging from 1 hour of daily exposure (OR, 1.05; 95% CI, 1.01-1.09) to 4 hours (OR, 1.97; 95% CI, 1.56-2.40). The dose-response curve showed myopia risk increasing significantly between 1 to 4 hours of daily screen time, and then rising more gradually after 4 hours. Conclusions and Relevance In this systematic review and DRMA, a daily 1-hour increment in digital screen time was associated with 21% higher odds of myopia and the dose-response pattern exhibited a sigmoidal shape, indicating a potential safety threshold of less than 1 hour per day of exposure, with an increase in odds up to 4 hours. These findings can offer guidance to clinicians and researchers regarding myopia risk.
Article
Objective Serum uric acid (SUA) may play positive roles in diseases associated with oxidative stress, such as osteoporosis (OP). Nevertheless, the specific impact of SUA levels on both bone mineral density (BMD) and the risk of OP remains uncertain. Considering such information crucial for clinicians when making decisions about urate‐lowering therapy (ULT), we sought to fill this gap by conducting dose–response meta‐analyses. Methods PubMed, EMBASE, and Cochrane Library were searched for studies that met the inclusion criteria. Pooled standardized mean difference (SMD) for BMDs and the odds ratio (OR) for OP between the highest and lowest SUA categories as well as the nonlinear dose–response relationships were estimated. Results Pooled SMDs indicate that participants in the highest category of SUA have greater BMDs at the lumbar spine (SMD = 0.37; 95% CI: 0.27, 0.46), femoral neck (SMD = 0.25; 95% CI: 0.21, 0.29), total hip (SMD = 0.34; 95% CI: 0.26, 0.42), and lower risk of OP (OR = 0.59, 95% CI: 0.52, 0.67) compared with the lowest. The nonlinear dose–response relationships were also observed. However, when the SUA level exceeded 6 mg/dL, the dose–response curve between SUA levels and the risk of OP tended to be flattened. Conclusion Nonlinear dose–response relationships were found that higher SUA levels are associated with greater BMDs and lower risk of OP. For patients receiving ULT, maintaining SUA level at around 6 mg/dL may be appropriate from the perspective of bone metabolism.
Article
Full-text available
Objective: To develop an evidence base for recommendations on the use of atypical antipsychotics for patients with schizophrenia. Design: Systematic overview and meta-regression analyses of randomised controlled trials, as a basis for formal development of guidelines. Subjects: 12 649 patients in 52 randomised trials comparing atypical antipsychotics (amisulpride, clozapine, olanzapine, quetiapine, risperidone, and sertindole) with conventional antipsychotics (usually haloperidol or chlorpromazine) or alternative atypical antipsychotics. Main outcome measures: Overall symptom scores. Rate of drop out (as a proxy for tolerability) and of side effects, notably extrapyramidal side effects. Results: For both symptom reduction and drop out, there was substantial heterogeneity between the results of trials, including those evaluating the same atypical antipsychotic and comparator drugs. Meta-regression suggested that dose of conventional antipsychotic explained the heterogeneity. When the dose was </=12 mg/day of haloperidol (or equivalent), atypical antipsychotics had no benefits in terms of efficacy or overall tolerability, but they still caused fewer extrapyramidal side effects. Conclusions: There is no clear evidence that atypical antipsychotics are more effective or are better tolerated than conventional antipsychotics. Conventional antipsychotics should usually be used in the initial treatment of an episode of schizophrenia unless the patient has previously not responded to these drugs or has unacceptable extrapyramidal side effects.
Article
Full-text available
An increasing number of quantitative reviews of epidemiological data includes a doseresponse analysis. Aims of this paper are to describe the main aspects of the methodology and to illustrate the novel R package dosresmeta developed for multivariate dose-response meta-analysis of summarized data. Specific topics covered are reconstructing covariances of correlated outcomes; pooling of study-specific trends; flexible modeling of the exposure; testing hypothesis; assessing statistical heterogeneity; and presenting in either a graphical or tabular way the overall dose-response association.
