1 s2.0 S0165032716303871 Main
1 s2.0 S0165032716303871 Main
1 s2.0 S0165032716303871 Main
Research paper
art ic l e i nf o a b s t r a c t
Article history: Background: The efficacy, safety, and tolerability of lisdexamfetamine dimesylate (LDX) augmentation of
Received 9 March 2016 antidepressant monotherapy in adults with major depressive disorder (MDD) from two phase 3 studies
Received in revised form are reported.
14 June 2016
Methods: Across study 1 (placebo, n ¼201; LDX, n ¼201) and study 2 (placebo, n¼ 213; LDX, n ¼211),
Accepted 3 July 2016
Available online 5 July 2016
most participants (placebo and LDX) in the safety analysis set were female (study 1: 66.2% and 64.2%;
study 2: 67.1% and 66.8%); mean7 SD ages were 41.87 12.04 with placebo and 42.2 712.32 with LDX in
Keywords: study 1 and 42.6 711.41 with placebo and 42.0 7 11.63 with LDX in study 2. Participants (18–65 y) had
Augmentation DSM-IV-TR–diagnosed MDD and lead-in baseline Montgomery-Åsberg Depression Rating Scale (MADRS)
Lisdexamfetamine dimesylate total scores Z24. Eight-week antidepressant lead-in phases prospectively assessed antidepressant re-
Major depressive disorder
sponse. Then, 8 weeks of randomized (1:1), double-blind treatment with dose-optimized LDX (20–
Selective serotonin reuptake inhibitors
70 mg) or placebo in participants exhibiting inadequate antidepressant monotherapy responses (aug-
Serotonin-norepinephrine reuptake in-
hibitors
mentation baseline MADRS total scores Z18 and o 50% MADRS total score reductions from lead-in
Amphetamine baseline to augmentation baseline) was initiated. The primary endpoint was MADRS total score change
from augmentation baseline to week 16. Safety and tolerability measures included the occurrence of
treatment-emergent adverse events (TEAEs).
Results: Least squares mean (95% CI) treatment differences (LDX–placebo) for MADRS total score changes
from augmentation baseline to week 16 were not statistically significant in study 1 (0.1 [–1.7, 2.0],
P ¼0.883) or study 2 (–0.5 [–2.3, 1.3], P ¼0.583). The only TEAE reported by 45% of LDX participants at
twice the placebo rate in both studies was dry mouth.
Limitations: Limitations include the exclusion of participants with psychiatric comorbidities/active
medical disorders, the inability to assess specific MDD symptom domains (eg, anhedonia, cognition) or
subtypes, the use of telephone-based depression assessments, and the potential influence of placebo
response.
Conclusion: Contrary to expectations, LDX augmentation was not superior to placebo in reducing
Abbreviations: ABAC-A, Abbreviated Brief Assessment of Cognition in Affective Disorders; ACSA, Amphetamine Cessation Symptom Assessment; ADHD, attention-deficit/
hyperactivity disorder; BMI, body mass index; CGI-I, Clinical Global Impressions–Improvement; CMH, Cochran-Mantel-Haenszel; C-SSRS, Columbia-Suicide Severity Rating
Scale; DBP, diastolic blood pressure; ECG, electrocardiogram; EOS, end of study; FAS, full analysis set; LDX, lisdexamfetamine dimesylate; MADRS, Montgomery-Åsberg
Depression Rating Scale; MAF-GFI, global fatigue index of the Multidimensional Assessment of Fatigue; MDD, major depressive disorder; MMRM, mixed-effects model for
repeated measures; QIDS-SR, Quick Inventory of Depressive Symptomatology–Self-Report; QTcF, Fridericia's corrected QTC interval; SCID-CT, Structured Clinical Interview for
Diagnostic and Statistical Manual for Mental Disorders, Fourth Edition–Text Revision, Clinical Trial Version; SBP, systolic blood pressure; SDS, Sheehan Disability Scale; SNRIs,
serotonin-norepinephrine reuptake inhibitors; SSRIs, selective serotonin reuptake inhibitors; STAR*D, Sequenced Treatment Alternatives to Relieve Depression; TEAEs,
treatment-emergent adverse events
n
Correspondence to: Shire, 300 Shire Way, Lexington, MA 02421.
