ArticlePDF Available

Second-generation antipsychotic effect on cognition in patients with schizophrenia—a meta-analysis of randomized clinical trials

Authors:
  • Psychiatry - Aalborg University Hospital

Abstract and Figures

Objective To investigate the effect of second-generation antipsychotics on cognitive function in patients diagnosed with schizophrenia or schizoaffective disorder.Method Multiple-treatments meta-analysis model.ResultsOn cognitive composite score, sertindole was superior to clozapine, effect size (ES) 0.87; 95% CI: 0.12–1.63, quetiapine, ES 0.75; 95% CI: 0.00–1.49, and first-generation antipsychotics (FGAs), ES 0.89; 95% CI: 0.14–1.64. Analyses on each cognitive domain showed clozapine, ES 0.37; 95% CI: 0.00–0.74, olanzapine, ES 0.31; 95%CI: 0.02–0.59, quetiapine, ES 0.34; 95% CI: 0.03–0.64, and FGAs, ES 0.51; 95% CI: 0.18–0.83 performing poorer on verbal working memory than ziprasidone, as well as FGAs performing poorer than risperidone, ES 0.31; 95% CI: 0.04–0.58. On executive function, sertindole performed better than clozapine, ES 0.82; 95% CI: 0.06–1.58, olanzapine, ES 0.81; 95% CI: 0.07–1.55, quetiapine, ES 0.76; 95% CI: 0.02–1.51, ziprasidone, ES 0.90; 95% CI: 0.14–1.67, and FGAs, ES 0.83; 95% CI: 0.08–1.58. On processing speed, FGAs performed poorer than sertindole, ES 0.97; 95% CI: 0.02–1.91, and quetiapine, ES 0.36; 95% CI: 0.01–0.72. On long-term verbal working memory, clozapine performed poorer than olanzapine, ES 0.41; 95% CI: 0.06–0.76. On verbal fluency, FGAs performed poorer than olanzapine, ES 0.26; 95% CI: 0.01–0.50, and clozapine, ES 0.44; 95% CI: 0.06–0.81. Lastly, FGAs, ES 0.41; 95% CI: 0.04–0.78, and clozapine, ES 0.44; 95% CI: 0.05–0.83, performed poorer on visuospatial skill compared to olanzapine.Conclusion The meta-analysis was able to detect some trends in the data analyzed, but did not show any drug having a uniform positive cognitive profile.
Content may be subject to copyright.
Meta-analysis
Second-generation antipsychotic effect on
cognition in patients with schizophrenia
a meta-analysis of randomized clinical trials
Nielsen RE, Levander S, Kjaersdam Tell
eus G, Jensen SOW,
Østergaard Christensen T, Leucht S. Second-generation antipsychotic
effect on cognition in patients with schizophreniaa meta-analysis of
randomized clinical trials.
Objective: To investigate the effect of second-generation antipsychotics
on cognitive function in patients diagnosed with schizophrenia or
schizoaffective disorder.
Method: Multiple-treatments meta-analysis model.
Results: On cognitive composite score, sertindole was superior to
clozapine, effect size (ES) 0.87; 95% CI: 0.121.63, quetiapine, ES 0.75;
95% CI: 0.001.49, and first-generation antipsychotics (FGAs), ES
0.89; 95% CI: 0.141.64. Analyses on each cognitive domain showed
clozapine, ES 0.37; 95% CI: 0.000.74, olanzapine, ES 0.31; 95%CI:
0.020.59, quetiapine, ES 0.34; 95% CI: 0.030.64, and FGAs, ES 0.51;
95% CI: 0.180.83 performing poorer on verbal working memory than
ziprasidone, as well as FGAs performing poorer than risperidone, ES
0.31; 95% CI: 0.040.58. On executive function, sertindole performed
better than clozapine, ES 0.82; 95% CI: 0.061.58, olanzapine, ES 0.81;
95% CI: 0.071.55, quetiapine, ES 0.76; 95% CI: 0.021.51,
ziprasidone, ES 0.90; 95% CI: 0.141.67, and FGAs, ES 0.83; 95% CI:
0.081.58. On processing speed, FGAs performed poorer than
sertindole, ES 0.97; 95% CI: 0.021.91, and quetiapine, ES 0.36; 95%
CI: 0.010.72. On long-term verbal working memory, clozapine
performed poorer than olanzapine, ES 0.41; 95% CI: 0.060.76. On
verbal fluency, FGAs performed poorer than olanzapine, ES 0.26; 95%
CI: 0.010.50, and clozapine, ES 0.44; 95% CI: 0.060.81. Lastly,
FGAs, ES 0.41; 95% CI: 0.040.78, and clozapine, ES 0.44; 95% CI:
0.050.83, performed poorer on visuospatial skill compared to
olanzapine.
Conclusion: The meta-analysis was able to detect some trends in the
data analyzed, but did not show any drug having a uniform positive
cognitive profile.
R. E. Nielsen
1
, S. Levander
2
,
G. Kjaersdam Tell
eus
1
,S.O.W.
Jensen
1
, T. Østergaard
Christensen
3
, S. Leucht
4
1
Aalborg University Hospital, Psychiatry, Aalborg,
Denmark,
2
Department of Criminology, Malmo
University, Malmo, Sweden,
3
Public Health and Quality
Improvement, Aarhus, Denmark and
4
Department of
Psychiatry and Psychotherapy, Technische Universit
at
M
unchen, Munich, Germany
Key words: schizophrenia; meta-analysis; antipsychotics;
cognition
Ren
e Ernst Nielsen, Unit for Psychiatric Research,
Aalborg University Hospital, Psychiatry, Mølleparkvej 10,
9000 Aalborg, Denmark. E-mail: ren@rn.dk
Accepted for publication November 7, 2014
Summations
The meta-analysis showed decreased verbal working memory in patients treated with clozapine, ola-
nzapine, quetiapine, and FGAs compared to ziprasidone.
Sertindole had a positive effect on executive function compared to clozapine, olanzapine, ziprasidone,
and FGAs.
Clozapine and olanzapine had positive effects on verbal fluency.
1
Acta Psychiatr Scand 2015: 1–12 ©2015 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
All rights reserved
DOI: 10.1111/acps.12374 ACTA PSYCHIATRICA SCANDINAVICA
Considerations
The results regarding sertindole were only based on a single direct comparison and all of the indirect
comparisons with haloperidol.
No conclusions can be drawn regarding the specific effects of drug dosage in the individual patients.
Introduction
Cognitive deficits are a core feature of schizophre-
nia (1, 2). Several studies have shown deficits of
one to more than three standard deviations com-
pared to healthy controls (2). The deficits are sta-
ble over time and modestly correlated to negative
symptoms, with more severe negative symptoms
being correlated to more severe cognitive deficits;
and positive symptoms only being correlated to
cognitive deficits to a minor degree (35). Neuro-
cognitive deficits are linked to family disposition
and predict longitudinal functional outcome (68).
Data on the general effect of first-generation anti-
psychotics (FGAs) on cognition in patients with
schizophrenia are sparse, as most studies have
used haloperidol as the comparator. FGAs are
generally associated with deficits in working mem-
ory, processing speed, and motor skills, perhaps
due to a higher affinity for the dopamine D2
receptor or because anticholinergics are often used
in combination with these drugs (9). A high affin-
ity for the cholinergic and the histaminergic recep-
tors is also seen in low-potency FGAs (10), with
antagonistic cholinergic effects being correlated
with decreases in attention, memory, and execu-
tive functions (9, 11). Second-generation antipsy-
chotics (SGAs) receptor profiles are more diverse,
and the group is more heterogeneous than FGAs
(12). Early, smaller, clinical studies suggested that
SGAs could reduce cognitive impairment in
patients with schizophrenia, but newer, larger,
clinical studies have only shown modest to negligi-
ble effects in both chronic and first-episode
patients (9, 13, 14).
Current studies have not shown a uniform pat-
tern of cognitive effects of antipsychotic drug, for
patients diagnosed with schizophrenia. Several
meta-analyses, all but one, conducted as simple
pairwise meta-analyses, have tried to investigate
the effects of antipsychotic drugs on cognition in
patients diagnosed with schizophrenia (1518).
One meta-analysis has used multiple-treatment
meta-analysis methodology, but has chosen only
to include studies with a duration of more than
6 months (19). Several large studies have been con-
ducted and results published as the latest large
meta-analysis on this topic was conducted and
published (15), making more data available for the
current analysis. In the recent years, multiple-treat-
ment meta-analysis (MTMA), also termed a net-
work meta-analysis, have increasingly been used to
investigate the effects of psychotropic drugs (20,
21), as the statistical model allows for investigation
of not only all directly compared drugdrug com-
binations, for example, head-to-head studies, but
also to investigate the effects of all indirect drug
drug comparisons; for example, if drug A vs. B
and drug B vs. drug C have been directly com-
pared, this model can investigate the effects of drug
A vs. drug C, even when no head-to-head study
has been conducted (22).
Aims of the study
In this meta-analysis, we aim to evaluate available
data on second-generation antipsychotic drugs
published between January 1, 1980 and April 15,
2013.
Material and methods
Search strategy
We searched for relevant primary studies and/or
secondary publications (systematic reviews) in
MEDLINE, EMBASE, PsycINFO, and the Coch-
rane Central Register of Controlled Trials (CEN-
TRAL) using the keywords schizophrenia or
schizophrenic AND cognition or cognitive or neu-
rocognition or neurocognitive AND amisulpride
or aripiprazole or asenapine or blonanserin or
clozapine or iloperidone or lurasidone or mosapr-
amine or olanzapine or paliperidone or perospi-
rone or quetiapine or remoxipride or risperidone
or sertindole or ziprasidone or zotepine. In addi-
tion, the Cochrane Database of Systematic
Reviews (CDSR), the Database of Abstracts of
Reviews of Effects (DARE), and the Cochrane
Health Technology Assessment (HTA) Database
were searched for relevant secondary publications.
A handsearch of primary and secondary publica-
tion reference lists was conducted to identify fur-
ther studies.
2
Nielsen et al.
