Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Journal of Psychiatric Research 46 (2012) 667e674

Contents lists available at SciVerse ScienceDirect

Journal of Psychiatric Research


journal homepage: www.elsevier.com/locate/psychires

Apathy in currently nondepressed patients treated with a SSRI for a major


depressive episode: Outcomes following randomized switch to either duloxetine
or escitalopram
Joel Raskin a, *, Thomas George b, Renee E. Granger c, Nadia Hussain a, George Weizhong Zhao c,
Lauren B. Marangell d, e
a
Lilly Research Laboratories, Eli Lilly Canada, 3650 Danforth Ave, Toronto, ON, Canada M1N 2E8
b
North West Specialist Centre, Everton Park, QLD, Australia
c
Development Centre of Excellence Asia-Pacific, Eli Lilly Australia Pty Ltd, Macquarie Park, NSW, Australia
d
Eli Lilly and Company, Lilly USA, LLC, Indianapolis, IN, USA
e
University of Texas Health Science Center School of Medicine, Houston, TX, USA

a r t i c l e i n f o a b s t r a c t

Article history: Apathy in the context of treated major depressive disorder (MDD) is a common but understudied
Received 10 August 2011 symptom. This multicenter, double-blind, randomized study investigated whether switching from
Received in revised form a selective serotonin reuptake inhibitor (SSRI) to a serotonin-norepinephrine reuptake inhibitor (SNRI),
6 February 2012
compared with switching to another SSRI, improved apathy symptoms in patients who had been treated
Accepted 17 February 2012
with a SSRI for MDD for !3 months, were no longer depressed (Montgomery-Åsberg Depression Rating
Scale [MADRS] total score "15), and continued to have apathy (Apathy Evaluation Scale e Clinician rated
Keywords:
version [AES-C] total score >30). Following 8 weeks of treatment, both the duloxetine (SNRI, 244
Apathy
Major depressive disorder
patients) and escitalopram (SSRI, 239 patients) groups significantly improved from baseline on the AES-C
Duloxetine hydrochloride total score (least squares mean change [standard error]: duloxetine #13.9 [0.54]; escitalopram #13.5
Escitalopram [0.54], both P < 0.001), and on the secondary apathy, depression, and functional outcomes. There were
Norepinephrine no significant differences between the two groups on any measure, including AES-C total score (least
Serotonin squares mean difference [95% confidence interval]: #0.4 [#1.87 to 1.10], P ¼ 0.612; primary objective).
There was a significant within-group improvement in apathy in the subgroup who received escitalopram
before and during the study. There were few differences in safety between the two groups. This study did
not support the hypothesis that switching from a SSRI to a SNRI has a beneficial effect on apathy
symptoms. However, given the study limitations, it is possible that more specific targeting of the
noradrenergic pathway would be of benefit.
! 2012 Elsevier Ltd. All rights reserved.

1. Introduction respectively, of 161 patients who had completed a course of treat-


ment for depression with a selective serotonin reuptake inhibitor
Residual symptoms in the context of “successful” antidepressant (SSRI). In a cross-sectional study, Fava et al. (2006) reported apathy in
treatment for major depressive disorder (MDD) are increasingly 32.5% of 117 depressed patients who were in remission or partial
recognized as a frequent occurrence (McClintock et al., 2011; Fava remission following at least 3 months of antidepressant treatment for
et al., 2006). These symptoms are relevant because of the acute MDD. Residual symptoms may be a remnant of the original depres-
discomfort felt by the patient and because they predict a greater sion, a side effect of the antidepressant, or perhaps both. While apathy
likelihood of poor outcomes overall, including relapse (Paykel et al., may occur in the context of treatment with any antidepressant, most
1995) and treatment discontinuation (Demyttenaere and Jaspers, reports (all small in number) have been in the context of SSRI treat-
2008; Bolling and Kohlenberg, 2004). Bolling and Kohlenberg ment, with speculation that this effect is treatment-emergent
(2004) reported apathy and loss of ambition in 18.6% and 16.1%, (Marangell et al., 2002; Demyttenaere and Jaspers, 2008).
Apathy has been associated with reduced activity of the
noradrenergic system (Birkmayer and Riederer, 1989; Gold and
* Corresponding author. Tel.: þ1 416 699 7260; fax: þ1 416 699 7352. Chrousos, 1999). In addition, noradrenergic activity is modulated
E-mail addresses: Raskin_Joel@lilly.com, joel.raskin@lilly.com (J. Raskin). by serotonin (5-hydroxytryptamine, 5-HT) such that unopposed

0022-3956/$ e see front matter ! 2012 Elsevier Ltd. All rights reserved.
doi:10.1016/j.jpsychires.2012.02.010
668 J. Raskin et al. / Journal of Psychiatric Research 46 (2012) 667e674