E-mail address: [email protected] (M. Madhoo).
http://dx.doi.org/10.1016/j.jad.2016.07.006
0165-0327/& 2016 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
152 C. Richards et al. / Journal of Affective Disorders 206 (2016) 151–160
1. Introduction 2. Methods
Currently approved treatments for major depressive disorder 2.1. Study design and treatment regimens
(MDD), including selective serotonin reuptake inhibitors (SSRIs)
and serotonin-norepinephrine reuptake inhibitors (SNRIs), effec- Study 1 (ClinicalTrials.gov identifier: NCT01436149) was con-
tively reduce MDD symptoms (Arroll et al., 2005; Cipriani et al., ducted at 76 sites across Canada, Croatia, Mexico, Spain, and the
2009). Nevertheless, more than half of patients with MDD do not United States. Study 2 (ClinicalTrials.gov identifier: NCT01436162)
experience full remission with antidepressant monotherapy or was conducted at 94 sites across the Czech Republic, Estonia,
Finland, Germany, Hungary, Poland, Romania, South Africa, Swe-
with augmentation therapy (Rush et al., 2006; Trivedi et al., 2006a,
den, and the United States. Both studies were conducted in ac-
2006b). For example, in the Sequenced Treatment Alternatives to
cordance with guidelines from the International Conference on
Relieve Depression (STAR*D) study, low remission rates (27.5%
Harmonization Good Clinical Practice and the principles of the
based on the 17-item Hamilton Depression Rating Scale, 32.9%
Declaration of Helsinki. All study protocols were approved by
based on the 16-item Quick Inventory of Depressive Symptoma- ethics committees and regulatory agencies before initiating the
tology–Self-Report [QIDS-SR]) were observed with citalopram studies, and written informed consent was obtained before per-
monotherapy (Trivedi et al., 2006b). In addition, only 29.7% to forming study-related procedures.
39.0% of individuals receiving second-step augmentation with Each study employed a multicenter, randomized, double-blind,
sustained-release bupropion or buspirone achieved remission parallel-group, placebo-controlled, flexible-dose design. With the
(Trivedi et al., 2006a). exception of differences in some of the secondary endpoints, the
Despite the relatively high partial remission rates following studies were identically designed and consisted of 4 phases
antidepressant therapy, there are few medications approved by (Fig. 1): a screening period of 1–4 weeks, an 8-week anti-
the US Food and Drug Administration for use as augmentation depressant lead-in phase (to prospectively confirm inadequate
agents in MDD. Psychostimulants have been investigated for use as response to antidepressant monotherapy), an 8-week double-
augmentation agents in MDD since the 1950s (Robin and Wise- blind treatment phase (3 weeks of dose optimization followed by
berg, 1958). However, in 2 meta-analyses (Candy et al., 2008; 5 weeks of dose maintenance), and a 7- to 9-day follow-up period.
Fleurence et al., 2009), mixed results were reported with respect After screening, eligible participants entered the 8-week sin-
to the efficacy of psychostimulants or eugorics (modafinil) as gle-blind antidepressant lead-in phase, which prospectively as-
sessed response to antidepressant monotherapy and blinded par-
augmentation therapy in MDD.
ticipants to the time of randomization. During the antidepressant
Lisdexamfetamine dimesylate (LDX) is approved in the United
lead-in phase, participants were assigned by investigators to 1 of
States and in other countries for use in patients 6 years and older
4 commercially available antidepressant medications: an SSRI
with attention-deficit/hyperactivity disorder (ADHD) and for use
(escitalopram oxalate [10 or 20 mg] or sertraline hydrochloride
in adults with moderate to severe binge eating disorder only in the
[50–200 mg]) or an SNRI (venlafaxine hydrochloride extended
United States Vyvanse® [lisdexamfetamine dimesylate] 2015. The release [37.5–375 mg] or duloxetine hydrochloride [30–120 mg]).