Only randomized controlled trials on humans,
diagnosed with schizophrenia or schizoaffective
disorders, published between January 1, 1980 and
April 15, 2013 were included. Studies with a dura-
tion of <8 weeks were excluded, as well as studies
on depot formulations of antipsychotics. The 8-
week minimum duration was chosen to minimize
the risk of carryover effects from previous treat-
ment and to allow for cognitive effects of the inves-
tigated drug to occur. Longer study durations have
been proposed by the MATRICS group (23). Par-
ticipants in included studies should be between the
ages of 18 and 65 years. Only head-to-head studies
of a SGA vs. another antipsychotic drug or pla-
cebo were included. Studies on combinations of
antipsychotic drugs vs. a single antipsychotic drug
or placebo were analyzed separately from the oth-
ers studies. Conference abstracts were not
included. Only studies with a neurocognitive test
were included in the meta-analysis.
Meta-analysis
Drugs were divided into FGAs or SGAs. All SGAs
were included in the meta-analysis as individual
drugs, whereas FGAs were grouped. A MTMA
was conducted with individual neurocognitive tests
being categorized as mainly measuring 1) atten-
tion, 2) executive function, 3) long-term non-verbal
memory, 4) long-term verbal memory, 5) motor
function, 6) non-verbal working memory, 7) verbal
working memory, 8) processing speed, 9) reason-
ing, 10) verbal fluency, 11) verbal IQ, 12) vigilance,
13) visuospatial skill, or none of the domains
listed. A composite score was calculated from the
listed cognitive domains, with each domain
weighted equally. The allocation of neurocognitive
tests to domains was conducted by authors GKT,
SL, and TOC, in a blinded fashion initially. Dis-
crepancies were discussed openly, in line with the
domain allocations specified in Lezak et al. (24).
The cognitive domain preferred by the majority of
the authors listed was chosen, if consensus could
not be reached. We preferred this approach, in
contrast to the MATRICS suggestions (25), for
two reasons: many studies were performed before
2004, and the Lezak approach is more general
being based on all kinds of tests and data from the
1930s and forward.
The chosen measure of effect was Cohen’s d
using the pooled standard deviation as a denomi-
nator. Effect sizes and the corresponding standard
errors were calculated for all the included studies.
We furthermore adjusted the effect size due to Co-
hen’s d being upwardly biased when based on
small samples.
To ensure a unidirectional interpretation of the
effect sizes, we recoded those measures where an
increasing score equaled worsening such that a
positive value of Cohen’s d indicated improve-
ment.
When studies reported multiple measures of the
same cognitive domain, an average effect size was
computed to ensure that the studies were given the
same weight in the subsequent analyses.
The meta-analyses were then performed for each
of the two groups using a random effects regres-
sion model where effect size was entered as the
dependent variable and the drug categories as
explanatory variables. The standard error of effect
size was also entered into the model as the estimate
of within study variance. In addition to the
MTMA, a simple pairwise meta-analysis of all
studies was conducted, which is available upon
request from the corresponding author.
If data for the meta-analysis were not readily
available in the published studies, the correspond-
ing author was contacted to request the missing
data.
Confounder analysis
We conducted linear regression analyses to investi-
gate the association between the effect size and
publication year, sample size, study duration,
duration of illness, sponsorship of study and blind-
ing. Univariate analyses were performed for each
of the mentioned variables, and subsequently, a
multivariate analysis was performed. In these
regression analyses, we excluded two studies as
they were deemed outliers. The assumptions
underlying the linear regression were investigated
by visual inspection of residual plots. These analy-
ses are available upon request from the corre-
sponding author.
Exploration of publication bias
Publication bias might occur in a meta-analytic
review. To investigate this, we obtained funnel
plots to detect publication bias in meta-analysis
(26). We assessed the symmetry of the funnel plots
and found two studies to be outliers. As variation
from the symmetric funnel can be sign of publica-
tion bias, these studies were excluded.
Consistency of meta-analyses
We conducted both simple pairwise meta-analyses,
analyses which are available if requested from the
corresponding author, and MTMA with similar
results. One of the main assumptions behind a
3
SGAs effect on cognition
MTMA is that the analyzed network is coherent
and that the direct and indirect comparisons do
not disagree beyond chance. Inconsistency was
evaluated by calculating and examining confidence
intervals of the difference between direct and indi-
rect estimates.
The analyses were carried out in Stata 11, and
the chosen level of significance was 5%.
Results
A total of 5854 possible studies were identified
through the initial search. By screening of study
title, a large portion of studies could be excluded
due to several reasons, for example, studies not
investigating cognition as an outcome measure,
studies on other patient groups, other age groups,
short study duration, studies with non-randomized
design, studies on animals, and studies on other
drugs than antipsychotics. A total of 229 studies
were read and evaluated for the meta-analyses,
and a total of 51 studies fulfilled all criteria for
inclusion in the meta-analyses. Not all studies
reported the necessary data to calculate effects
sizes, and first authors of these studies were con-
tacted regarding missing data, resulting in further
14 studies being excluded due to lack of data, as
shown in Fig. 1.
Thirty-seven studies with 3526 patients were
included in the two meta-analyses, with three stud-
ies investigating the effects of augmenting cloza-
pine with a second antipsychotic (146 patients).
The mean duration of all studies was 23.6 weeks,
95% CI (17.729.5) weeks. The average age for
patients included in the meta-analyses was
35.4 years, 95% CI (33.137.8) years, and the
mean duration of illness was 10.6 years, 95% CI
(8.013.3) years. The included studies are listed in
bold in Table 1.
The most investigated drug in the MTMA on
non-clozapine augmentation was olanzapine
(n=970), followed by risperidone (n=894), que-
tiapine (n=514), clozapine (n=238), ziprasidone
(n=206), amisulpride (n=92), perospirone (n=
18), and sertindole (n=17). FGAs were used
as comparator drugs in 354 patients. No studies
with asenapine, blonanserin, iloperidone, lurasi-
done, mosapramine, paliperidone, perospirone,
remoxipride, and zotepine were included in the
meta-analyses.
Initial allocation of cognitive tests to cognitive
domains resulted in total agreement on 38.0% of
neurocognitive tests, remaining tests were dis-
cussed openly, and consensus was reached on all
tests allocated to cognitive domains. The alloca-
tion of cognitive tests to domains can be requested
from the corresponding author.
We administered Begg’s plot and Egger’s test to
investigate potential publication bias in our data-
set, and the funnel plot found two outliers. These
were excluded from further analyses. Egger’s test
showed no significant publication bias, P=0.272,
after excluding outliers.
Second, we conducted a confounder analysis to
investigate the effects of publication year, duration
of illness, study duration, gender, sponsorship of
study, blinding, and number of patients and found
no significant influences on outcome. The analyses
can be requested from the corresponding author.
Our analyses on antipsychotic monotherapy did
not show any significant differences on the cogni-
tive composite score when comparing all SGA
drugs to each other and to a group of FGAs,
except for sertindole being superior to clozapine
with an effect size (ES) of 0.87; 95% CI: 0.121.63,
quetiapine, ES 0.75; 95% CI: 0.001.49, and
FGAs, ES -0.89; 95% CI: -1.64(-0.14), as shown
in Table 2.
The analyses on each cognitive domain are
shown in Table 3, where we found clozapine, ES
0.37; 95% CI: 0.000.74, olanzapine, ES 0.31; 95%
CI: 0.020.59, quetiapine, ES 0.34; 95% CI: 0.03
0.64, and FGAs, ES 0.51; 95% CI: 0.180.83 per-
forming poorer on verbal working memory than
ziprasidone, as well as FGAs performing poorer
on verbal working memory as compared to risperi-
done, ES 0.31, 95% CI: 0.040.58. Examination of
executive function showed sertindole performing
better than clozapine, ES 0.82; 95% CI: 0.061.58,
olanzapine, ES 0.81; 95% CI: 0.071.55, quetia-
pine, ES 0.76, 95% CI: 0.021.51, ziprasidone, ES
0.90; 95% CI: 0.141.67, and FGAs, ES 0.83; 95%
CI: 0.081.58. On processing speed, FGAs per-
formed poorer than sertindole, ES 0.97; 95% CI:
0.021.91, and quetiapine, ES 0.36; 95% CI: 0.01
0.72. On long-term verbal working memory,
patients treated with clozapine performed poorer
than those treated with olanzapine, ES 0.41; 95%
CI: 0.060.76. On verbal fluency, FGAs performed
poorer than olanzapine, ES 0.26; 95% CI: 0.01
0.50, and clozapine, ES 0.44; 95% CI: 0.060.81.
Lastly, FGAs performed poorer on visuospatial
skill compared to olanzapine, ES 0.41; 95% CI:
0.040.78, as well as olanzapine patients perform-
ing better as compared to clozapine, ES 0.44; 95%
CI: 0.050.83. We did not find any other significant
differences between any of the investigated drugs
as shown in Table 3.
Our analyses showed no significant effect of
adjunctive treatment with SGAs to clozapine
4
Nielsen et al.
utilizing a cognitive composite score as shown in
Table 2.
The simple pairwise meta-analysis with only
direct comparisons did not show any significant
differences between any of the drugs investigated
on the composite score. On attention, motor func-
tion, reasoning, processing speed, and long-term
verbal memory, results were similar between the
simple pairwise and MTMA. The simple pairwise
meta-analysis found no differences between any of
the drugs compared directly on verbal fluency, but
the MTMA found clozapine and olanzapine to be
superior as compared to FGAs. On verbal working
memory, the simple pairwise analysis showed a
superior effect of risperidone compared to quetia-
pine, but with confidence intervals close to zero.
On the visuospatial domain, both analyses showed
a superior effect of olanzapine as compared to
clozapine, but the MTMA was the only one to find
a superior effect of olanzapine as compared to
FGAs. Finally, both analyses found olanzapine to
be superior to clozapine in long-term verbal mem-
ory, but only the simple pairwise meta-analysis
found risperidone to be superior to clozapine on
the same cognitive domain. Forest plots are avail-
able by request from the corresponding author,
and a network plot of all direct comparisons is
shown in Fig. 2. To estimate differences between
the direct comparisons in the simple pairwise
meta-analysis and the both direct and indirect
comparisons in the MTMA, we conducted an
analysis of consistency showing our data to be con-
sistent. All analyses regarding consistency are
available by request from the corresponding
author.