serotonergic manipulation may decrease noradrenergic activity (escitalopram or other SSRI). The study consisted of four periods
(Segal, 1979), possibly via stimulation of 5-HT 2C receptors (Gobert that included 8 weeks of treatment after randomization followed
et al., 2000). Therefore, increasing noradrenergic activity by by an optional tapering period. Patients remained on their current
switching to a serotonin-norepinephrine reuptake inhibitor (SNRI) SSRI treatment during the Screening Period while their eligibility
is hypothesized to result in a greater improvement in apathy than for the study was evaluated. During the Acute Therapy Period,
switching to another SSRI. patients were enrolled and randomized to switch from their
At present, there is no consensus on treating apathy in the current SSRI treatment to duloxetine 60 mg/day or escitalopram
context of MDD. In order to assess the acute benefits of switching to 10 mg/day in a double-blinded manner for 1 week. During the 7-
a SNRI, the current study was conducted in patients with apathy week Optimization Period, patients who experienced a worsening
who were nondepressed following at least 3 months of treatment in their depressive symptoms (based on the clinical judgment of
with a SSRI for a major depressive episode. The primary objective the investigator) could have their doses optimized (duloxetine
was to compare the mean baseline to endpoint change in the 120 mg/day or escitalopram 20 mg/day), at the discretion of the
Apathy Evaluation Scale e Clinician rated version (AES-C) total investigator. This was followed by a 2-week Optional Taper Period,
score between patients receiving duloxetine or escitalopram after 8 during which patients could taper off their study treatment to
weeks of treatment. Secondary objectives included comparison reduce the likelihood of discontinuation-emergent adverse events.
between the two treatment groups of mean changes from baseline
scores and/or longitudinal profiles in a variety of apathy, depres- 2.4. Study assessments
sion, and functional measures, and comparison of the safety and
tolerability of duloxetine and escitalopram. Investigator-assessed instruments included the AES-C (Marin
et al., 1991), the Rothschild Scale for Antidepressant Tachyphy-
2. Materials and methods laxis (RSAT) (Rothschild, 2008), the MADRS (Montgomery and
Åsberg, 1979), and the Clinical Global Impression of Severity (CGI-
2.1. Study information Severity) scale (Guy, 1976). The AES-C (18 items) assesses cognitive,
behavioral, emotional, and other symptoms of apathy based on
This was a multicenter, double-blind, randomized study con- verbal and nonverbal information provided by the patient. In the
ducted in eight countries (Australia, Canada, China, Italy, Mexico, current study, residual apathy was defined as an AES-C total score
Korea, Russia, Taiwan) between September 2009 and December of >30. The RSAT (7 items) assesses symptoms of apathy or
2010. The study was conducted in accordance with the latest decreased motivation among depressed patients who have ach-
version of the Declaration of Helsinki and Good Clinical Practice ieved symptomatic remission with antidepressant treatment. The
guidelines. The study design was reviewed and approved by instrument consists of 6 self-report items (energy level, motivation
appropriate institutional ethics review boards. Written informed and interest, cognitive functioning, weight gain, sleep, and sexual
consent was obtained from the study participants after the nature functioning) and affect (assessed by the interviewer). An RSAT score
of the study procedures had been fully explained. The study was of !7 has been hypothesized to reflect antidepressant tachyphy-
registered at www.clinicaltrials.gov (NCT00985504). laxis (Rothschild, 2008). The MADRS (10 items) measures severity
of depressive mood symptoms. The MADRS Item 8 assesses the
2.2. Eligibility criteria patient’s inability to feel (through evaluation of their interest in
their surroundings or activities that normally give pleasure) and
Eligible patients were outpatients aged !18 years with a histor- their ability to react with adequate emotion to circumstances or
ical diagnosis of MDD, as defined by the Diagnostic and Statistical people. The CGI-Severity scale is used in the presence of the patient
Manual of Mental Disorders, 4th edition (DSM-IV). Patients were to record the severity of illness at the time of assessment.
required to have received treatment with a SSRI (escitalopram, The patient-reported instruments included the Patient’s Global
sertraline, paroxetine, or citalopram) for MDD for at least 3 months Impression of Improvement (PGI-Improvement) scale (Guy, 1976),
before randomization, and to no longer be experiencing an episode the Massachusetts General Hospital Cognitive and Physical Func-
of MDD, based on a Montgomery-Åsberg Depression Rating Scale tioning Questionnaire (CPFQ) (Fava et al., 2009), and the Sheehan
(MADRS) total score "15 and Item 1 (apparent sadness) score of <2 Disability Scale (SDS) (Sheehan et al., 1998). The PGI-Improvement
at the start and end of the Screening Period (Visits 1 and 2). Patients scale rates the improvement of symptoms since the previous visit.
were required to have an AES-C total score >30 at Visits 1 and 2. A The CPFQ (7 items) evaluates cognitive and physical well being. This
negative urine test for pregnancy at the time of enrollment and questionnaire assesses motivation, wakefulness, energy, focus,
agreement to use a reliable method of birth control was required for word-finding difficulty, and mental acuity. The SDS assesses the
females of child-bearing potential. Exclusion criteria included: functional impact of symptoms on several inter-related domains.
a previous lack of response to an adequate trial of duloxetine or Patients are also asked to provide their impression of how many
escitalopram within the past 12 months; any current or historical days were lost or rendered unproductive during the past week
DSM-IV diagnosis of mania, bipolar disorder, treatment-resistant because of their symptoms.
depression (defined as failure of two antidepressant trials of
adequate dose and duration), or psychosis; current suicide risk, as 2.5. Study outcomes
assessed by the Mini International Neuropsychiatric Interview and
Columbia Suicide Severity Rating Scale; pregnant or breast-feeding; The primary endpoint was the comparison of the mean baseline
and abnormal thyroid stimulating hormone concentration. to endpoint change in AES-C total score between patients
randomized to switch treatment to duloxetine or escitalopram for 8
2.3. Study design weeks. The secondary endpoints included comparison between the
duloxetine and escitalopram treatment groups on: mean changes
Eligible patients were randomly assigned (1:1) to double-blind from baseline scores and longitudinal profiles for RSAT total score,
(double-dummy) duloxetine or escitalopram treatment. Randomi- MADRS total and Item 8 scores, CGI-S rating scale, and PGI-I rating
zation was achieved with an interactive voice response system scale; mean changes from baseline scores for CPFQ total score and
(IVRS) and was stratified by site and previous SSRI treatment SDS total score; the proportion of patients who relapsed (defined as
J. Raskin et al. / Journal of Psychiatric Research 46 (2012) 667e674 669