efficacy of LDX in MDD has been investigated in small, phase 2, Antidepressant assignment was unblinded to both investigators
randomized, double-blind, placebo-controlled studies in in- and participants and was based on clinical factors, including prior
dividuals with MDD who exhibited inadequate response to anti- antidepressant use, response, and tolerability. Each individual in-
depressant monotherapy (Madhoo et al., 2014; Trivedi et al., 2013). vestigator was asked to distribute antidepressant treatments
In one study, LDX augmentation of escitalopram met signal-de- equally at their site, attempting to not assign any 1 antidepressant
tection criteria (prespecified 2-sided significance level α¼0.10) for to 440% of participants. Investigators managed antidepressant
significant reduction in mean Montgomery-Åsberg Depression distribution at the site level and the study sponsor provided on-
Rating Scale (MADRS) total score versus placebo in escitalopram going internal review of the overall distribution. No action from
nonremitters (Trivedi et al., 2013). In a separate study that focused the sponsor was taken to ensure that the protocol-stated anti-
on treating executive dysfunction in individuals with fully or depressant distribution was attained. The initial antidepressant
partially remitted MDD (defined as MADRS total scores r18), LDX dose was taken after the lead-in baseline visit (week 0), with
augmentation of SSRI monotherapy produced significantly greater treatment titrated to the maximum tolerated approved dose by
week 4; dose adjustments were not permitted after week 4.
reductions in MADRS total score than placebo (Madhoo et al.,
Weekly dose increases were made by the investigator according to
2014).
product label guidelines. Antidepressant strength and doses varied
This report presents the findings of two phase 3 studies de-
based on commercial availability and country and were adminis-
signed to assess the efficacy, safety, and tolerability of LDX aug-
tered according to local guidelines. All participants also received
mentation in adults with MDD who exhibit inadequate response
LDX-matched placebo during this phase.
to an 8-week prospective course of antidepressant therapy. The Following the lead-in phase, participants exhibiting an in-
primary objective of each study was to assess the efficacy of LDX adequate response to antidepressant monotherapy entered the
augmentation, as measured by change in MADRS total score. double-blind treatment phase and were randomized (1:1) to LDX
Secondary objectives included assessment of LDX augmentation or placebo. Randomization criteria included having a MADRS total
effects on the Sheehan Disability Scale (SDS; the key secondary score of Z18 at augmentation baseline (week 8), having a MADRS
endpoint), on other secondary efficacy endpoints, and on safety total score reduction of o50% from lead-in baseline to augmen-
and tolerability. tation baseline, exhibiting improvement in depressive symptoms
C. Richards et al. / Journal of Affective Disorders 206 (2016) 151–160 153
Fig. 1. Study designs for randomized participants (A) and non-randomized participants (B). LDX ¼ lisdexamfetamine; V ¼ visit; Wk¼ week.
Fig. 2. Participant disposition. DBP¼ diastolic blood pressure; ECG ¼ electrocardiogram; LDX¼ lisdexamfetamine dimesylate; MADRS ¼ Montgomery-Åsberg Depression
Rating Scale; SBP ¼systolic blood pressure. *Participants could have had multiple reasons for not being randomized to double-blind treatment. †Sustained elevations in
average (of 3 readings) sitting SBP (an increase of Z 10 mmHg from lead-in baseline and an average value Z 140 mmHg on 2 consecutive visits), or sitting DBP (an increase
of Z 10 mmHg from lead-in baseline and an average value Z 90 mmHg on 2 consecutive visits), or pulse rate (an increase of Z 20 bpm from lead-in baseline and an average
value Z100 bpm on 2 consecutive visits).
administration of the first randomized treatment dose (excluding 96.2% (205/213), LDX: 98.1% (207/211)]) had compliance rates
the assigned background antidepressant in the current studies). (defined as [total number of capsules taken 100]/total planned
Prior MDD medication use was reported by 57.0% (229/402) and days of dosing) in the range of 80% to 120%.
64.2% (272/424) of participants in studies 1 and 2, respectively.