Discussion
The meta-analyses on composite cognitive score
did not show any general differences between
Citations retrieved from
bibliographic databases:
n= 5,854
Non-relevant citations:
n = 5,625
Excluded with reasons: n= 178
Excluded due to:
Published abstract: n= 32
Age: n= 12
Data published before: n= 6
Design: n= 99
Diagnosis: n= 5
Duration: n= 23
Non-human: n= 1
Full-text publication
retrieved for more detailed
evaluation:
n= 229
Overall number of meeting
inclusion criteria:
n= 51
Relevant articles identified
by handsearch:
n= 0
Overall number of studies
included in meta-analyses:
n= 35
Excluded due to missing data:
n= 14
Excluded due to bias analysis:
n= 2
Fig. 1. Flowchart: Study selection.
5
SGAs effect on cognition
SGAs and FGAs, except for the findings of sertin-
dole’s superior effects compared to clozapine, que-
tiapine, and FGAs, which were primarily driven by
a single smaller study (76), but due to the MTMA
model also driven by indirect comparisons from
studies with haloperidol as a comparator.
On the verbal working memory domain cloza-
pine, olanzapine, quetiapine, and FGAs did
poorer than ziprasidone, and FGAs did poorer
than risperidone. The main features shared by
clozapine, olanzapine, and quetiapine are a low
affinity for the dopaminergic receptor, as well as a
Table 1. Studies full-filling criteria to be included in the meta-analyses
Author Year Drug Duration (weeks) N
included
N
baseline
N
in meta-analysis
Abdolahian et al.* (27) 2008 Risperidone, haloperidol 8 65 65 65
Bellack et al. (28) 2004 Clozapine, risperidone 29 107 65 22
Bender et al. (29) 2006 Clozapine, olanzapine 26 54 54 31
Bilder et al. (30) 2002 Clozapine, olanzapine, risperidone, haloperidol 14 101 101 101
Boulay et al. (31) 2007 Olanzapine, haloperidol 8 27 27
Buchanan et al. (32) 1994 Clozapine, haloperidol 10 38 38 38
Davidson et al. (14) 2009 Amisulpride, quetiapine, ziprasidone, haloperidol 26 498 286 320
Fagerlund et al. (33) 2004 Risperidone, zuclopenthixol 13 31 25
Gallhofer et al. (34) 2007 Sertindole, haloperidol 12 40 34 32
Goldberg et al. (35) 2007 Olanzapine, risperidone 16 104 104
Green et al. (36) 1997 Risperidone, haloperidol 8 59 59
Green et al. (37) 2002 Risperidone, haloperidol 26 62 62
Grootens et al. (38) 2010 Olanzapine, ziprasidone 24 74 74
Gurpegui et al. (39) 2007 Olanzapine, risperidone 48 235 235 163
Harvey et al. (40) 2008 Clozapine, ziprasidone 12 130 130 100
Harvey et al. (41) 2003 Olanzapine, risperidone 8 377 377 267
Harvey et al. (42) 2006 Olanzapine, ziprasidone 26 133 72 72
Harvey et al. (43) 2006 Quetiapine, risperidone 8 673 289 289
Honer et al. (44) 2006 Clozapine + risperidone or clozapine + placebo 8 68 68 65
Jerrell et al. (45) 2008 Olanzapine, risperidone, typical antipsychotic drugs 52 108 108 108
Keefe et al. (46) 2006 Olanzapine, risperidone, haloperidol 52 414 414
Keefe et al. (47) 2007 Olanzapine, quetiapine, risperidone 52 400 400 81
Keefe et al. (13) 2007 Olanzapine, quetiapine, risperidone, ziprasidone, perphenazine 78 1460 1460 128
Kern et al. (48) 2006 Aripiprazole, olanzapine 26 169 169
Krakowski et al. (49) 2008 Clozapine, olanzapine, haloperidol 12 100 101 100
Lee et al. (50) 1994 Clozapine, typical antipsychotic drugs 52 59 47
Lee et al. (51) 1999 Clozapine, typical antipsychotic drugs 52 64 64 52
Lee et al. (52) 2007 Risperidone, haloperidol 8 20 20 20
Lindenmayer et al. (53) 2007 Olanzapine, haloperidol 12 35 35 33
Ljubin et al. (54) 2000 Olanzapine, uphenazine 22 18 18 18
McGurk et al. (55) 2005 Clozapine, risperidone 29 97 97 35
Meltzer et al. (56) 2008 Clozapine, olanzapine 26 40 40 24
Mori et al. (57) 2004 Olanzapine, perospirone, quetiapine, risperidone 12 77 77 77
Mortimer et al. (58) 2007 Amisulpride, olanzapine 26 26 26 18
Muscatello et al. (59) 2011 Aripiprazole + clozapine or placebo + clozapine 24 40 31 31
Nielsen et al. (60) 2012 Clozapine + sertindole or clozapine + placebo 12 50 50 50
Purdon et al. (61) 2000 Olanzapine, risperidone, haloperidol 54 65 65 65
Purdon et al. (62) 2001 Quetiapine, haloperidol 26 25 25 25
Remillard et al. (63) 2005 Risperidone, haloperidol 52 31 31
Remillard et al. (64) 2008 Risperidone. Haloperidol 52 28 28
Remillard et al. (65) 2010 Risperidone, haloperidol 52 26 26
Riedel et al. (66) 2007 Olanzapine, quetiapine 8 52 33 33
Riedel et al. (67) 2006 Quetiapine, risperidone 12 44 44 34
Rosenheck et al. (68) 2003 Olanzapine, haloperidol 52 309 309
Sergi et al. (69) 2007 Olanzapine, risperidone, haloperidol 8 100 77 73
Smith et al. (70) 2001 Olanzapine, haloperidol 21 45 45 29
Van Veelen et al. (71) 2010 Olanzapine, ziprasidone 8 74 74 56
Velligan et al. (72) 2002 Quetiapine, haloperidol 24 58 58 34
Velligan et al. (73) 2003 Quetiapine, typical antipsychotic drugs 26 40 40
Voruganti et al.* (74) 2007 Olanzapine, quetiapine 52 86 85 85
Wagner et al. (75) 2005 Amisulpride, olanzapine 8 52 36 36
Studies highlighted in bold are included in the first meta-analysis of the effects of atypical antipsychotic treatment. Studies highlighted with bold and underlined are included in
the second meta-analysis on the effects of using atypical antipsychotic drugs as adjunctive treatment to clozapine.
Year, year of publication.
*Studies excluded from the meta-analyses, as funnel plots found results were outliers.
6
Nielsen et al.
relatively high affinity for the cholinergic receptor
(12). The FGAs used as comparator drugs are
primarily haloperidol and perphenazine, with a
moderate to high affinity for the dopaminergic
receptor system and a low to moderate affinity for
the muscarinergic receptor system (10). Many of
the studies included allowed treatment with anti-
cholinergics in patients treated with FGAs, if EPS
occurred, thereby exposing patients to a high
degree of antagonism on the muscarinergic recep-
tors. The use of anticholinergic drugs was not
described in all studies. Studies have shown the
muscarinergic receptor system being implicated
with cognitive function (11, 13, 7779), with
antagonizing drugs worsening symptoms and
drugs with agonizing effects improving function in
attention and working memory domains (11, 80
83). By comparison, ziprasidone is characterized
by a relatively low affinity for the muscarinergic
receptor system. Finally, the superior effect of ris-
peridone to FGAs could perhaps be explained by
a greater use of anticholinergics in patients treated
with FGAs, a hypothesis we cannot investigate, as
all direct comparisons between risperidone and
FGAs do not report the use of anticholinergic
drugs.
Sertindole showed superiority compared to
clozapine, olanzapine, ziprasidone, and FGAs on
executive functions, primarily driven by the results
of a single study (76), but also indirectly by all
studies using haloperidol as a comparator drug via
the MTMA. Animal studies have shown remitting
effects of sertindole on phencyclidine (PCP)-
induced executive function deficits, as well as sug-
gested the 5-HT
6
affinity of sertindole as a possible
mechanism of action (8486), possibly explaining
our findings.
In regard to processing speed, both sertindole
and quetiapine performed better than FGAs. The
receptor profiles of sertindole and quetiapine are
not similar, as quetiapine is characterized by a low
affinity for the dopamine D2 receptor and a high
affinity for both the muscarinergic and histaminer-
gic receptors as opposed to sertindole (12). They
do share the same high affinity for the adrenergic
receptor, but this receptor affinity is also seen in
especially clozapine, which does not improve pro-
cessing speed (12).
Our finding of verbal fluency improvement in
patients treated with clozapine or olanzapine as
compared to FGAs could be suggestive of low
dopaminergic affinity being correlated to verbal
fluency, especially as quetiapine shows a trend
toward similar findings. No studies have examined
this in patients diagnosed with schizophrenia, but
low dopaminergic states have been associated with
decreased verbal fluency in patients diagnosed with
Parkinson’s disease (87).
Table 2. Effect sizes and confidence intervals for cognitive composite scores
Amis Arip
(Cloz)
-0.14 Cloz 0.07 Placebo
-0.49; 0.21 -1.58; 1.72 (Cloz)
0.02 0.16 Olan -0.25 -0.32 Risp
-0.29; 0.33 -
0.05; 0.37 -2.09; 1.59 -
1.59; 0.95 (Cloz)
0.07 0.21 0.05 Pero -0.14 -0.22 0.11 Sert
-0.66; 0.80 -0.48; 0.90 -0.62; 0. 73 -2.08; 1.79 -1.62; 1.19 -1.51; 1.72 (Cloz)
-0.01 0.13 -0.03 -
0.08 Quet
-0.34; 0.32 -0.11; 0.37 -0.21; 0.15 -0.77; 0.60
0.01 0.15 -0.01 -0.06 0.02 Risp
-0.30; 0.33 -0.06; 0.36 -0.16; 0.14 -0.74; 0.61 -
0.16; 0.21
0.74 0.87 0.72 0.66 0.75 0.72 Sert
-0.05; 1.52 0.12; 1.63 -0.02; 1.45 -0.33; 1.65 0.00; 1.49 -0 .01; 1.46
0.00 0.13 -0.02 -0.08 0.01 -0.02 -0.74 Zipr
-0.36; 0.36 -0.14; 0.41 -0.25; 0.20 -0.78; 0.62 -
0.25; 0.26 -0.25; 0.21 -1.50 ; 0.02
-0.16 -0.02 -0.18 -0.23 -0.15 -0.17 -0.89 -0.15 FGA
-0.49; 0.18 -0.26; 0.23 -0.36; 0.01 -0.92; 0.46 -
0.36; 0.07 -0.36; 0.03 -1.64; -0. 14 -0.41; 0.11
Adjunctive treatment to clozapine is shown on the right. The column headings correspond to the respective reference group. Significant values are in white font color and black fill
in.