MADRS total score !16) and the time to relapse; and serious groups who met the relapse criterion were compared at endpoint
adverse events (SAEs), treatment-emergent adverse events using a last observation carried forward approach and the Fisher’s
(TEAEs), discontinuations due to adverse events, and vital signs. exact test. Time to relapse was analyzed using the KaplaneMeier
method. The incidence of SAEs, TEAEs, and discontinuations due
to adverse events was compared between the two treatment
2.6. Statistical analysis
groups using Fisher’s exact test. Mean changes from baseline to last
observation for vital signs were assessed using an MMRM
Assuming an effect size of 0.28, the study was estimated to have
approach. Statistical analyses were conducted using SAS Version
approximately 86% power for the primary objective with 240
9.1.3 (SAS Institute, Cary, N.C.).
patients in each treatment group using a two-sided test at an alpha
level of 0.05. Based on the estimation that approximately 4% of
patients would not have a postbaseline measure of AES-C, the 3. Results
planned sample size was 500 patients. Patients who were switched
directly from escitalopram to escitalopram formed a control 3.1. Patient disposition
subgroup. In addition, all patients who had received escitalopram
for at least 3 months before the study were excluded in a sensitivity In total, 483 patients met the eligibility criteria and were
analysis of the primary objective. Efficacy analyses included all randomized in the study, with 244 and 239 patients randomized to
patients with a baseline and at least one postbaseline observation. the duloxetine and escitalopram treatment groups, respectively
Safety analyses included all randomized patients who received at (Fig. 1). The majority of the randomized patients (407/483 patients,
least one dose of study drug. The primary objective was assessed 84.3%) completed 8 weeks of treatment (Fig. 1).
using a mixed-effects model with repeated measures (MMRM)
approach. Mean changes from baseline in secondary efficacy 3.2. Patient characteristics
measures were analyzed by MMRM or by analysis of covariance
(ANCOVA). Effect sizes (ES) were calculated for apathy measures Patient characteristics are summarized in Table 1. The median
using Cohen’s d. The percentages of patients in the two treatment age of the patients enrolled in the study was 45.0 years and

Fig. 1. Patient disposition.


670 J. Raskin et al. / Journal of Psychiatric Research 46 (2012) 667e674

Table 1
Patient characteristics.

Characteristic Duloxetine (N ¼ 244) Escitalopram (N ¼ 239) Overall (N ¼ 483)


Median age, years 45.1 45.0 45.0
Minimum, maximum 18.7, 73.9 19.2, 78.0 18.7, 78.0
Gender, n (%)
Female 187 (76.6) 179 (74.9) 366 (75.8)
Male 57 (23.4) 60 (25.1) 117 (24.2)
Ethnicity, n (%)
African/African American 0 (0) 1 (0.4) 1 (0.2)
Asian 90 (36.9) 82 (34.3) 172 (35.6)
Caucasian 120 (49.2) 119 (49.8) 239 (49.5)
Native American/Alaskan Native 34 (13.9) 37 (15.5) 71 (14.7)
Native Hawaiian/Pacific Islander 1 (0.4) 0 (0) 1 (0.2)
Mean (SD) BMI, kg/m2 25.6 (5.56) 26.8 (7.03) 26.2 (6.36)
Previous treatment, n (%)
Escitalopram 67 (27.5) 60 (25.1) 127 (26.3)
Other SSRI 177 (72.5) 179 (74.9) 356 (73.7)
Median age at first MDD episode, years (range) 34.0 (6.0e72.4) 34.7 (8.0e76.6) 34.2 (6.0e76.6)
Previous episodes of MDD
Experienced one or more, n (%) 175 (71.7) 182 (76.2) 357 (73.9)
Number of previous episodes, median (range) 1.0 (0e63.0) 2.0 (0e12.0) 1.0 (0e63.0)
Median duration of current episode, months (range) 5.0 (0e204.0) 6.0 (0e360.0) 5.8 (0e360.0)
Median time between last/current episode, months (range) 12.0 (0e360.0) 17.0 (0e252.0) 14.0 (0e360.0)

BMI ¼ body mass index; MDD ¼ major depressive disorder; SD ¼ standard deviation; SSRI ¼ selective serotonin reuptake inhibitor.