The most frequently used MDD medications (Z5% of participants) 3.4. Efficacy
were escitalopram (16.4% [66/402]), sertraline (13.2% [53/402]),
duloxetine (12.4% [50/402]), fluoxetine (11.7% [47/402]), citalo- 3.4.1. Primary endpoint – change in MADRS total score
pram (11.4% [46/402]), bupropion (10.9% [44/402]), paroxetine In both studies, least squares (LS) mean MADRS total score
(9.0% [36/402]), and venlafaxine (7.7% [31/402]) in study 1 and decreases from augmentation baseline to week 16 were observed
citalopram (18.9% [80/424]), sertraline (18.6% [79/424]), venlafax- with placebo and LDX (Fig. 3). As reported in Table 2, the between-
ine (17.7% [75/424]), escitalopram (16.0% [68/424]), fluoxetine treatment difference (LDX – placebo) for mean change from aug-
(11.6% [49/424]), bupropion (8.0% [34/424]), paroxetine (8.0% [34/ mentation baseline to week 16 in MADRS total score in the entire
424]), duloxetine (7.8% [33/424]), and any investigational drug FAS was not statistically significant in either study (both P4 0.05).
(5.9% [25/424]) in study 2. Findings were similar when assessed by sex or antidepressant
class (Supplemental Fig. 1).
3.3. Extent of exposure and treatment compliance
3.4.2. Secondary efficacy endpoints
During double-blind treatment, mean 7SD exposure days for Findings for the SDS (key secondary endpoint) and other sec-
placebo and LDX, respectively, were 49.5 7 15.54 and 50.1 714.76 ondary efficacy endpoints are presented in Table 2. In both studies,
in study 1 and 52.5 712.33 and 51.57 13.17 in study 2. During the there were no statistically significant or clinically meaningful
double-blind treatment periods in studies 1 and 2, respectively, treatment differences for the change from augmentation baseline
74.1% (149/201) and 62.6% (132/211) of participants had their LDX to week 16 between placebo and LDX for SDS total score (Table 2)
dose increased to 50 mg and 39.3% (79/201) and 33.6% (71/211) or for the individual SDS domain scores (data not shown). Across
had their dose increased to 70 mg; 6.0% (12/201) and 9.5% (20/211) most of the other secondary efficacy endpoints (dichotomized CGI-
had their dose down-titrated to 20 mg. The mean 7SD daily LDX I, QIDS-SR, ABAC-A, MADRS remission rate, MADRS 50% response
dose during double-blind treatment was 46.5 713.74 mg in study rate) similar findings were observed, with few nominally statisti-
1 and 43.4 714.35 mg in study 2. Most participants (study 1 cally significant treatment effects being observed between placebo
[placebo: 88.1% (177/201), LDX: 91.5% (184/201)]; study 2 [placebo: and LDX at week 16/EOS (Table 2). However, because the primary
156 C. Richards et al. / Journal of Affective Disorders 206 (2016) 151–160
Table 1
Demographic and baseline clinical characteristics, safety analysis set.
Study 1 Study 2
Placebo (n¼ 201) LDX (n¼ 201) Placebo (n ¼213) LDX (n¼ 211)
Mean 7 SD age, years 41.8 712.04 42.2 7 12.32 42.6 7 11.41 42.0 7 11.63
Sex, n (%)
Male 68 (33.8) 72 (35.8) 70 (32.9) 70 (33.2)
Female 133 (66.2) 129 (64.2) 143 (67.1) 141 (66.8)
Race, n (%)
White 160 (79.6) 154 (76.6) 183 (85.9) 174 (82.5)
Black/African American 36 (17.9) 39 (19.4) 24 (11.3) 34 (16.1)
Native Hawaiian/Pacific Islander 0 0 1 (0.5) 1 (0.5)