Amis, amisulpride; Cloz, clozapine; Olan, olanzapine; Pero, perospirone; Quet, quetiapine; Risp, risperidone; Sert, sertindole; Zipr, ziprasidone; FGA, first-generation antipsychotic.
7
SGAs effect on cognition
The findings on long-term verbal memory and
visuospatial skill seem less uniform, and no obvi-
ous explanation is apparent for these findings.
Contrary to our expectations, we did not find
any significant differences on motor function
between any of the FGAs and SGAs investigated,
probably as a consequence of comedication with
anticholinergic drugs, and some studies using low
dose FGA treatment.
Newer studies suggest an association between
antipsychotic high dosage or polypharmaceutical
treatment and cognitive impairment, with
improved cognitive function after dose reduction
(88, 89). In the meta-analysis of adjunctive treat-
ment to clozapine with another antipsychotic drug,
we are unable to show any differential effect on
cognition between the groups treated with two
antipsychotic drugs compared to the groups only
treated with clozapine, but the study population is
small and highly selected due to the primary treat-
ment with clozapine. In the main meta-analysis of
non-adjunctively treated patients, most studies
have been conducted with a variable dosage para-
digm, with patients being treated with the optimum
dosage defined by effects and side-effects, making
an investigation of possible drug dose-dependent
cognitive effects impossible. Mean drug dosages
and drug ranges can available by request from the
corresponding author.
Several meta-analyses have previously been con-
ducted on the effects of FGAs and SGAs on cogni-
tion in patients diagnosed with schizophrenia (15
19). However, the current meta-analysis is the larg-
est conducted to date on the cognitive effects anti-
psychotic drugs, including 18 studies published in
2007 and onwards. Furthermore, the current meta-
analysis is the first to include all SGAs and to com-
pare them directly and indirectly to each other uti-
lizing a MTMA (22). Other meta-analyses have
used predefined cognitive domains with accompa-
nying cognitive tests, thereby excluding a portion
of the cognitive tests utilized in the studies (15, 16),
contrary to this meta-analysis where all cognitive
tests have been included and categorized under
specific predefined domains, as also done by Keefe
et al. and Mishara et al. (17, 18).
Generally, in meta-analyses, a high degree of
homogeneity is needed, as data are pooled from
several different studies into a single analysis. In
MTMA, this is even more so needed, as both direct
and indirect comparisons are conducted. In a
meta-analysis, there is a risk of the included studies
Table 3. Effect sizes and confidence intervals for cognitive domain scores
VERBAL WORKING MEMORYLONGTERMVERBALWORKINGMEMORY
ATTENTION
Amis -0.04 0.02 0.02 -0.01 0.13 0.33 -0.18
VERBAL FLUENCY
Amis
-0.48; 0.39 -0.35; 0.39 -
0.76; 0.80 -
0.39; 0.37 -0.25; 0.51 -0.09; 0.74 -0.57; 0.22
Cloz 0.06 0.06 0.03 0.17 0.37 -0.14 0.19 Cloz 0. 41 0.32 0.32 0.20 0.34
-0.25; 0.37 -
0.69; 0.81 -0.29; 0.36 -0.14; 0.49 0.00; 0.74 -0.48; 0.21 -0.46; 0.83 0.06; 0.76 -0. 10; 0.74 -0.04; 0.69 -0.31; 0.71 -0.05; 0.73
0.13 Olan 0.00 -0.03 0.11 0.31 -0.20 0.00 -0.18 Olan -0.09 -0.09 -0.21 -0.07
-0.63; 0.90 -0.71; 0.72 -0.26; 0.20 -0.11; 0.34 0.02; 0.59 -0.45; 0.06 -0.57; 0.58 -0.52; 0.16 -0. 40; 0.22 -0.32; 0.15 -0.63; 0.21 -0.35; 0.20
Pero -0.03 0.11 0.31 -0.20 0.00 -0.19 -0.01 Quet 0.00 -0.12 0.02
-0.75; 0.69 -0.61; 0.83 -0.43; 1.05 -0.93; 0.53 -0.60; 0.59 -0.55; 0.17 -0.24 ; 0.23 -0.32; 0.33 -0.60; 0. 36 -0.34; 0.38
-0.01 -0.14 Quet 0.14 0.34 -0.17 -0.07 -0.25 -0.07 -0.06 Risp -0.12 0.01
-0.79; 0.78 -0.45;0.17 -0.10; 0.39 0.03; 0.6 4 -0 .44; 0.11 -0.64; 0.51 -0.59; 0.08 -0.26; 0.12 -0.29; 0.17 -0.56; 0.31 -0.27; 0.30
0.10 -0.03 0.11 Risp 0.20 -0.31 0.04 -0.14 0.04 0.04 0.11 Zipr 0.14
-0.66; 0.86 -0.27; 0.21 -0.19; 0.41 -0.10; 0.49 -0.58; -0.04 -0.62; 0.71 -0.62; 0.33 -0.34; 0.42 -0.36; 0.44 -0.27; 0.48 -0.32 ; 0.60
0.16 0.03 0.17 0.06 Zipr -0.51 -0.25 -0.44 -0.26 -0.25 -0.19 -
0.29 FGA
-0.80; 1.13 -0.61; 0.67 -0.50; 0.83 -0.58; 0.70 -0.83; -0.18 -0.85; 0.35 -0.81; -0.06 -0.50 ; -0.01 -0.53; 0.03 -0.43; 0.06 -0.70; 0.12
-0.03 -0.16 -0.02 -0.13 -0.19 FGA
-1.00; 0.95 -0.82; 0.49 -0.70; 0.66 -0.78; 0.52 -1.07;0.69 VISUO - SPATIAL SKILL
MOTOR FUNCTION
Amis -0.26 0.18 0.03 0.01 -0.23
PROCESSING SPEED -1.02;0.50 -0.58; 0. 94 -1.07; 1.13 -0.83; 0.85 -0.99; 0.52
EXECUTIVE FUNCTION
Amis -0.24 -0.07 0.05 -0.16 0.65 -0.31 -0.32 0.48 Cloz 0.44 0.29 0.27 0.03
-0.81; 0.34 -0.56; 0.43 -0.49; 0.58 -0.67; 0.36 -0.38; 1.67 -0.86; 0.25 -0.84; 0.20 -0.25;1.21 0.05; 0.83 -0.59 ; 1.17 -0.25; 0.79 -0.34;0.40
-0.14 Cloz 0.17 0.28 0.08 0. 88 -0.07 -0.08 0.04 -0.44 Olan -0.15 -0.17 -0.41
-0.50; 0.23 -0.21; 0.54 -0.15; 0.71 -0.32; 0.48 -0.09;1.86 -0.53; 0.39 -0.50; 0.33 -0.39;0.47 -1.10; 0.22 -1.03; 0.73 -0.69 ; 0.35 -0.78; -0.04
-0.13 0.01 Olan 0.11 -
0.09 0.72 -0.24 -0.25 -0.02 -0.49 -0.06 Quet -0.02 -0.26
-0.44; 0.19 -
0.23; 0.25 -0.20; 0.43 -0.36; 0.19 -0.21; 1.65 -0.59;0.12 -0.54;0.04 -0.48; 0.45 -1.18;0.19 -0.40; 0.29 -0.97;0.93 -1.14; 0.61
-0.08 0.05 0.04 Quet -0.20 0.60 -
0.35 -0.03 -0.50 -0.07 -0.01 Risp -0.24
-0.42; 0.25 -0.21; 0.32 -
0.15; 0.24 -0.54; 0.14 -0.35; 1.55 -0.76; 0.06 -0.49; 0.44 -
1.19; 0.18 -0.41; 0.27 -0.40; 0.38 -0.75; 0.26
-0.06 0.08 0.07 0.03 Risp 0.80 -0. 15 -0.16 -0.04 -0.52 -0.08 -0.02 -0.01 Zipr
-0.38; 0.26 -0.16; 0.32 -0.09; 0.23 -0.17;0.22 -0. 13; 1.74 -0.53; 0.23 -0.48; 0.16 -0.62; 0.54 -1.28; 0.25 -0.57; 0.41 -0.55; 0.50 -0.53; 0.51
0.68 0.82 0.81 0.76 0.74 Sert -0.96 0.00 -0.47 -0.04 0.02 0.03 0.04 FGA
-0.11; 1.47 0.06; 1.58 0.07; 1.55 0.02; 1.51 -0. 00; 1.48 -1.92; 0.01 -0.45; 0.45 -1.15; 0.20 -0.36;0. 28 -0.35; 0.39 -0.34; 0.40 -0.47;0.55
-0.22 -0.09 -0.10 -0.14 -0.17 -0.90 Zipr -0.01
-0.59; 0.15 -0.39; 0.22 -0.34; 0.15 -0.41; 0.13 -0.42; 0.08 -1.67; -0.14 -0.41; 0. 38 LONG TERM NON -VERBAL
-0.15 -0.01 -0.02 -0.06 -0.09 -0.83 0.08 FGA
REASONING
Cloz 0.10 0.41 0.03 -0.12
-0.49; 0.20 -0.28; 0.26 -
0.22; 0.18 -0.30; 0.17 -
0.30; 0.12 -1.58; -0.08 -0. 20; 0.36 -0.49;0.68 -0. 26; 1.08 -0.60; 0.66 -0.73;0.48
Olan 0.32 -0.07 -0.22
-0.25; 0.89 -0.59; 0.46 -0.71; 0.27
0.20 Quet -0.38 -0.54
-0.50;0.90 -1.00 ; 0.23 -1.12; 0.05
0.10 -0.10 Ri sp -0.15
-0.53;0. 73 -0.8 5; 0.64 -0.69; 0.39
0.25 0.04 0.15 Zipr
-0.50;0.99 -0.80; 0.89 -0.64; 0.94
0.25 0.05 0.15 0.00 FGA
-0.43;0.92 -0.73; 0.83 -0.57; 0.87 -0.82; 0.82
-0.36
-0.72; -0.01
-0.97
-1.91; -0.02
The column headings correspond to the respective reference group. Significant values are in white font color and black fill in.