approximately three-quarters of the enrolled patients (75.8%) were on a higher than minimum dose of SSRI. During the study, 87/244
female. Before the study, 87 patients (18.0%) had received cit- patients (35.7%) in the duloxetine treatment group had a dose
alopram, 127 patients (26.3%) escitalopram, 130 patients (26.9%) escalation to 120 mg/day and 93/239 patients (38.9%) in the esci-
paroxetine, 134 patients (27.7%) sertraline, and 5 patients (1.0%) had talopram treatment group had a dose escalation to 20 mg/day.
received other drugs (fluoxetine hydrochloride, 2; fluvoxamine, 1;
metoclopramide, 1; reboxetine, 1). The median age at the first MDD 3.4. Clinical outcomes
episode was 34.2 years. Approximately three-quarters of patients
(73.9%) had experienced one or more episodes of MDD, and the The mean baseline to endpoint change in AES-C total score after 8
median (range) number of episodes was 1.0 (0e63.0). There was weeks of treatment was not statistically significantly different
clinically significant apathy in the study population at baseline between the duloxetine and escitalopram treatment groups (Table 3,
and patients were nonsyndromic but symptomatic for MDD: the Fig. 2). The least squares (LS) mean difference (95% confidence
mean AES-C total score (standard deviation [SD]) was 46.3 (7.97) interval [CI]) was #0.4 (#1.87 to 1.10, P ¼ 0.612, ES #0.05) (Table 3).
and the mean MADRS total score (SD) was 10.4 (3.59) (Table 2). There were improvements in apathy (AES-C, RSAT, MADRS Item 8),
Patient characteristics and apathy, depression, and functioning mood (MADRS), and functioning (CGI-S, PGI-I, CPFQ, SDS) (Table 3)
were similar in the duloxetine and escitalopram treatment groups in both the duloxetine and escitalopram treatment groups after 8
at baseline (Tables 1and 2). weeks of treatment. There were no statistically significant differ-
ences between the duloxetine and escitalopram treatment groups
for any of the secondary apathy, depression, or functional outcomes
3.3. Dose escalations after 8 weeks of treatment (Table 3).
There was a significant within-group improvement in AES-C
Before the study, 80/241 patients (33.2%) in the duloxetine total score in the group of patients who switched directly from
treatment group, 89/236 patients (37.7%) in the escitalopram escitalopram to escitalopram (escitalopram to escitalopram
treatment group, and 30/58 patients (51.7%) in the group of patients subgroup, n ¼ 57) over the 8-week treatment period. The LS mean
who switched directly from escitalopram to escitalopram had been change over the treatment period (as an average of all visits)
was #6.6 (standard error [SE] 0.85, P < 0.001). There was no
Table 2
statistically significant difference between the escitalopram to
Apathy, depression, and functioning at baseline.
escitalopram subgroup and the subgroup who switched from
Scalea Scale Duloxetine Escitalopram Overall escitalopram to duloxetine (P ¼ 0.600). In addition, there was no
range (N ¼ 244) (N ¼ 239) (N ¼ 483)
statistically significant difference between the subgroups who
Mean (SD) Mean (SD) Mean (SD) switched to escitalopram or duloxetine from another SSRI (i.e., not
AES-C total 18e72 46.3 (7.82) 46.3 (8.14) 46.3 (7.97) escitalopram, P ¼ 0.954). The sensitivity analysis, which removed
RSAT total 0e28 10.7 (4.92) 10.5 (4.95) 10.6 (4.93) all patients who had received escitalopram for at least 3 months
MADRS total 0e60 10.6 (3.49) 10.3 (3.68) 10.4 (3.59)
MADRS item 8 0e6 1.8 (1.10) 1.8 (1.15) 1.8 (1.13)
before the study, had no significant effect on the difference
CGI-S 1e7 3.1 (0.91) 3.0 (0.91) 3.1 (0.91) between the two treatment groups in the mean baseline to
CPFQ total 7e42 24.8 (5.73) 24.7 (5.83) 24.7 (5.78) endpoint change in AES-C total score after 8 weeks of treatment (LS
SDS total 0e30 15.3 (6.76) 14.6 (6.39) 15.0 (6.58) mean difference [95% CI]: 0.1 [#1.70 to 1.80], P ¼ 0.954).
AES-C ¼ Apathy Evaluation Scale e Clinician rated version; CGI-S ¼ Clinical Global An ad hoc analysis showed that the mean baseline to endpoint
Impression of Severity; CPFQ ¼ Cognitive and Physical Functioning Questionnaire; change in AES-C total score after 8 weeks of treatment was greater
MADRS ¼ Montgomery-Åsberg Depression Rating Scale; RSAT ¼ Rothschild Scale in the group of patients who were on the minimum SSRI dose
for Antidepressant Tachyphylaxis; SD ¼ standard deviation; SDS ¼ Sheehan
Disability Scale.
before the study compared with the group on a higher than
a
For each of the scales, the higher the score, the more severe the condition under minimum SSRI dose. The LS mean (SE) for the group on the
assessment. minimum SSRI dose before the study was #14.5 (0.51) and the LS
J. Raskin et al. / Journal of Psychiatric Research 46 (2012) 667e674 671

Table 3
Change in apathy, depression, and functioning from baseline to 8 weeks.

Scalea Duloxetine (N ¼ 244) Escitalopram (N ¼ 239) Difference Effect sizeb P-value

LS mean (SE) LS mean (SE) LS mean (95% CI) Week 8c Treatmentd Treatment by visite
AES-C total #13.9 (0.54) #13.5 (0.54) #0.4 (#1.87 to 1.10) #0.05 0.612 0.674 0.964
RSAT total #5.5 (0.26) #5.0 (0.26) #0.5 (#1.22 to 0.20) #0.15 0.157 0.500 0.085
MADRS total #4.2 (0.32) #4.1 (0.33) #0.1 (#0.96 to 0.82) NC 0.880 0.866 0.360
MADRS item 8 #1.0 (0.06) #0.9 (0.06) #0.1 (#0.27 to 0.06) #0.09 0.224 0.101 0.061
CGI-S #0.9 (0.06) #0.9 (0.06) 0.1 (#0.09 to 0.22) NC 0.410 0.552 0.840
PGI-If 2.6 (0.08) 2.6 (0.08) 0 (#0.19 to 0.27) NC 0.723 0.792 0.170
CPFQ total #7.0 (0.34) #6.9 (0.35) #0.1 (#0.97 to 0.87) #0.01 0.910 0.910 N/A
SDS total #7.6 (0.40) #7.7 (0.41) 0.1 (#0.96 to 1.22) NC 0.821 0.821 N/A