Asian 5 (2.5) 5 (2.5) 3 (1.4) 1 (0.5)
American Indian/Alaska Native 0 1 (0.5) 0 0
Other 0 2 (1.0) 2 (0.9) 1 (0.5)
Mean 7 SD weight, kg 82.4 717.90 81.3 718.34 80.8 7 18.52 82.2 7 17.66
Mean 7 SD body mass index, kg/m2 28.9 75.43 28.5 7 5.66 28.1 75.47 28.4 7 5.45
Mean 7 SD MADRS total score
Lead-in baseline 33.27 4.63 33.87 5.01 34.1 75.21 33.47 4.39
Augmentation baseline 25.2 75.03 25.4 7 4.75 25.7 7 5.29 26.0 7 5.15
Mean 7 SD SDS scores at augmentation baselinea
Total score 15.6 7 5.65 15.9 7 5.99 16.8 7 5.75 16.8 7 5.34
Disrupted family life score 5.2 72.05 5.2 7 2.19 5.5 7 2.18 5.6 7 1.98
Disrupted social life score 5.6 72.16 5.7 7 2.09 5.8 7 2.14 5.8 7 1.95
Disrupted work/school score 4.7 72.26 5.0 7 2.36 5.5 7 2.23 5.3 7 2.27
Lead-in baseline antidepressant, n (%)
Venlafaxine HCl extended-release (SNRI) 25 (12.4) 21 (10.4) 52 (24.4) 48 (22.7)
Duloxetine HCl (SNRI) 62 (30.8) 65 (32.3) 46 (21.6) 48 (22.7)
Escitalopram oxalate (SSRI) 67 (33.3) 80 (39.8) 66 (31.0) 63 (29.9)
Sertraline HCl (SSRI) 47 (23.4) 35 (17.4) 49 (23.0) 52 (24.6)
Final antidepressant dose, n (%)
Venlafaxine HCl extended-release (SNRI)
37.5 mg 1 (0.5) 0 – –
75 mg 0 1 (0.5) 7 (3.3) 7 (3.3)
150 mg 3 (1.5) 4 (2.0) 18 (8.5) 17 (8.1)
225 mg 21 (10.4) 16 (8.0) 22 (10.3) 20 (9.5)
300 mg – – 3 (1.4) 3 (1.4)
375 mg – – 2 (0.9) 1 (0.5)
Duloxetine HCl (SNRI)
30 mg 1 (0.5) 2 (1.0) 0 2 (0.9)
40 mg 5 (2.5) 5 (2.5) 1 (0.5) 1 (0.5)
60 mg 48 (23.9) 44 (21.9) 31 (14.6) 37 (17.5)
90 mg 3 (1.5) 6 (3.0) 3 (1.4) 2 (0.9)
120 mg 5 (2.5) 8 (4.0) 11 (5.2) 6 (2.8)
Escitalopram oxalate (SSRI)
10 mg 7 (3.5) 13 (6.5) 12 (5.6) 11 (5.2)
20 mg 60 (29.9) 67 (33.3) 54 (25.4) 52 (24.6)
Sertraline HCl (SSRI)
50 mg 3 (1.5) 2 (1.0) 4 (1.9) 8 (3.8)
100 mg 8 (4.0) 7 (3.5) 13 (6.1) 18 (8.5)
150 mg 14 (7.0) 12 (6.0) 4 (1.9) 15 (7.1)
200 mg 22 (10.9) 14 (7.0) 28 (13.1) 11 (5.2)
LDX¼ lisdexamfetamine dimesylate; MADRS ¼ Montgomery-Åsberg Depression Rating Scale; SDS ¼Sheehan Disability Scale; SNRI ¼ serotonin-norepinephrine reuptake in-
hibitor; SSRI¼ selective serotonin reuptake inhibitor.
a
Based on Full Analysis set: study 1 (total score [placebo, n ¼198; LDX, n ¼198], disrupted family life score [placebo, n ¼199; LDX, n ¼199], disrupted social life score
[placebo, n ¼ 199; LDX, n ¼ 198], disrupted work/school score [placebo, n ¼ 198; LDX, n¼ 199]); study 2 (placebo, n ¼213; LDX, n ¼ 209).
endpoint was not statistically significant, any nominal differences 1 (syncope in an LDX participant; suicide attempt in a placebo
in secondary endpoints should not be considered statistically participant; both resolved) and 2 cases in study 2 (anxiety in an
significant. LDX participant; suicidal ideation in a placebo participant). The
most frequently occurring TEAEs are summarized in Table 3. The
3.5. Safety and tolerability TEAEs reported by Z5% of participants treated with LDX and at
twice the rate of placebo were dry mouth and nasopharyngitis in
3.5.1. Adverse events study 1 and dry mouth and decreased appetite in study 2. There
Treatment-emergent AE (TEAEs) are summarized in Table 3. In were no instances of psychosis/mania or aggression reported as
both studies, higher percentages of TEAEs were reported with LDX TEAEs or deaths in either study.