Amis, amisulpride; Cloz, clozapine; Olan, olanzapine; Pero, perospirone; Quet, quetiapine; Risp, risperidone; Sert, sertindole; Zipr, ziprasidone; FGA, first-generation antipsychotic.
8
Nielsen et al.
being too heterogeneous, making the interpreta-
tion of results hard or even impossible. With the
criteria used for inclusion of studies, we have tried
to diminish the heterogeneousity, without being
too restrictive, and thereby excluding too many
studies. In the current meta-analysis, the inclusion
criteria used have ensured inclusion of well-
designed studies only, for example, randomization,
diagnosis, and duration of study, thereby increas-
ing homogeneity of the studies. We have chosen to
include newly diagnosed, more chronically ill, and
even treatment-resistant patients diagnosed with
schizophrenia or schizoaffective disorder and
investigated the effects in the sensitivity analyses.
Optimally, investigation of, for example, duration
of untreated psychosis, previous treatment, and
effects of low- or high-dose treatment would have
been included in the sensitivity analyses, but most
of the included studies have not reported these
data, and we have only been able to investigate the
effects of associations between the effect size and
publication year, sample size, study duration,
duration of illness, sponsorship of study and
blinding.
The allocation of cognitive tests to cognitive
domains was carried out by three experienced clini-
cians with both clinical and research experience
with cognitive testing. The initial allocation of tests
to domains was carried out blinded and resulted in
a 38% agreement on tests to domains between the
raters, which could seem low. The reason for these
results is that cognitive tests do not measure a sin-
gle cognitive domain, but always measures several
domains, with a single domain dominating the test.
After the initial blinded allocation of cognitive
tests to domains, the remaining tests were dis-
cussed openly, and total agreement was reached by
the three raters.
The meta-analysis was able to detect some
trends in the data analyzed, suggesting a decreased
verbal working memory in patients treated with
clozapine, olanzapine, quetiapine, and FGAs com-
pared to ziprasidone and a positive effect of sertin-
dole on executive function compared to clozapine,
olanzapine, ziprasidone, and FGAs, as well as a
possible positive effect of clozapine and olanzapine
on verbal fluency.
Acknowledgements
None.
Declaration of interest
All expenses toward the authors’ salaries were paid by the
affiliated institutions. Expenses for retrieval of manuscripts
were covered by the Unit of Psychiatric Research, Aalborg
Psychiatric Hospital, Aalborg University Hospital, Aalborg,
Denmark.
Amisulpride
Clozapine
Quetiapine
Sertindole
Ziprasidone
FGA
Risperidone Olanzapine
Perospirone
Fig. 2. Network plot of direct comparisons between non-augmented studies. Thicknesses of lines symbolize number of studies on
each direct comparison, and size of node symbolizes number of participants for each drug.
9
SGAs effect on cognition
R. E. Nielsen has received research grants from H. Lund-
beck for clinical trials, received speaking fees from Bristol-
Myers Squibb, Astra Zeneca, Janssen & Cilag, Lundbeck,
Servier, Otsuka Pharmaceuticals, and Eli Lilly and has acted
as advisor to Astra Zeneca, Eli Lilly, Lundbeck, Otsuka Phar-
maceuticals, Takeda, and Medivir.
S. Leucht has received honoraria for consulting/advisory
boards from Alkermes, Bristol-Myers Squibb, Eli Lilly, Jans-
sen & Cilag, Johnson & Johnson, Lundbeck, MedAvante,
Roche; has received lecture honoraria from Astra Zeneca, Bris-
tol-Myers Squibb, Eli Lilly, Essex Pharma, Janssen & Cilag,
Johnson & Johnson, Lundbeck, Pfizer, Sanofi-Aventis, and Eli
Lilly; and has provided medication for a trial with SL as the
primary investigator.
S. Levander, G. Kjærsdam Tell
eus, S. Olrik Wallenstein
Jensen, and T. Østergaard Christensen have nothing to
declare.
References
1. Nielsen RE. Cognition in schizophrenia a systematic
review. Drug Discov Today Ther Strateg 2011;8:4348.
2. Buchanan RW, Keefe RS, Umbricht D, Green MF, Laugh-
ren T, Marder SR. The FDA-NIMH-MATRICS guide-
lines for clinical trial design of cognitive-enhancing drugs:
what do we know 5 years later? Schizophr Bull
2011;37:12091217.
3. Szoke A, Trandafir A, Dupont ME, Meary A, Schurhoff F,
Leboyer M. Longitudinal studies of cognition in schizo-
phrenia: meta-analysis. Br J Psychiatry 2008;192:248
257.
4. Dominguez MG, Viechtbauer W, Simons CJ, Van OJ, Krab-
bendam L. Are psychotic psychopathology and neurocogni-
tion orthogonal? A systematic review of their associations.
Psychol Bull 2009;135:157171.
5. Mohamed S, Rosenheck R, Swartz M, Stroup S, Lieberman
JA, Keefe RS. Relationship of cognition and psychopa-
thology to functional impairment in schizophrenia. Am J
Psychiatry 2008;165:978987.
6. Quee PJ, van der Meer L, Krabbendam L et al. Insight
change in psychosis: relationship with neurocognition,
social cognition, clinical symptoms and phase of illness.
Acta Psychiatr Scand 2014;129:126133.
7. Fervaha G, Foussias G, Agid O, Remington G. Motivational
and neurocognitive deficits are central to the prediction of
longitudinal functional outcome in schizophrenia. Acta
Psychiatr Scand 2014;130:290299.
8. Kakela J, Panula J, Oinas E, Hirvonen N, Jaaskelainen
E, Miettunen J. Family history of psychosis and social,
occupational and global outcome in schizophrenia:
a meta-analysis. Acta Psychiatr Scand 2014t;130:
269278.
9. Hill SK, Bishop JR, Palumbo D, Sweeney JA. Effect of sec-
ond-generation antipsychotics on cognition: current issues
and future challenges. Expert Rev Neurother 2010;10:43
57.
10. Nielsen RE, Nielsen J. Antipsychotic drug treatment for
patients with schizophrenia. Clin Med Therap
2009;1:10531068.
11. Minzenberg MJ, Poole JH, Benton C, Vinogradov S. Asso-
ciation of anticholinergic load with impairment of com-
plex attention and memory in schizophrenia. Am J
Psychiatry 2004;161:116124.
12. Correll CU. From receptor pharmacology to improved
outcomes: individualising the selection, dosing, and
switching of antipsychotics. Eur Psychiatry 2010;25(Suppl.
2):S12S21.
13. Keefe RSE, Bilder RM, Davis SM et al. Neurocognitive
effects of antipsychotic medications in patients with
chronic schizophrenia in the CATIE trial. Arch Gen Psy-
chiatry 2007;64:633647.
14. Davidson M, Galderisi S, Weiser M et al. Cognitive effects
of antipsychotic drugs in first-episode schizophrenia and
schizophreniform disorder: a randomized, open-label
clinical trial (EUFEST). Am J Psychiatry 2009;166:675
682.
15. Woodward ND, Purdon SE, Meltzer HY, Zald DH. A
meta-analysis of cognitive change with haloperidol in
clinical trials of atypical antipsychotics: Dose effects and
comparison to practice effects. Schizophr Res 2007;89:
211224.
16. Woodward ND, Purdon SE, Meltzer HY, Zald DH. A
meta-analysis of neuropsychological change to clozapine,
olanzapine, quetiapine, and risperidone in schizophrenia.
Int J Neuropsychopharmacol 2005;8:457472.
17. Keefe RS, Silva SG, Perkins DO, Lieberman JA. The effects
of atypical antipsychotic drugs on neurocognitive impair-
ment in schizophrenia: a review and meta-analysis. Schiz-
ophr Bull 1999;25:201222.
18. Mishara AL, Goldberg TE. A meta-analysis and critical
review of the effects of conventional neuroleptic treatment
on cognition in schizophrenia: opening a closed book. Biol
Psychiatry 2004;55:10131022.
19. Desamericq G, Schurhoff F, Meary A et al. Long-term
neurocognitive effects of antipsychotics in schizophrenia: a
network meta-analysis. Eur J Clin Pharmacol 2014;
70:127134.
20. Leucht S, Cipriani A, Spineli L et al. Comparative efficacy
and tolerability of 15 antipsychotic drugs in schizophrenia:
a multiple-treatments meta-analysis. Lancet 2013;382:951
962.
21. Cipriani A, Furukawa TA, Salanti G et al. Comparative
efficacy and acceptability of 12 new-generation antidepres-
sants: a multiple-treatments meta-analysis. Lancet 2009;
373:746758.
22. Cipriani A, Higgins JP, Geddes JR, Salanti G. Conceptual
and technical challenges in network meta-analysis. Ann
Intern Med 2013;159:130137.
23. Buchanan RW, Davis M, Goff D et al. A summary of the
FDA-NIMH-MATRICS workshop on clinical trial design
for neurocognitive drugs for schizophrenia. Schizophr Bull
2005;31:519.
24. Lezak M, Howieson D, Bigler E, Tranel D. Neuropsycho-
logical assessment. 5th edn. New York, NY: Oxford Uni-
versity Press Inc, 2012.
25. Green MF, Nuechterlein KH, Gold JM et al. Approaching
a consensus cognitive battery for clinical trials in schizo-
phrenia: the NIMH-MATRICS conference to select cogni-
tive domains and test criteria. Biol Psychiatry 2004;
56:301307.
26. Duval S, Tweedie R. Trim, and fill: a simple funnel-plot-
based method of testing and adjusting for publication bias
in meta-analysis. Biometrics 2000;56:455463.
27. Abdolahian E, Mohareri F, Bordbar MRF. Haloperidol
versus risperidone: a comparison of beneficial effect on
cognitive function of patients with chronic schizophrenia.
Iran J Psychiatry Behav Sci 2008;2:1420.
28. Bellack AS, Schooler NR, Marder SR, Kane JM, Brown
CH, Yang Y. Do clozapine and risperidone affect social
competence and problem solving? Am J Psychiatry
2004;161:364367.
29. Bender S, Dittmann-Balcar A, Schall U et al. Influence of
atypical neuroleptics on executive functioning in patients
with schizophrenia: a randomized, double-blind compari-
10
Nielsen et al.
son of olanzapine vs. clozapine. Int J Neuropsychophar-
macol 2006;9:135145.