All within-group P-values from t test on LS mean change were <0.001. All models were adjusted by including the fixed effect of investigator and baseline measure (except PGI).
AES-C ¼ Apathy Evaluation Scale e Clinician rated version; ANCOVA ¼ analysis of covariance; CGI-S ¼ Clinical Global Impression of Severity; CI ¼ confidence interval;
CPFQ ¼ Cognitive and Physical Functioning Questionnaire; LS ¼ least squares; MADRS ¼ Montgomery-Åsberg Depression Rating Scale; MMRM ¼ mixed-effects model
repeated measures; N/A ¼ not applicable; NC ¼ not calculated; PGI-I ¼ Patient Global Impression of Improvement; RSAT ¼ Rothschild Scale for Antidepressant Tachyphylaxis;
SDS ¼ Sheehan Disability Scale; SE ¼ standard error.
a
For each of the scales, a negative LS mean value denotes improvement on the scale.
b
Effect sizes calculated using Cohen’s d.
c
Between group P-values from t test on LS mean difference based on MMRM or ANCOVA (CPFQ and SDS only).
d
Main effect F test for treatment in MMRM or ANCOVA model.
e
Main effect F test for interaction term (treatment by visit) in MMRM over the 8-week treatment period.
f
PGI-I scores are presented as means rather than mean change from baseline values.

mean (SE) for the group on a higher than minimum SSRI dose duloxetine and escitalopram treatment groups (Table 4). There was 1
was #12.7 (0.70); the LS mean difference (95% CI) was #1.84 (#3.57 death (suicide) in the escitalopram treatment group. Approximately
to #0.11, P ¼ 0.037). half of the patients in the study experienced one or more TEAE; the
most commonly reported TEAEs were nausea, headache, con-
3.5. Relapse stipation, and insomnia (Table 4). More patients in the duloxetine
compared with the escitalopram treatment group experienced
The relapse criterion (MADRS total score !16) was met by 55/ TEAEs of constipation (9.4% vs 3.8%) and anxiety (3.7% vs 0.4%).
480 patients (11.5%) in the study population, and 29/243 patients Statistically significantly greater increases in heart rate and
(11.9%) and 26/237 patients (11.0%) in the duloxetine and escitalo- diastolic blood pressure were observed in the duloxetine compared
pram treatment groups, respectively. The minimum and maximum with the escitalopram treatment group after 8 weeks of treatment.
time to relapse was 4 and 81 days, respectively. The median time to The LS mean change (SE) in heart rate was 3.6 (0.51) and 1.0 (0.52)
relapse could not be calculated using the KaplaneMeier procedure. in the duloxetine (N ¼ 212) and escitalopram (N ¼ 207) treatment
The relapse criterion was met by 10/59 patients (17.0%) in the groups, respectively (LS mean difference [95% CI]: 2.7 [1.26e4.08],
escitalopram to escitalopram subgroup. P < 0.001). The LS mean change (SE) in diastolic blood pressure was
1.4 (0.49) and #0.4 (0.50) in the duloxetine (N ¼ 212) and escita-
lopram (N ¼ 207) treatment groups, respectively (LS mean differ-
3.6. Safety ence [95% CI]: 1.8 [0.43e3.11], P ¼ 0.010).

The rates of SAEs, discontinuations due to adverse events, or


discontinuations due to lack of efficacy were similar in the
Table 4
Summary of adverse events and most common treatment-emergent adverse events
(!5%).

Adverse event Duloxetine Escitalopram Overall P-valuea


(N ¼ 244) (N ¼ 239) (N ¼ 483)

n (%) n (%) n (%)


SAEsb 3 (1.2) 3 (1.3) 6 (1.2) N/A
Deaths 0 (0) 1 (0.4) 1 (0.2) N/A
Discontinuations due 10 (4.1) 13 (5.4) 23 (4.8) 0.528
to an adverse event
Discontinuations due 5 (2.0) 3 (1.3) 8 (1.7) 0.724
to lack of efficacy
!1 TEAE 132 (54.1) 100 (41.8) 232 (48.0) 0.008
Nausea 26 (10.7) 28 (11.7) 54 (11.2) N/Ac
Headache 16 (6.6) 20 (8.4) 36 (7.5) N/Ac
Constipation 23 (9.4) 9 (3.8) 32 (6.6) N/Ac
Insomnia 17 (7.0) 11 (4.6) 28 (5.8) N/Ac

N/A ¼ not applicable; SAE ¼ serious adverse event; TEAE ¼ treatment-emergent


adverse event.
a
P-values from Fisher’s exact test.
b
3 patients in the duloxetine treatment group experienced 3 SAEs (depression,
cholelithiasis, and asthma) and 3 patients in the escitalopram treatment group
experienced 4 SAEs (suicide attempt, insomnia, melaena, and syncope).
c
Fig. 2. Change in AES-C total score in the duloxetine and escitalopram treatment Statistical significance for individual TEAEs are not reported because of the
groups over the 8-week treatment period. AES-C ¼ Apathy Evaluation Scale e Clinician issues associated with multiplicity and the suitability of the sample size for such
rated version; CI ¼ confidence interval; LS ¼ least squares. comparisons.
672 J. Raskin et al. / Journal of Psychiatric Research 46 (2012) 667e674