than with placebo. Most TEAEs were mild or moderate in in- Few serious TEAEs were reported in either study (Table 3). In
tensity; the frequency of severe TEAEs was low in both studies (see study 1, there were 3 serious TEAEs in the LDX group (nonsuicidal
footnote to Table 3 for a complete listing). TEAEs leading to dis- overdose of diphenhydramine, syncope, and appendicitis; none
continuation were generally of mild to moderate intensity in both were considered treatment related by the investigator) and 7 ser-
studies (see footnote to Table 3 for a complete listing of TEAEs ious TEAEs in the placebo group (suicide attempt, major depres-
leading to discontinuation), with the exception of 2 cases in study sion, vertigo, syncope, appendectomy, laceration, and skin
C. Richards et al. / Journal of Affective Disorders 206 (2016) 151–160 157
4. Discussion
Table 2
Efficacy assessments at end of double-blind treatment, full analysis set.
Study 1 Study 2
ABAC-A ¼ Abbreviated Brief Assessment of Cognition in Affective Disorders; CGI-I ¼Clinical Global Impressions-Improvement; EOS ¼ end of study; MADRS ¼ Montgomery-
Åsberg Depression Rating Scale; MAF-GFI=global fatigue index of the Multidimensional Assessment of Fatigue; QIDS-SR=Quick Inventory of Depressive Symptomatology–
Self-Report; SDS ¼ Sheehan Disability Scale.
a
MADRS sample sizes: augmentation baseline (study 1 [placebo (n ¼200), LDX (n¼ 200)]; study 2 [placebo (n¼ 213), LDX (n ¼ 209)]); week 16, LS mean change from
augmentation baseline, and LS mean treatment difference at week 16 (study 1 [placebo (n ¼165), LDX (n ¼ 163)]; study 2 [placebo (n ¼189), LDX (n ¼181)]).
b
SDS sample sizes: augmentation baseline (study 1 [placebo (n¼ 198), LDX (n ¼198)]; study 2 [placebo (n ¼ 213), LDX (n ¼209)]); week 16 (study 1 [placebo (n¼ 165),
LDX (n ¼163)]; study 2 [placebo (n¼ 189), LDX (n ¼ 180)]); LS mean change from augmentation baseline and LS mean treatment difference at week 16 (study 1 [placebo
(n¼ 164), LDX (n ¼ 162)]), study 2 (placebo [n¼ 189], LDX [n ¼180]).
c
MAF-GFI sample sizes: augmentation baseline (study 2 [placebo (n¼ 212), LDX (n ¼209)]); week 16, LS mean change from augmentation baseline and LS mean
treatment difference at week 16 (study 2 [placebo (n ¼205), LDX (n¼ 197)]).
d
QIDS-SR sample sizes: augmentation baseline (study 1 [placebo (n¼ 197), LDX (n¼ 199)]); week 16 (study 1 [placebo (n ¼ 189), LDX (n¼ 186)]); LS mean change from
augmentation baseline and LS mean treatment difference at week 16 (study 1 [placebo (n ¼186), LDX (n ¼185)]).
e
ABAC-A sample sizes: augmentation baseline (study 2 [placebo (n ¼213), LDX (n ¼209)]); week 16, LS mean change from augmentation baseline and LS mean
treatment difference at week 16 (study 2 [placebo (n ¼198), LDX (n ¼ 194)]).
was related to the use of the MADRS seems unlikely because of the are needed to understand the nuanced effects of psychostimulants
consistent lack of effect observed across all study assessments, and to further delineate the biological differences between
including the QIDS-SR. In support of the latter hypothesis, me- symptoms of apathy and mood enhancement in individuals with
thylphenidate has been reported to be efficacious in reducing MDD.
apathy in individuals with Alzheimer's disease (Rosenberg et al., The overall safety and tolerability findings observed in these
2013) and apathy and fatigue in individuals with MDD (Ravindran studies are consistent with previously published studies of LDX
et al., 2008). Additionally, a previously published study reported augmentation for MDD with other clinical studies of LDX for which
LDX augmentation significantly improved self-reported and in- LDX is approved for use (Adler et al., 2008; Madhoo et al., 2014;
formant-reported executive dysfunction, as measured by the Be- McElroy et al., 2016; Trivedi et al., 2013 Wigal et al., 2010). In-
havior Rating Inventory of Executive Function-Adult Version, in somnia, dry mouth, headache, decreased appetite, and nasophar-
participants with mild MDD (Madhoo et al., 2014). However, given yngitis were among the most frequently reported TEAEs with LDX.