30. Bilder RM, Goldman RS, Volavka J et al. Neurocognitive
effects of clozapine, olanzapine, risperidone, and haloperi-
dol in patients with chronic schizophrenia or schizoaffec-
tive disorder. Am J Psychiatry 2002;159:10181028.
31. Boulay LJ, Labelle A, Bourget D et al. Dissociating medi-
cation effects from learning and practice effects in a neuro-
cognitive study of schizophrenia: olanzapine versus
haloperidol. Cogn Neuropsychiatry 2007;12:322338.
32. Buchanan RW, Holstein C, Breier A. The comparative effi-
cacy and long-term effect of clozapine treatment on neuro-
psychological test performance. Biol Psychiatry
1994;36:717725.
33. Fagerlund B, Mackeprang T, Gade A, Glenthoj BY. Effects
of low-dose risperidone and low-dose zuclopenthixol on
cognitive functions in first-episode drug-naive schizo-
phrenic patients. CNS Spectr 2004;9:364374.
34. Gallhofer B, Bauer U, Lis S, Krieger S, Gruppe H. Cogni-
tive dysfunction in schizophrenia: comparison of treat-
ment with atypical antipsychotic agents and conventional
neuroleptic drugs. Eur Neuropsychopharmacol 1996;6
(Suppl. 2):S13S20.
35. Goldberg TE, Goldman RS, Burdick KE et al. Cognitive
improvement after treatment with second-generation
antipsychotic medications in first-episode schizophrenia:
is it a practice effect? Arch Gen Psychiatry 2007;64:
11151122.
36. Green MF, Marshall BD Jr, Wirshing WC et al. Does ris-
peridone improve verbal working memory in treatment-
resistant schizophrenia? Am J Psychiatry 1997;154:799
804.
37. Green MF, Marder SR, Glynn SM et al. The neurocogni-
tive effects of low-dose haloperidol: a two-year compari-
son with risperidone. Biol Psychiatry 2002;51:972978.
38. Grootens KP, Van Veelen NMJ, Sitskoorn MM et al.
Effects on cognitive functioning after olanzapineziprasi-
done crossover in recent-onset schizophrenia. Eur Neuro-
psychopharmacol 2010;20:907912.
39. Gurpegui M,
Alvarez E, Bouso~
no M, Ciudad A, G
omez JC,
Olivares JM. Effect of olanzapine or risperidone treatment
on some cognitive functions in a one-year follow-up of
schizophrenia outpatients with prominent negative symp-
toms. Eur Neuropsychopharmacol 2007;17:725734.
40. Harvey PD, Sacchetti E, Galluzzo A et al. A randomized
double-blind comparison of ziprasidone vs. Clozapine for
cognition in patients with schizophrenia selected for resis-
tance or intolerance to previous treatment. Schizophr Res
2008;105:138143.
41. Harvey PD, Green MF, McGurk SR, Meltzer HY.
Changes in cognitive functioning with risperidone and ola-
nzapine treatment: a large-scale, double blind, randomized
study. Psychopharmacology 2003;169:404411.
42. Harvey PD, Bowie CR, Loebel A. Neuropsychological nor-
malization with long-term atypical antipsychotic treat-
ment: results of a six-month randomized, double-blind
comparison of ziprasidone vs. olanzapine. J Neuropsychi-
atry Clin Neurosci 2006;18:5463.
43. Harvey PD, Patterson TL, Potter LS, Zhong K, Brecher
M. Improvement in social competence with short-term
atypical antipsychotic treatment: a randomized, double-
blind comparison of quetiapine versus risperidone for
social competence, social cognition, and neuropsychologi-
cal functioning. Am J Psychiatry 2006;163:19181925.
44. Honer WG, Thornton AE, Chen EY et al. Clozapine alone
versus clozapine and risperidone with refractory schizo-
phrenia. N Engl J Med 2006;354:472482.
45. Jerrell JM, Ramirez PM. Changes in neuropsychological
functioning following treatment with risperidone, olanza-
pine, and conventional antipsychotic medications. Hum
Psychopharmacol 2008;23:595604.
46. Keefe RS, Young CA, Rock SL et al. One-year double-
blind study of the neurocognitive efficacy of olanzapine,
risperidone, and haloperidol in schizophrenia. Schizophr
Res 2006;81:115.
47. Keefe RS, Sweeney JA, Gu H et al. Effects of olanzapine,
quetiapine, and risperidone on neurocognitive function in
early psychosis: a randomized, double-blind 52-week com-
parison. Am J Psychiatry 2007;164:10611071.
48. Kern RS, Green MF, Cornblatt BA et al. The neurocogni-
tive effects of aripiprazole: an open-label comparison with
olanzapine. Psychopharmacology 2006;187:312320.
49. Krakowski MI, Czobor P, Nolan KA. Atypical antipsy-
chotics, neurocognitive deficits, and aggression in schizo-
phrenic patients. J Clin Psychopharmacol 2008;28:485
493.
50. Lee MA, Thompson PA, Meltzer HY. Effects of clozapine
in cognitive function in schizophrenia. J Clin Psychiatry
1994;55:8287.
51. Lee MA, Jayathilake K, Meltzer HY. A comparison of the
effect of clozapine with typical neuroleptics on cognitive
function in neuroleptic-responsive schizophrenia. Schiz-
ophr Res 1999;37:111.
52. Lee S, Chou Y, Li M, Wan F, Yen M. Effects of antipsy-
chotics on cognitive performance in drug-naive schizo-
phrenic patients. Prog Neuropsychopharmacol Biol
Psychiatry 2007;31:11011107.
53. Lindenmayer JP, Khan A, Iskander A, Abad MT, Parker B.
A randomized controlled trial of olanzapine versus halo-
peridol in the treatment of primary negative symptoms
and neurocognitive deficits in schizophrenia. J Clin Psychi-
atry 2007;68:368379.
54. Ljubin T, Milas DZ, Mimica N, Folnegovic-Smalc V, Maka-
ric G. A preliminary study of the comparative effects of
olanzapine and fluphenazine on cognition in schizophrenic
patients. Hum Psychopharmacol 2000;15:513519.
55. McGurk SR, Carter C, Goldman R et al. The effects of
clozapine and risperidone on spatial working memory in
schizophrenia. Am J Psychiatry 2005;162:10131016.
56. Meltzer HY, Bobo WV, Roy A et al. A randomized, dou-
ble-blind comparison of clozapine and high-dose olanza-
pine in treatment-resistant patients with schizophrenia. J
Clin Psychiatry 2008;69:274285.
57. Mori K, Nagao M, Yamashita H, Morinobu S, Yamawaki S.
Effect of switching to atypical antipsychotics on
memory in patients with chronic schizophrenia. Prog
Neuropsychopharmacol Biol Psychiatry 2004;28:659665.
58. Mortimer AM, Joyce E, Balasubramaniam K, Choudhary
PC, Saleem PT. Treatment with amisulpride and olanza-
pine improve neuropsychological function in schizophre-
nia. Hum Psychopharmacol 2007;22:445454.
59. Muscatello MRA, Bruno A, Pandolfo G et al. Effect of
aripiprazole augmentation of clozapine in schizophrenia: a
double-blind, placebo-controlled study. Schizophr Res
2011;127:9399.
60. Nielsen R, Odur F, Østergaard T, Munk-Jørgensen P, Niel-
sen J. Comparison of the effects of Sertindole and Olanza-
pine on Cognition: a double-blind randomized 12-week
study of patients diagnosed with schizophrenia. Ther Adv
Psychopharmacol 2013;4:414.
61. Purdon SE, Jones BDW, Stip E et al. Neuropsychological
change in early phase schizophrenia during 12 months of
treatment with olanzapine, risperidone, or haloperidol.
Arch Gen Psychiatry 2000;57:249258.
11
SGAs effect on cognition
62. Purdon SE, Malla A, Labelle A, Lit W. Neuropsychologi-
cal change in patients with schizophrenia after treatment
with quetiapine or haloperidol. J Psychiatry Neurosci
2001;26:137149.
63. Remillard S, Pourcher E, Cohen H. The effect of neurolep-
tic treatments on executive function and symptomatology
in schizophrenia: a 1-year follow up study. Schizophr Res
2005;80:99106.
64. Remillard S, Pourcher E, Cohen H. Long-term effects of
risperidone versus haloperidol on verbal memory, atten-
tion, and symptomatology in schizophrenia. J Int Neuro-
psychol Soc 2008;14:110118.
65. Remillard S, Pourcher E, Cohen H. Long-term skill proced-
uralization in schizophrenia. J Int Neuropsychol Soc
2010;16:148156.
66. Riedel M, M
uller N, Spellmann I et al. Efficacy of olanza-
pine versus quetiapine on cognitive dysfunctions in
patients with an acute episode of schizophrenia. Eur Arch
Psychiatry Clin Neurosci 2007;257:402412.
67. Riedel M, Spellmann I, Strassnig M et al. Effects of risperi-
done and quetiapine on cognition in patients with schizo-
phrenia and predominantly negative symptoms. Eur Arch
Psychiatry Clin Neurosci 2007;257:360370.
68. Rosenheck R, Perlick D, Bingham S et al. Effectiveness and
cost of olanzapine and haloperidol in the treatment of
schizophrenia: a randomized controlled trial. JAMA
2003;290:26932702.
69. Sergi MJ, Green MF, Widmark C et al. Cognition and neu-
rocognition: effects of risperidone, olanzapine, and halo-
peridol. Am J Psychiatry 2007;164:15851592.
70. Smith RC, Infante M, Singh A, Khandat A. The effects of
olanzapine on neurocognitive functioning in medication-
refractory schizophrenia. Int J Neuropsychopharmacol
2001;4:239250.
71. van Veelen NM, Grootens KP, Peuskens J et al. Short term
neurocognitive effects of treatment with ziprasidone and
olanzapine in recent onset schizophrenia. Schizophr Res
2010;120:191198.
72. Velligan DI, Newcomer J, Pultz J et al. Does cognitive
function improve with quetiapine in comparison to halo-
peridol? Schizophr Res 2002;53:239248.
73. Velligan DI, Prihoda TJ, Sui D, Ritch JL, Maples N,
Miller AL. The effectiveness of quetiapine versus conven-
tional antipsychotics in improving cognitive and func-
tional outcomes in standard treatment settings. J Clin
Psychiatry 2003;64:524531.