4. Discussion population was more depressed than expected (mean MADRS


total score 10.4 [SD 3.59]). Other evidence that the patients may
To our knowledge, this is the largest controlled study designed have been more depressed at study entry was the relapse rate
to evaluate apathy outcomes in patients with MDD. However, (11.5% for the overall study population), which was higher than
several limitations must be considered. The results of this study are expected for a stable population over 8 weeks. The relapses were
only applicable to patients who were treated with a SSRI for MDD spread out over the study period, which suggests that the relapse
for 3 or more months and continued to experience apathy in the rate was not related to a dosing issue; if patients were switched to
absence of syndromic depression. The current study does not too low a dose of antidepressant, the relapses would be expected
address if apathy is a residual symptom from the initial MDD or to occur in the first week or two of treatment. However, the
a treatment-emergent symptom of SSRIs. Further, the study was changing doses of SSRI that patients received before and during
designed to allow patients to enter the trial from a SSRI at any the study may also have had an effect on apathy. For example,
approved dose. This approach is important for generalizability and a patient with apathy on a high-dose SSRI before the study may
feasibility, but the variability in treatment may have contributed to have had an improvement in apathy on receiving a lower dose in
the inability to find a difference between groups. However, post hoc the study (Hoehn-Saric et al., 1990; Garland and Baerg, 2001;
sensitivity analyses did not suggest that one or another SSRI Barnhart et al., 2004). Conversely, a patient on a low-dose SSRI
subgroup had a markedly different response than the group as before the study may have received a higher dose in the study,
a whole (see Supplementary Table). Of note, improvements in which could have improved the patient’s depression and, as
apathy were observed in patients in the escitalopram to escitalo- a result, improved their apathy. Analysis of the change in AES-C
pram subgroup, who had not switched antidepressant. These total score by SSRI dose before the study suggested that there
patients had received escitalopram before the study and were was an improvement in apathy in both the group of patients who
randomized to remain on escitalopram during the study. Apathy had been on a higher than minimum SSRI dose before the study
improvements in this subgroup suggest that nonspecific effects of and the group of patients who had been on the minimum SSRI
time and participation in a study were relevant factors for both dose. In total, 35.4% of patients entered the study on higher doses
groups. of SSRIs and a similar proportion (37.3%) had their antidepressant
The SNRI (duloxetine) treatment group showed improvements dose optimized during the study.
in apathy during the study, as shown by statistically significant and There have been few assessments of apathy outcomes in
clinically significant within-group improvements in the AES-C total patients with MDD to date. The suggestion that apathy may be
score and MADRS Item 8. However, there was a similar degree of a treatment-emergent effect of SSRI treatment arose from reports
improvement in patients in the escitalopram treatment group, who of apathy in children, adolescents, and adults receiving a SSRI for
had not switched class of antidepressant (i.e., continued to receive various conditions, including depression (Hoehn-Saric et al., 1990,
treatment with a SSRI during the study). Although this was 1991; Garland and Baerg, 2001; Barnhart et al., 2004). The apathy
a randomized controlled study, the lack of a placebo arm prevents occurred after weeks or months of otherwise effective treatment
a full assessment of the effect of participation in the trial on and could be alleviated by reducing the dose of SSRI or stopping
treatment outcomes, although the escitalopram to escitalopram treatment. Preliminary studies supported this proposed effect of
subgroup was included to allow for some interpretation. Partici- SSRI treatment and suggested that switching from a SSRI to a dual
pation in the trial may have improved outcomes in the following reuptake inhibitor may improve apathy symptoms. A retrospective
ways: (i) patients knew they were receiving an active treatment analysis of antidepressant tachyphylaxis across antidepressant
and that its effect on apathy was being assessed, and this may have classes found lower rates in patients receiving dual reuptake
resulted in higher expectations of therapy; (ii) participation may inhibitors (venlafaxine and tricyclic antidepressants) compared
have resulted in improved treatment adherence; and (iii) partici- with those receiving SSRIs (Posternak and Zimmerman, 2005). A
pation required actions that may have counteracted apathy to some case control study found that elderly depressed patients treated
extent, for example, leaving the house for study visits and engaging with SSRIs may have more apathy symptoms at discharge
with new individuals. As patients were not asked to guess treat- compared with those treated with non-SSRI antidepressants
ment assignment, the extent of blinding cannot be fully assessed. (Wongpakaran et al., 2007). In addition, a study conducted by
There may also have been a nonspecific effect of time if the duration Marangell et al. (2002) found a significant reduction in apathy
of treatment before study entry had not been sufficient to treat the symptoms after the addition of olanzapine to SSRI treatment in
patient’s depression fully. In this case, continuing antidepressant patients with MDD who had achieved remission. The rationale for
treatment, regardless of the class of drug, may have been sufficient adding olanzapine to SSRI treatment was based on the suggestion
to improve depression and, consequently, apathy. Although that increased levels of dopamine may improve apathy, and olan-
patients were nonsyndromic for MDD, a mean MADRS total score of zapine may reverse the inhibition of dopamine release in the
10.4 at baseline suggests many patients still had moderate MDD frontal lobe that arises from chronic serotonergic stimulation by
symptoms. Moreover, the numerically higher relapse rate in the SSRIs. The current study was intended to address the lack of large
escitalopram to escitalopram subgroup compared with the primary controlled studies comparing antidepressants in treating residual
treatment groups implies that patients’ symptoms of depression or antidepressant-induced apathy. While there were no statistically
may not have been under good control in this subgroup, despite significant differences between duloxetine and escitalopram on
being treated for at least 3 months. Given the above factors, apathy and while the conclusions that can be drawn from the study
a randomized treatment period longer than 8 weeks may have are limited, this study (i) suggests that improving depressive
allowed differences between the two antidepressants, if any, to be symptoms can improve symptoms of apathy; (ii) provides infor-
revealed. mation on apathy symptoms in a large group of patients with MDD
The requirement for patients to have received treatment with receiving antidepressant treatment; and (iii) provides valuable data
a SSRI for at least 3 months before the study was intended to for the design of future studies of apathy in patients with MDD.
minimize symptoms of depression and allow for the appearance An issue to consider for future studies investigating apathy is the
of SSRI treatment-emergent effects. As there was no prospective choice of scale. There are few scales for assessing apathy and those
run-in to the study, it is not possible to state the cause of apathy that are available are not well known to physicians. The AES treats
with certainty. The baseline assessments suggest that the study apathy as a psychological dimension that may be evaluated in
J. Raskin et al. / Journal of Psychiatric Research 46 (2012) 667e674 673