the lack of effect of LDX on the ABAC-A in the current studies, this LDX was associated with greater mean increases in pulse rate and
finding also needs to be cautiously considered. Nevertheless, the blood pressure than placebo and with decreases in weight and
possibility that psychostimulants may effectively reduce apathy, BMI.
inattention, or indifference (symptoms that may not be effectively These findings should be considered in light of several limita-
captured by the MADRS or ABAC-A) remains. Additional studies tions. First, these studies used remote telephone assessment of the
C. Richards et al. / Journal of Affective Disorders 206 (2016) 151–160 159
Table 3
Summary of safety and tolerability during double-blind treatment, safety analysis set.
Study 1 Study 2
Placebo (n¼ 201) LDX (n¼ 201) Placebo (n ¼213) LDX (n¼ 211)
Any TEAE, n (%) 118 (58.7) 131 (65.2) Any TEAE, n (%) 108 (50.7) 139 (65.9)
Serious TEAEs 5 (2.5) 3 (1.5) Serious TEAEs 1 (0.5) 1 (0.5)
Severe TEAEsa 6 (3.0) 4 (2.0) Severe TEAEs a
3 (1.4) 7 (3.3)
Treatment-related TEAEs 56 (27.9) 75 (37.3) Treatment-related TEAEs 43 (20.2) 85 (40.3)
TEAEs leading to discontinuationb 7 (3.5) 8 (4.0) TEAEs leading to 1 (0.5) 2 (0.9)
discontinuationb
TEAEs reported by 45% of participants in either treatment group TEAEs reported by 45% of participants in either treatment group
Insomnia 15 (7.5) 19 (9.5) Headache 16 (7.5) 25 (11.8)
Dry mouth 6 (3.0) 19 (9.5) Dry mouth 6 (2.8) 25 (11.8)
Decreased appetite 8 (4.0) 15 (7.5) Nasopharyngitis 19 (8.9) 14 (6.6)
Headache 21 (10.4) 13 (6.5) Decreased appetite 5 (2.3) 13 (6.2)
Nausea 10 (5.0) 13 (6.5) Insomnia 7 (3.3) 11 (5.2)
Nasopharyngitis 4 (2.0) 11 (5.5) Hyperhidrosis 1 (0.5) 11 (5.2)
Dizziness 10 (5.0) 9 (4.5)
Changes (mean 7 SD) from augmentation baseline at week 16/EOS Changes (mean 7 SD) from augmentation baseline at week 16/EOS
c
Diastolic blood pressure, mmHg 0.4 77.69 1.5 78.60 Diastolic blood pressure, mmHgc 0.1 76.55 2.6 7 7.17
c
Systolic blood pressure, mmHg 0.4 710.04 1.2 79.70 Systolic blood pressure, mmHgc 0.3 7 8.39 2.6 7 9.79
c c
Pulse, bpm 0.7 7 9.75 5.9 7 10.64 Pulse, bpm 0.5 7 8.31 5.2 7 10.34
ECG heart rate, bpmd 0.3 7 8.84 6.17 10.29 ECG heart rate, bpmd 0.5 7 8.01 5.2 7 10.67
ECG QT duration, msd 1.8 7 21.67 11.4 7 23.57 ECG QT duration, msd 1.6 7 19.80 10.7 7 25.23
ECG QTcF, msd 0.8 711.30 0.8 7 12.80 ECG QTcF, msd 0.0 7 12.27 1.7 7 13.03
Weight, kgc 0.3 7 1.90 1.0 7 2.16 Weight, kgc 0.2 7 1.64 1.0 7 2.21
2c 2c
BMI, kg/m 0.17 0.67 0.4 7 0.77 BMI, kg/m 0.1 70.58 0.3 70.77
Potentially clinically important vital signs at 2 consecutive visits (of which 1 was the Potentially clinically important vital signs at 2 consecutive visits (of which 1 was
last visit)c the last visit)c
Systolic blood pressure 2 (1.0) 0 Systolic blood pressure 0 3 (1.4)
Z 140 mmHgc Z 140 mmHgc
Diastolic blood pressure 2 (1.0) 0 Diastolic blood pressure 0 5 (2.4)
Z 90 mmHgc Z 90 mmHgc
c c
Pulse Z100 bpm 0 0 Pulse Z 100 bpm 0 3 (1.4)
BMI¼ body mass index; ECG ¼electrocardiogram; EOS ¼end of study; QTcF ¼Fridericia's corrected QTC interval; TEAE ¼treatment-emergent adverse event.