74. Voruganti LP, Awad AG, Parker G et al. Cognition, func-
tioning and quality of life in schizophrenia treatment:
results of a one-year randomized controlled trial of ola-
nzapine and quetiapine. Schizophr Res 2007;96:146
155.
75. Wagner M, Quednow BB, Westheide J, Schlaepfer TE, Ma-
ier W, Kuhn K. Cognitive improvement in schizophrenic
patients does not require a serotonergic mechanism: ran-
domized controlled trial of olanzapine vs amisulpride.
Neuropsychopharmacology 2005;30:381390.
76. Gallhofer B, Jaanson P, Mittoux A, Tanghoj P, Lis S, Krie-
ger S. Course of recovery of cognitive impairment in
patients with schizophrenia: a randomised double-blind
study comparing sertindole and haloperidol. Pharmacop-
sychiatry 2007;40:275286.
77. Fagerlund B, Soholm B, Fink-Jensen A, Lublin H, Glenthoj
BY. Effects of donepezil adjunctive treatment to ziprasi-
done on cognitive deficits in schizophrenia: a double-blind,
placebo-controlled study. Clin Neuropharmacol 2007;30:
312.
78. Shekhar A, Potter WZ, Lightfoot J et al. Selective musca-
rinic receptor agonist xanomeline as a novel treatment
approach for schizophrenia. Am J Psychiatry
2008;165:10331039.
79. Freedman R, Olincy A, Buchanan RW et al. Initial phase 2
trial of a nicotinic agonist in schizophrenia. Am J Psychia-
try 2008;165:10401047.
80. Pae CU. Role of the cholinesterase inhibitors in the treat-
ment of schizophrenia. Expert Opin Investig Drugs
2013;22:293298.
81. Desmarais JE, Beauclair L, Margolese HC. Anticholiner-
gics in the era of atypical antipsychotics: short-term or
long-term treatment? J Psychopharmacol 2012;26:1167
1174.
82. McGurk SR, Green MF, Wirshing WC et al. Antipsychotic
and anticholinergic effects on two types of spatial
memory in schizophrenia. Schizophr Res 2004;68:225
233.
83. Tracy JI, Monaco CA, Abraham G, Josiassen RC, Pollock
BG. Relation of serum anticholinergicity to cognitive sta-
tus in schizophrenia patients taking clozapine or risperi-
done. J Clin Psychiatry 1998;59:184188.
84. Mork A, Witten LM, Arnt J. Effect of sertindole on extra-
cellular dopamine, acetylcholine, and glutamate in the
medial prefrontal cortex of conscious rats: a comparison
with risperidone and exploration of mechanisms involved.
Psychopharmacology 2009;206:3949.
85. Burnham KE, Baxter MG, Bainton JR et al. Activation of
5-HT(6) receptors facilitates attentional set shifting. Psy-
chopharmacology 2010;208:1321.
86. Rodefer JS, Nguyen TN, Karlsson JJ, Arnt J. Reversal of
subchronic PCP-induced deficits in attentional set shifting
in rats by sertindole and a 5-HT6 receptor antagonist:
comparison among antipsychotics. Neuropsychopharma-
cology 2008;33:26572666.
87. Herrera E, Cuetos F, Ribacoba R. Verbal fluency in Par-
kinson’s disease patients on/off dopamine medication.
Neuropsychologia 2012;50:36363640.
88. Kontis D, Theochari E, Kleisas S et al. Doubtful associa-
tion of antipsychotic polypharmacy and high dosage with
cognition in chronic schizophrenia. Prog Neuropsycho-
pharmacol Biol Psychiatry 2010;34:13331341.
89. Kawai N, Yamakawa Y, Baba A et al. High-dose of multi-
ple antipsychotics and cognitive function in schizophrenia:
the effect of dose-reduction. Prog Neuropsychopharmacol
Biol Psychiatry 2006;30:10091014.
12
Nielsen et al.
... 50 The differences between antipsychotics are not entirely clear. 51 One study suggested that lurasidone may be better than quetiapine for cognitive symptoms. 52 Benzodiazepines and anticholinergics for the treatment of parkinsonian side effects should be avoided in the long term. ...
Article
Full-text available
Background Schizophrenia is most times a chronic and often debilitating illness associated with poor mental health outcomes. Early and effective treatment of schizophrenia in the most appropriate setting can make a significant difference in the long-term recovery. The aim of this narrative review was to provide suggestions and recommendations for effectively managing patients with schizophrenia during acute exacerbations and to enhance awareness and skills related to personalized medicine. Methods A panel of academics and clinicians with experience in the field of psychosis met virtually on July 13th 2023 to narratively review and discuss the research evidence and their clinical experience about the most appropriate acute treatments for patients with schizophrenia. This manuscript represents a synthesis of the panel analysis and discussion. Results First contact is very important for service users, as is finding the most adequate treatment setting. If patients present to the emergency department, which may be a traumatic setting for service users, a dedicated environment with adequate space and specialized mental health support, including personnel trained in de-escalation techniques, is recommended. A well-connected continuum of care is strongly recommended, possibly with seamless links between inpatient units, day hospital services, outpatient facilities and rehabilitation services. Ideally, this should be structured as part of a coordinated step-down service line. Treatment challenges include suboptimal response, side effects, and nonadherence, which is reduced by the use of long-acting injectable antipsychotics. Conclusion Individual circumstances, including age, gender, and presence of hostility/aggression or self-harm, cognitive impairment and negative symptoms, comorbidities (depression, substance use disorders) or associated symptoms (anxiety, insomnia), should be considered when selecting the most appropriate treatment for the acute phase of schizophrenia. Efficacy and feasibility, as well as acceptability and tolerability of treatments, require joint consideration from the early stages of schizophrenia, thereby enhancing the possibility of improved short- and long-term outcomes.
... Although cognitive enhancers have not been identified, pharmacological interventions such as minimising the anticholinergic load (75,76) by means of selecting (newer) antipsychotics with less anticholinergic burden and using anti-muscarinic agents for extra-pyramidal side-effects sparingly, have been demonstrated to improve cognitive deficits in several studies (77)(78)(79)(80)(81)(82)(83). ...
... Second-generation Disclaimer/Publisher's Note: The statements, opinions, and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions, or products referred to in the content. 2 antipsychotics are believed to slightly improve patients' cognitive function, while none of them are found to have a satisfactory profile [15]. ...
Preprint
Full-text available
In patients with schizophrenia, metabolic syndrome is associated with cognitive impairments. We carried out a second analysis of data from a longitudinal trial to examine the relationship between weight gain and cognitive function in patients with first-episode schizophrenia (FES) over the first 6-month antipsychotic treatment. Baseline and 6-month endpoint measurements were taken for both cognitive function and body weight. Linear and logistic regression analyses were applied to investigate the bidirectional relationship between weight gain and cognitive function. Clinically relevant weight gain (CRW) was defined as an increase in body weight ex-ceeding 7%. The final analysis included 337 participants. Lower baseline scores in processing speed (OR=0.837, p=0.007), working memory and attention (OR=0.889, p=0.043), and executive function (OR=0.863, p=0.006) domains were significantly associated with CRW at the 6-month endpoint. After adjusting for confounders, CRW was found to be associated with improvement in the Brief Visuospatial Memory Test (p=0.036). The findings suggest that patients with lower baseline cognitive performance experienced more substantial weight gain. Conversely, weight gain was correlated with cognitive improvements, particularly in the domain of visual learning and memory. This suggested a potential bidirectional relationship between weight gain and cog-nitive function in patients with first-episode schizophrenia.
Article
Full-text available
Introduction Schizophrenia is a severe mental illness that usually begins in late adolescence or early adulthood. Current pharmacological treatments, while acceptably effective for many patients, are rarely clinically tailored or individualized. The lack of sufficient etiopathological knowledge of the disease, together with overall comparable effect sizes for efficacy between available antipsychotics and the absence of clinically actionable biomarkers, has hindered the advance of individualized medicine in the treatment of schizophrenia. Nevertheless, some degree of stratification based on clinical markers could guide treatment choices and help clinicians move toward individualized psychiatry. To this end, a panel of experts met to formally discuss the current approach to individualized treatment in schizophrenia and to define how treatment individualization could help improve clinical outcomes. Methods A task force of seven experts iteratively developed, evaluated, and refined questionnaire items, which were then evaluated using the Delphi method. Descriptive statistics were used to summarize and rank expert responses. Expert discussion, informed by the results of a scoping review on personalizing the pharmacologic treatment of adults and adolescents with schizophrenia, ultimately generated recommendations to guide individualized pharmacologic treatment in this population. Results There was substantial agreement among the expert group members, resulting in the following recommendations: 1) individualization of treatment requires consideration of the patient’s diagnosis, clinical presentation, comorbidities, previous treatment response, drug tolerability, adherence patterns, and social factors; 2) patient preferences should be considered in a shared decision-making approach; 3) identified barriers to personalized care that need to be overcome include the lack of actionable biomarkers and mechanistic similarities between available treatments, but digital tools should be increasingly used to enhance individualized treatment. Conclusion Individualized care can help provide effective, tailored treatments based on an individual’s clinical characteristics, disease trajectory, family and social environment, and goals and preferences.
Article
Cognitive Impairment Associated with Schizophrenia (CIAS) represents one of the core dimensions of Schizophrenia Spectrum Disorders (SSD), with an important negative impact on real-world functional outcomes of people living with SSD. Treatment of CIAS represents a therapeutic goal of considerable importance, and while cognition-oriented evidence-based psychosocial interventions are available, effective pharmacological treatment could represent a game-changer in the lives of people with SSD. The present critical review reports and discusses the evidence regarding the effects of several pharmacological agents that are available in clinical practice or are under study, commenting on both current and future perspectives of CIAS treatment. In particular, the effects on CIAS of antipsychotic medications, anticholinergic medications, benzodiazepines, which are currently commonly used in the treatment of SSD, and of iclepertin, D-serine, luvadaxistat, xanomeline-trospium, ulotaront, anti-inflammatory molecules, and oxytocin, which are undergoing regulatory trials or can be considered as experimental agents, will be reported and discussed. Currently available pharmacological agents do not appear to provide substantial benefits on CIAS, but an accurate management of antipsychotic medications and avoiding treatments that can further exacerbate CIAS represent important strategies. Some molecules that are currently being investigated in Phase 2 and Phase 3 trials have provided very promising preliminary results, but more information is currently required to assess their effectiveness in real-world contexts and to provide clear recommendations regarding their use in clinical practice. The results of ongoing and future studies will reveal whether any of these molecules represents the awaited pharmacological game-changer in the treatment of CIAS.