patients whose apathy characterizes their overall clinical state as Role of contributors
well as in those in whom apathy is a symptom of another
syndrome, such as depression (Marin et al., 1991). The clinician JR, REG, and LM were involved in study design, NH was involved
rated version of the AES (AES-C) was chosen as the primary means in data collection, GWZ was involved in the statistical analysis, and
of assessing apathy in this study. Also used were the RSAT TG was a Principal Investigator. All authors participated in the
(Rothschild, 2008), which was designed to assess symptoms of interpretation of the study results, and in the drafting, critical
antidepressant tachyphylaxis in patients who do not meet the revision, and approval of the final version of the manuscript.
criteria for a relapse or recurrence of an episode of major depres-
sion and includes “motivation and interest” and “energy level” Conflicts of interest
items, and the MADRS Item 8 (Montgomery and Åsberg, 1979),
which assesses the patient’s inability to feel. Significant within- JR, REG, GWZ, and LM are full-time employees of, and NH is
group improvements in apathy symptoms were observed in both contractually employed by, Eli Lilly and Company. JR and LM own
treatment groups on each of these scales. stock in Eli Lilly and Company. TG has no conflicts of interest to
There were few differences in safety and tolerability observed disclose.
between the duloxetine and escitalopram treatment groups;
however, the study was not powered for the safety endpoints. More
Acknowledgments
patients reported one or more TEAE in the duloxetine treatment
group than the escitalopram treatment group (132 vs 100) over the
The authors acknowledge Richard Walton (Eli Lilly) for his
8-week treatment period. However, patients switching from a SSRI
assistance in reviewing the manuscript. The authors would also like
to another SSRI would be expected to have fewer side effects than
to thank the investigators, their staff, and their patients for their
those switching to another class (SNRI). In addition, approximately
participation in this study.
25% of patients in the escitalopram treatment group had already
been receiving treatment with escitalopram before the study and,
therefore, would be expected to experience even fewer adverse Appendix. Supplementary material
events compared with patients switching from other SSRIs. Overall,
duloxetine-treated patients more commonly experienced Supplementary material related to this article can be found
treatment-emergent anxiety or constipation, and experienced online at doi:10.1016/j.jpsychires.2012.02.010
greater increases in heart rate and diastolic blood pressure
compared with escitalopram-treated patients, as expected given References
that duloxetine inhibits the reuptake of norepinephrine (Stahl et al.,
2005). The safety findings in the duloxetine treatment group were Barnhart WJ, Makela EH, Latocha MJ. SSRI-induced apathy syndrome: a clinical
review. Journal of Psychiatric Practice 2004;10:196e9.
similar to those observed in other clinical trials of duloxetine and Birkmayer W, Riederer P. Understanding the neurotransmitters: keys to the work-
are what would be expected of a SNRI (Hudson et al., 2005; Thase ings of the brain. Blau K, trans.. New York, NY: Springer-Verlag Wien; 1989
et al., 2005; Gahimer et al., 2007; Norman and Olver, 2010). Bolling MY, Kohlenberg RJ. Reasons for quitting serotonin reuptake inhibitor
therapy: paradoxical psychological side effects and patient satisfaction.
In conclusion, this study did not detect a difference between Psychotherapy and Psychosomatics 2004;73:380e5.
duloxetine and escitalopram on apathy, depression, or functional Demyttenaere K, Jaspers L. Review: bupropion and SSRI-induced side effects.
outcomes after 8 weeks of treatment in nonsyndromal depressed Journal of Psychopharmacology 2008;22:792e804.
Fava M, Graves LM, Benazzi F, Scalia MJ, Iosifescu DV, Alpert JE, et al. A cross-
patients with residual apathy; there were significant improvements sectional study of the prevalence of cognitive and physical symptoms during
in all outcomes in both treatment groups. Patients in the escitalo- long-term antidepressant treatment. Journal of Clinical Psychiatry 2006;67:
pram to escitalopram subgroup also experienced improvements in 1754e9.
Fava M, Iosifescu DV, Pedrelli P, Baer L. Reliability and validity of the Massachusetts
apathy over the 8-week treatment period. Given the limitations
general hospital cognitive and physical functioning questionnaire. Psycho-
discussed above, this study does not support our hypothesis that therapy and Psychosomatics 2009;78:91e7.
switching from a SSRI to a SNRI alleviates apathy. Duloxetine has Gahimer J, Wernicke J, Yalcin I, Ossanna MJ, Wulster-Radcliffe M, Viktrup L.
A retrospective pooled analysis of duloxetine safety in 23,983 subjects. Current
effects on both serotonin and norepinephrine reuptake. While
Medical Research and Opinion 2007;23:175e84.
speculative, it is possible that a noradrenergic agent without Garland EJ, Baerg EA. A motivational syndrome associated with selective serotonin
concomitant serotonergic effects would be effective i.e., the sero- reuptake inhibitors in children and adolescents. Journal of Child and Adolescent
tonergic component of duloxetine might have been problematic Psychopharmacology 2001;11:181e6.
Gobert A, Rivet JM, Lejeune F, Newman-Tancredi A, Adhumeau-Auclair A, Nicolas JP,
with regard to alleviating apathy. Further study of the natural et al. Serotonin2C receptors tonically suppress the activity of mesocortical
course of apathy in patients with MDD and the relative benefits of dopaminergic and adrenergic, but not serotonergic, pathways: a combined
different treatments is warranted. dialysis and electrophysical analysis in the rat. Synapse 2000;36:205e21.
Gold PW, Chrousos GP. The endocrinology of melancholic and atypical depression:
relation to neurocircuitry and somatic consequences. Proceedings of the
Role of funding source Association of American Physicians 1999;111:22e34.
Guy W. ECDEU assessment manual for psychopharmacology, revised. DHEW
Publication ADM-76-338. Rockville (MD): US Department of Health, Education,
This study (F1J-CR-HMGM) was sponsored by Eli Lilly and and Welfare; 1976.
Company. Eli Lilly was involved in the study design, data collection, Hoehn-Saric R, Lipsey JR, McLeod DR. Apathy and indifference in patients on flu-
data analysis, and preparation of the manuscript. In compliance voxamine and fluoxetine. Journal of Clinical Psychopharmacology 1990;10:
343e5.
with the Uniform Requirements for Manuscripts, established by the Hoehn-Saric R, Harris GJ, Pearlson GD, Cox CS, Machlin SR, Camargo EE.
International Committee of Medical Journal Editors, the sponsor of A fluoxetine-induced frontal lobe syndrome in an obsessive compulsive patient.
this study did not impose any impediment, directly or indirectly, on Journal of Clinical Psychiatry 1991;52:131e3.
Hudson JI, Wohlreich MM, Kajdasz DK, Mallinckrodt CH, Watkin JG, Martynov OV.
the publication of the study’s results. The authors acknowledge the
Safety and tolerability of duloxetine in the treatment of major depressive
independent medical writing assistance provided by Justine disorder: analysis of pooled data from eight placebo-controlled clinical trials.
Southby, PhD, and Serina Stretton, PhD, of ProScribe Medical Human Psychopharmacology 2005;20:327e41.
Communications, funded from an unrestricted financial grant from Marangell LB, Johnson CR, Kertz B, Zboyan HA, Martinez JM. Olanzapine in the
treatment of apathy in previously depressed participants maintained with
Eli Lilly Australia. ProScribe’s services complied with international selective serotonin reuptake inhibitors: an open-label, flexible dose study.
guidelines for Good Publication Practice (GPP2). Journal of Clinical Psychiatry 2002;63:391e5.
674 J. Raskin et al. / Journal of Psychiatric Research 46 (2012) 667e674