Article
Full-text available
It is a major goal of clinical pharmacology to understand the dose-effect relationship in therapeutics. Much progress towards this goal has been made in the last 2 decades through the development of pharmacokinetics as a discipline. The study of pharmacokinetics seeks to explain the time course of drug concentration in the body. Recognition of the crucial concepts of clearance and volume of distribution has provided an important link to the physiological determinants of drug disposition. Mathematical models of absorption, distribution, metabolism and elimination have been extensively applied, and generally their predictions agree remarkably well with actual observations. However, the time course of drug concentration cannot in itself predict the time course or magnitude of drug effect. When drug concentrations at the effect site have reached equilibrium and the response is constant, the concentration-effect relationship is known as pharmacodynamics. Mathematical models of pharmacodynamics have been used widely by pharmacologists to describe drug effects on isolated tissues. The crucial concepts of pharmacodynamics are potency — reflecting the sensitivity of the organ or tissue to a drug, and efficacy — describing the maximum response. These concepts have been embodied in a simple mathematical expression, the Emax model, which provides a practical tool for predicting drug response analogous to the compartmental model in pharmacokinetics for predicting drug concentration. The application of pharmacodynamics to the study of drug action in vivo requires the linking of pharmacokinetics and pharmacodynamics to predict firstly the dose-concentration, and then the concentration-effect relationship. This may be done directly by equating the concentration predicted by a pharmacokinetic model to the effect site concentration, but this simplistic approach is often not appropriate for various reasons, including delay in drug equilibrium with the receptor site, use of indirect measures of drug action, the presence of active metabolites, or homeostatic responses, thus often necessitating the use of more complex models. The relative pharmacodynamic bioavailability of different preparations of the same drug may be determined from the time course of a drug effect. Bioavailability determined in this way may differ markedly from bioavailability defined by measurements of drug concentration if active metabolites are formed or if effects are produced in the non-linear region of the concentration-effect relationship. The influence of changes in the extent of plasma protein binding may be important in the interpretation of drug concentration measurements since it is generally held that only the unbound fraction is pharmacologically active. Clear examples of this phenomenon are few, but this reflects the general paucity of adequate observations rather than casting doubt on the usual assumption. The design of rational dosing regimens for clinical therapeutics cannot be performed with a knowledge of pharmacokinelics alone. The time course of drug effect may be essentially independent of concentration when a dose produces near maximal effects throughout the dosing interval. If effects are between 20 and 80% of maximum, the response will decrease linearly even though concentrations are declining exponentially. Finally, at relatively small degrees of effect, the time course of drug effect and concentration will be in parallel. The usual ‘rule of thumb’ of dosing every half-life is a conservative strategy for limiting wide fluctuations in drug effect, but demands more from the patient in terms of dosing frequency than may be necessary to achieve consistent drug action. On the other hand, if therapeutic success is dependent more on cumulative response than moment to moment activity, the use of extended dosing intervals may markedly reduce the effectiveness of the same average dose. Considerations of these factors can be incorporated into a dosing scheme by combined application of the principles of pharmacokinelics and pharmacodynamics.
Article
Low‐dimensional parametric models are well understood, straightforward to communicate to other workers, have very smooth curves and may easily be checked for consistency with background scientific knowledge or understanding. They should therefore be ideal tools with which to represent smooth relationships between a continuous predictor and an outcome variable in medicine and epidemiology. Unfortunately, a seriously restricted set of such models is used routinely in practical data analysis – typically, linear, quadratic or occasionally cubic polynomials, or sometimes a power or logarithmic transformation of a covariate. Since their flexibility is limited, it is not surprising that the fit of such models is often poor. Royston and Altman's recent work on fractional polynomials has extended the range of available functions. It is clearly crucial that the chosen final model fits the data well. Achieving a good fit with minimal restriction on the functional form has been the motivation behind the major recent research effort on non‐parametric curve‐fitting techniques. Here I propose that one such model, a (possibly over‐fitted) cubic smoothing spline, may be used to define a suitable reference curve against which the fit of a parametric model may be checked. I suggest a significance test for the purpose and examine its type I error and power in a small simulation study. Several families of parametric models, including some with sigmoid curves, are considered. Their suitability in fitting regression relationships found in several real data sets is investigated. With all the example data sets, a simple parametric model can be found which fits the data approximately as well as a cubic smoothing spline, but without the latter's tendency towards artefacts in the fitted curve. Copyright © 2000 John Wiley & Sons, Ltd.