Article
Background The effectiveness of isolated resistance training (RT) on cognitive function among older adults with schizophrenia is insufficiently investigated. This study investigated the effectiveness of 12‐weeks POWER rehabilitation, a novel RT regimen, on cognitive function among older patients with schizophrenia and frailty. Methods Thirty‐two older adults with schizophrenia and frailty were enrolled and randomized to receive either a 12‐week, twice weekly POWER rehabilitation, or without add‐on training. Cognitive functioning was assessed using mini‐mental state examination (MMSE), digit symbol substitution test, color trail task (CTT), and digit span task (DST). Physical performance was assessed by walking speed and hand grip strength. The generalized estimating equations was used to compare pre‐ and post‐training outcome measure between groups. Results Between‐group analysis revealed significant improvement in CTT1 and hand grip strength in the intervention group compared to the controls. Subgroup analyses showed CTT1 performance significantly improved after 12 weeks of POWER rehabilitation in the intervention group (time, p < .001), independent of age, educational level, global cognition, depressive symptoms, and psychotropic medication use. Increased hand grip strength was significantly associated with improved performance in MMSE, CTT1, and DST forward at study endpoint. Conclusion A 12‐week POWER rehabilitation for older patients with schizophrenia and frailty is safe and feasible, and may benefit physical and some domains of cognitive functioning.
Article
Full-text available
Aim Baseline cognitive functions of patients predicted the efficacy of cognitive remediation therapy (CRT), but results are mixed. Eye movement is a more objective and advanced assessment of cognitive functions than neuropsychological testing. We aimed to investigate the applicability of eye movements in predicting cognitive improvement after patients with schizophrenia were treated with CRT. Methods We recruited 79 patients with schizophrenia to complete 8 weeks of CRT and assessed their cognitive improvement outcomes. Eye movements were assessed by prosaccades, antisaccades, and free-viewing tasks at baseline, and neuropsychological tests in four cognitive domains were assessed before and after treatment to calculate treatment outcomes. Predictors of demographic information, clinical characteristics, and eye movement measures at baseline on cognitive improvement outcomes were analyzed using logistic regression analysis. We further compared the predictive performance between eye movement measurements and neuropsychological test regarding the effect of CRT on cognitive improvement, and explored factors that could be affect the treatment outcomes in different cognitive domains. Results As operationally defined, 33 patients showed improved in cognition (improved group) and 46 patients did not (non-improved group) after CRT. Patients with schizophrenia being employed, lower directional error rate in antisaccade task, and lower the gap effect (i.e., the difference in saccadic latency between the gap condition and overlap condition) in prosaccade task at baseline predicted cognitive improvement in CRT. However, performance in the free-viewing task not associated with cognitive improvement in patients in CRT. Our results show that eye-movement prediction model predicted the effect of CRT on cognitive improvement in patients with schizophrenia better than neuropsychological prediction model in CRT. In addition, baseline eye-movements, cognitive reserve, antipsychotic medication dose, anticholinergic cognitive burden change, and number of training sessions were associated with improvements in four cognitive domains. Conclusion Eye movements as a non-invasiveness, objective, and sensitive method of evaluating cognitive function, and combined saccadic measurements in pro- and anti-saccades tasks could be more beneficial than free-viewing task in predicting the effect of CRT on cognitive improvement in patients with schizophrenia.
Article
Full-text available
Patients with schizophrenia often encounter notable weight gain during their illness, heightening the risk of metabolic diseases. While previous studies have noted a correlation between obesity and cognitive impairment in schizophrenia, many were cross-sectional, posing challenges in establishing a causal relationship between weight gain and cognitive function. The aim of this longitudinal study is to examine the relationship between weight gain and cognitive function in patients with first-episode schizophrenia (FES) during the initial 6-month antipsychotic treatments. Employing linear and logistic regression analyses, the study involved 337 participants. Significantly, baseline scores in processing speed (OR = 0.834, p = 0.007), working memory and attention (OR = 0.889, p = 0.043), and executive function (OR = 0.862, p = 0.006) were associated with clinically relevant weight gain (CRW, defined as an increase in body weight > 7%) at the 6-month endpoint. On the other hand, CRW correlated with improvements in the Brief Visuospatial Memory Test (p = 0.037). These findings suggest that patients with lower baseline cognitive performance undergo more substantial weight gain. Conversely, weight gain was correlated with cognitive improvements, particularly in the domain of visual learning and memory. This suggested a potential bidirectional relationship between weight gain and cognitive function in first-episode schizophrenia patients.
Article
Background: Studies on the effects of antipsychotics on cognitive deficits in schizophrenia mostly suggest a superior effect of atypical over typical compounds, although findings are inconsistent and effect sizes small. Several methodological issues, such as heterogenous patient samples, incomparable drug doses, effects of prior medication, construct validity, and retest effects on neuropsychological tasks, confound most results and the comparability between studies. Consequently, the conclusion concerning effects of antipsychotics on cognition is still equivocal. Objective: The present randomized clinical trial examined the effects on cognition of comparatively low doses of a typical antipsychotic (zuclopenthixol) and an atypical antipsychotic (risperidone) in a homogenous group of drug-naïve first-episode schizophrenic patients in a longitudinal setting. Methods: First-episode schizophrenic patients who had never previously been exposed to antipsychotic treatment (N-25) were randomly allocated to treatment with flexible doses of zuclopenthixol or risperidone in an open-label design. Cognitive functions were examined both when patients were drug-naïve, and after 13 weeks of treatment. A comprehensive neuropsychological battery was used in order to optimize construct validity, and principal components of cognitive functions were extrapolated in order to reduce type I errors. A healthy control group was tested at baseline and after 13 weeks, in order to examine retest effects. The cognitive domains studied were executive functions, selective attention, and reaction time. Results: The patients showed considerable cognitive deficits when drug-naïve. There were few differential effects of risperidone and zuclopenthixol on cognitive deficits, except for a differential significance, respectively, tendency towards improved reaction and movement times in the risperidone group, and a lack of such in the zuclopenthixol group. These differences were no longer significant after covarying for extrapyramidal side effects and anticholinergic medication that were more prevalent in the zuclopenthixol group and the increases after medication were comparable with retest effects in controls. Conclusion: The study underscores the importance of examining impact of factors, such as clinical improvement, extrapyramidal side effects, anticholinergic medication and retest effects in longitudinal efficacy studies. This study does not support efficacy of either risperidone or zuclopenthixol on cognitive functions in drug-naïve schizophrenia patients after 3 months of medication, because neither could be distinguished from retest effects of the healthy control group.
Article
Background: Neurocognitive deficits are core features of schizophrenia that are linked to functional outcome for the disorder. Recent studies and reviews have concluded that newer antipsychotic medications are better for neurocognitive deficits than conventional antipsychotic medications; however, one difficulty in interpreting this literature is that the comparisons have mainly been with high doses of conventional medications. This study examined the neurocognitive effects of low-dose haloperidol compared with risperidone over a 2-year period. Methods: Sixty-two patients were randomly assigned to medication (starting at 6 mg of each medication) and administered neurocognitive batteries six times over the course of follow-up. At 6 months, the mean dose of haloperidol was 5.0 mg, and the mean dose of risperidone was 6.0 mg. Neurocognitive data were reduced into cluster scores and a global summary score. Results: We found no significant overall differences in treatment effects on the cluster scores or the global score. The global score revealed a significant group by time interaction, reflecting the fact that the haloperidol group tended to improve initially and then stay stable, whereas the risperidone group improved more gradually over the follow-up period. Conclusions: This study did not provide support for neurocognitive advantages of a newer antipsychotic medication over a low-dose conventional medication. We speculate that conventional medications may have neurocognitive benefits at low doses that are neutralized or reversed at higher doses.
Article
Objective: The current study was performed to evaluate the cognitive improvements of the chronic schizophrenic patients treated with risperidone in comparison with those treated with haloperidol according to Wisconsin Card Sorting Test (WCST). Methods: In a double blind clinical trial, 65 patients with a diagnosis of chronic schizophrenia were randomly allocated into two groups. They received a 7 days washout and then during an eight weeks period one group was treated with risperidone 4-8 mg daily while patients in the other group received haloperidol 10-15 mg daily. Patients of the two groups were assessed by positive and Negative Syndrome Scale (PANSS) and Brief Psychiatric Rating Scale (BPRS). Patients' cognitive abilities were assessed by WCST. Treatment side effects were also evaluated in both groups. Results: The overall PANSS score, the scores of the positive and negative subscales and BPRS scores revealed that risperidone was significantly superior to haloperidol in the treatment of psychotic symptoms (p<0.001). Risperidone caused less marked dyskinetic side effects in comparison with halopridol (p<0.001). Haloperidol produced more symptoms of parkinsonism and tardive dykinesa than risperidone. The positive cognitive effect of risperidone was significantly better than haloperidol at 4th (p<0.001) and 8th (p<0.001) weeks. Conclusion: Apart from being more effective in improving positive and negative symptoms of psychotic disorders, risperidone is also more beneficial in reducing the symptoms of cognitive impairment in chronic and long standing form of schizophrenia. It also seems to be better tolerated than haloperidol.
Article
Objective We aimed to investigate associations between family history of psychosis and long-term occupational, social and global (i.e. combined occupational, social and clinical) outcome in schizophrenia.MethodA systematic search to identify potentially relevant studies was conducted using seven electronic databases and a manual search of literature. Only observational studies with a follow-up period of at least 2 years were included.ResultsThe search identified 4081 unique potentially relevant articles, of which 14 met our inclusion criteria. The presence of family history of psychosis was associated with poor occupational and global outcome (n = 3; r = 0.17; P = 0.008, n = 11; r = 0.13; P = 0.002, respectively).Conclusion This was the first systematic review on the effects of family history of psychosis on occupational and social outcome in schizophrenia. Based on the review, the presence of family history of psychosis has a relatively small but statistically significant association with long-term occupational and global outcome in patients with schizophrenia.