Marin RS, Biedrzycki RC, Firinciogullari S. Reliability and validity of the apathy Rothschild AJ. The Rothschild Scale for Antidepressant Tachyphylaxis: reliability and
evaluation scale. Psychiatry Research 1991;38:143e62. validity. Comprehensive Psychiatry 2008;49:508e13.
McClintock SM, Husain MM, Wisniewski SR, Nierenberg AA, Stewart JW, Segal M. Serotonergic innervation of the locus coeruleus from the dorsal raphe and its
Trivedi MH, et al. Residual symptoms in depressed outpatients who respond by action on responses to noxious stimuli. Journal of Physiology 1979;286:401e15.
50% but do not remit to antidepressant medication. Journal of Clinical Sheehan DV, Lecrubier Y, Sheehan KH, Amorim P, Janavs J, Weiller E, et al. The Mini-
Psychopharmacology 2011;31:180e6. International Neuropsychiatric Interview (M.I.N.I.): the development and vali-
Montgomery SA, Åsberg M. A new depression scale designed to be sensitive to dation of a structured diagnostic psychiatric interview for DSM-IV and ICD-10.
change. British Journal of Psychiatry 1979;134:382e9. Journal of Clinical Psychiatry 1998;59(Suppl. 20):22e33.
Norman TR, Olver JS. Continuation treatment of major depressive disorder: is there Stahl SM, Grady MM, Moret C, Briley M. SNRIs: their pharmacology, clinical efficacy,
a case for duloxetine? Journal of Drug Design, Development and Therapy 2010; and tolerability in comparison with other classes of antidepressants. CNS
4:19e31. Spectrums 2005;10:732e47.
Paykel ES, Ramana R, Cooper Z, Hayhurst H, Kerr J, Barocka A. Residual symptoms Thase ME, Tran PV, Wiltse C, Pangallo BA, Mallinckrodt C, Detke MJ. Cardiovascular
after partial remission: an important outcome in depression. Psychological profile of duloxetine, a dual reuptake inhibitor of serotonin and norepinephrine.
Medicine 1995;25:1171e80. Journal of Clinical Psychopharmacology 2005;25:132e40.
Posternak MA, Zimmerman M. Dual reuptake inhibitors incur lower rates of Wongpakaran N, van Reekum R, Wongpakaran T, Clarke D. Selective serotonin
tachyphylaxis than selective serotonin reuptake inhibitors: a retrospective reuptake inhibitor use associates with apathy among depressed elderly: a case-
study. Journal of Clinical Psychiatry 2005;66:705e7. control study. Annals of General Psychiatry 2007;6:7.

You might also like