Article
This paper presents a command, glst, for trend estimation across different exposure levels for either single or multiple summarized case-control, incidence-rate, and cumulative incidence data. This approach is based on constructing an approximate covariance estimate for the log relative risks and estimating a corrected linear trend using generalized least squares. For trend analysis of multiple studies, glst can estimate fixed- and random-effects metaregression models. Copyright 2006 by StataCorp LP.
Article
This article reports the results of a meta-analysis based on dose–response studies conducted by a large pharmaceutical company between 1998–2009. Data collection targeted efficacy endpoints from all compounds with evidence of clinical efficacy during the time period. Safety data were not extracted. The goal of the meta-analysis was to identify consistent quantitative patterns in dose–response across different compounds and diseases. The article presents summaries of the study designs, including the number of studies conducted for each compound, dosing range, the number of doses evaluated, and the number of patients per dose. The Emax model, ubiquitous in pharmacology research, was fit for each compound. It described the data well, except for a single compound, which had nonmonotone dose–response. Compound-specific estimates and Bayesian hierarchical modeling showed that dose–response curves for most compounds can be approximated by Emax models with “Hill” parameters close to 1.0. Summaries of the potency estimates show pharmacometric predictions of potency made before the first dose ranging study within a (1/10, 10) multiple of the final estimates for 90% of compounds. The results of the meta-analysis, when combined with compound-specific information, provide an empirical basis for designing and analyzing new dose finding studies using parametric Emax models and Bayesian estimation with empirically derived prior distributions.
Article
Dose-finding experiments define the safe dosage of a drug in development, in terms of the quantity given to a patient. Statistical methods play a crucial role in identifying optimal dosage. Used appropriately, these methods provide reliable results and reduce trial duration and costs. In practice, however, dose-finding is often done poorly, with widely used conventional methods frequently being unreliable, leading to inaccurate results. However, there have been many advances in recent years, with new statistical techniques being developed and it is important that these new techniques are utilized correctly. Statistical Methods for Dose-Finding Experiments reviews the main statistical approaches for dose-finding in phase I/II clinical trials and presents practical guidance on their correct use. Includes an introductory section, summarizing the essential concepts in dose-finding. Contains a section on algorithm-based approaches, such as the traditional 3+3 design, and a section on model-based approaches, such as the continual reassessment method. Explains fundamental issues, such as how to stop trials early and how to cope with delayed or ordinal outcomes. Discusses in detail the main websites and software used to implement the methods. Features numerous worked examples making use of real data. Statistical Methods for Dose-Finding Experiments is an important collaboration from the leading experts in the area. Primarily aimed at statisticians and clinicians working in clinical trials and medical research, there is also much to benefit graduate students of biostatistics.
Book
If you have ever wondered when visiting the pharmacy how the dosage of your prescription is determined this book will answer your questions. Dosing information on drug labels is based on discussion between the pharmaceutical manufacturer and the drug regulatory agency, and the label is a summary of results obtained from many scientific experiments. The book introduces the drug development process, the design and the analysis of clinical trials. Many of the discussions are based on applications of statistical methods in the design and analysis of dose response studies. Important procedural steps from a pharmaceutical industry perspective are also examined.
Article
The abstract for this document is available on CSA Illumina.To view the Abstract, click the Abstract button above the document title.