a
Severe TEAEs: study 1 (placebo: gingivitis, wound infection, laceration, headache, insomnia, suicide attempt [n¼ 1 for all]; LDX: fatigue, appendicitis, migraine, syncope,
apathy [n ¼1 for all]); study 2 (placebo: seasonal allergy, sleep disorder, suicidal ideation [n¼ 1 for all]; LDX: dental caries, dry mouth, dyspepsia, nasopharyngitis, anxiety,
insomnia, restlessness, hot flush [n¼ 1 for all]).
b
TEAEs leading to discontinuation: study 1 (placebo: diarrhea, disturbance in attention, liver function test abnormal, syncope, suicide attempt, major depression, and
rash [n¼ 1 for all]; LDX: accidental overdose, sedation, systolic blood pressure increased, liver function test abnormal, musculoskeletal pain, pain in extremity, syncope, self-
injurious behavior, and palpitations [n¼ 1 for all]); study 2 (placebo: suicidal ideation [n¼ 1]; LDX: headache and anxiety [both n ¼1]).
c
Study 1 (placebo, n ¼ 200; LDX, n¼ 201); study 2 (placebo, n ¼ 213; LDX, n ¼ 209).
d
Study 1 (placebo, n ¼ 189; LDX, n ¼ 189); study 2 (placebo, n ¼202; LDX, n ¼ 196).
primary efficacy endpoint by a small pool of calibrated raters to subtypes. It should also be noted that the treatment duration in
reduce the overall number of raters and the potential for varia- the current studies was relatively short. A longer duration anti-
bility across raters. As inter-rater reliability was not assessed in the depressant lead-in phase may have increased the number of par-
current studies, it is a limitation that variability across raters is ticipants exhibiting antidepressant responses, and a longer dura-
unknown. The use of telephone measures is a method now being tion double-blind treatment phase may have increased the mag-
used in clinical trials to reduce bias (Kobak et al., 2008). Even
nitude of LDX responses. Finally, clinical trials for MDD are sus-
though it may offer less specificity and more variability than a
ceptible to having high placebo responses (Walsh et al., 2002),
face-to-face clinical assessment, due in part to the inability to vi-
which could mask a potential therapeutic effect.
sually observe the participant, it is important to note that the
In conclusion, in the 2 largest, randomized clinical trials of
validity of using standardized central rating via telephone corre-
lates well with face-to-face interviews (Kobak et al., 2008) and stimulant augmentation of antidepressant therapy completed to
that site- and participant-rated secondary assessments were con- date, LDX was not superior to placebo in reducing the depressive
sistent with the primary efficacy assessment. In addition, as al- symptoms of MDD in individuals with inadequate responses to
luded to above, it is possible that psychostimulants provide di- standard antidepressant monotherapy. Although these studies did
mensional efficacy and that assessment of specific MADRS items or not show a treatment difference, the potential exists that stimu-
clusters may have revealed effects of LDX that were not apparent lant augmentation may be appropriate in subgroups of individuals
in the total score. However, a detailed assessment of MADRS do- with MDD (eg, treatment-resistant MDD) or with a different sti-
mains was not performed because the primary and key secondary mulant augmentation.
endpoints consistently failed in both studies, making such as-
sessments statistically inappropriate and unreliable. Also, partici-
pants in the current studies lacked diversity in terms of race (most
were White) and gender (most were female). Those with psy- Appendix A. Supporting information
chiatric comorbidities (including ADHD) and/or active medical
disorders were excluded and executive functioning was not fully Supplementary data associated with this article can be found in
examined in these studies. Therefore, these findings are limited to the online version at http://dx.doi:10.1016/j.jad.2016.07.006.
a specific MDD population and do not extend to potential MDD
160 C. Richards et al. / Journal of Affective Disorders 206 (2016) 151–160