You are on page 1of 11

Expert Opinion on Drug Metabolism & Toxicology

ISSN: 1742-5255 (Print) 1744-7607 (Online) Journal homepage: http://www.tandfonline.com/loi/iemt20

Lisdexamfetamine: chemistry,
pharmacodynamics, pharmacokinetics, and
clinical efficacy, safety, and tolerability in the
treatment of binge eating disorder

Kristen Ward & Leslie Citrome

To cite this article: Kristen Ward & Leslie Citrome (2018) Lisdexamfetamine: chemistry,
pharmacodynamics, pharmacokinetics, and clinical efficacy, safety, and tolerability in the treatment
of binge eating disorder, Expert Opinion on Drug Metabolism & Toxicology, 14:2, 229-238, DOI:
10.1080/17425255.2018.1420163

To link to this article: https://doi.org/10.1080/17425255.2018.1420163

Accepted author version posted online: 20


Dec 2017.
Published online: 04 Jan 2018.

Submit your article to this journal

Article views: 168

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iemt20
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY, 2018
VOL. 14, NO. 2, 229–238
https://doi.org/10.1080/17425255.2018.1420163

DRUG EVALUATION

Lisdexamfetamine: chemistry, pharmacodynamics, pharmacokinetics, and clinical


efficacy, safety, and tolerability in the treatment of binge eating disorder
Kristen Warda and Leslie Citromeb
a
University of Michigan College of Pharmacy, Ann Arbor, MI, USA; bPsychiatry and Behavioral Sciences, New York Medical College, Valhalla, NY USA

ABSTRACT ARTICLE HISTORY


Introduction: The indications for lisdexamfetamine (LDX), a central nervous system stimulant, were Received 26 September 2017
recently expanded to include treatment of moderate to severe binge eating disorder (BED). Accepted 18 December 2017
Areas covered: This review aims to describe the chemistry and pharmacology of LDX, as well as the KEYWORDS
clinical trials investigating the efficacy and safety of this medication for the management of BED. Binge eating;
Expert opinion: LDX is the first medication with United States Food and Drug Administration approval for d-amphetamine;
the treatment of BED. It is an inactive prodrug of d-amphetamine that extends the half-life of d-ampheta- LDX; lisdexamfetamine;
mine to allow for once daily dosing. D-amphetamine acts primarily to increase the concentrations of pharmacodynamics;
synaptic dopamine and norepinephrine. Metabolism of LDX to d-amphetamine occurs when peptidases pharmacokinetics; SPD489;
in red blood cells cleave the covalent bond between d-amphetamine and l-lysine. D-amphetamine is then safety; stimulant; tolerability
further metabolized by CYP2D6. Excretion is primarily through renal mechanisms. In clinical trials, LDX
demonstrated statistical and clinical superiority over placebo in reducing binge eating days per week at
doses of 50 and 70 mg daily. Commonly reported side effects of LDX include dry mouth, insomnia, weight
loss, and headache, and its use should be avoided in patients with known structural cardiac abnormalities,
cardiomyopathy, serious heart arrhythmia or coronary artery disease. As with all CNS stimulants, risk of
abuse needs to be assessed prior to prescribing.

1. Introduction patients with BED range from slightly less than 50% [3] to
approximately 70% [4], obese patients with BED are more
Binge eating disorder (BED) can be summarized as repetitive
likely to have decreased functional capacity and quality of
episodes of eating more food than would normally be con-
life than obese patients without BED [4].
sumed by an average person in the same setting, without
American Psychiatric Association treatment guidelines,
compensatory purging or exercising, and feeling unable to
last published in 2006, strongly recommend the use of
control this behavior [1]. Although the BED diagnosis was
cognitive behavioral therapy (CBT) and guided self-help
officially recognized for the first time in DSM-5, the DSM-IV
programs in the treatment of BED, and there is adequate
and the text revision version of the DSM-IV (DSM-IV-TR) [2]
evidence to support the off-label use of antidepressants,
contained preliminary BED diagnostic criteria that were
particularly selective serotonin reuptake inhibitors (SSRIs),
commonly used in research studies, including those dis-
and the mood stabilizer topiramate [5]. Topiramate [6], but
cussed in this review. A key difference between DSM-5 and
not antidepressants [7], has also been associated with
DSM-IV-TR criteria is that the required average binge eating
improved response to CBT in the treatment of BED. In
(BE) episode occurrence was modified from twice weekly for
2015 lisdexamfetamine dimesylate (LDX) became the first
the past 6 months, to once weekly for the past 3 months
medication approved by the United States Food and Drug
[1,2]. BED is the most common eating disorder, with an
Administration (FDA) for the treatment of moderate to
estimated lifetime prevalence of 2.8% (3.5% females, 2.0%
severe BED (Box 1) [8]. As it can be often difficult for
males) [3], representing a large number of patients who
patients to access therapy providers specialized in eating
would potentially benefit from treatment for BED over the
disorders in their community, this development addresses a
course of their lifetime.
significant unmet medical need for more effective drug
An important consideration when selecting treatment
therapies that are not associated with increased weight, as
strategies for patients with BED is the high rate of comorbid
with many antidepressants, or the potential for cognitive
psychiatric diagnoses. Results from the National
impairment, as with topiramate.
Comorbidity Survey Replication (NCS-R) of approximately
In this review, we compile and present the available evi-
10,000 people noted that 79% of responders with BED
dence on the chemistry, mechanism of action, and pharmaco-
met criteria for at least one comorbid DSM-IV diagnosis,
kinetics of LDX, and will describe clinical trials investigating the
and that 49% met criteria for at least three additional
efficacy, tolerability and safety of LDX for the treatment of BED.
diagnoses [3]. Additionally, while observed obesity rates in

CONTACT Leslie Citrome citrome@cnsconsultant.com Psychiatry and Behavioral Sciences, New York Medical College, 11 Medical Park Drive, Suite
106, Pomona, NY 10970, USA
© 2018 Informa UK Limited, trading as Taylor & Francis Group
230 K. WARD AND L. CITROME

Box 1. Drug summary.


Drug name (generic) Lisdexamfetamine dimesylate
Phase (for indication under Product is approved for use
discussion)
Pharmacology description/ Stimulant/Prodrug of d-amphetamine, a non-catecholamine sympathomimetic amine, that increases synaptic
mechanism of action concentration of norepinephrine and dopamine via multiple mechanisms
Route of administration Oral
Chemical structure (2S)-2,6-diamino-N-[(1S)-1-methyl-2-phenylethyl] hexanamide dimethanesulfonate

Pivotal trials One Phase II [49], and two Phase III [50], 11–12 week randomized controlled trials in patients with moderate to severe
BED. All trials showed improvement over placebo in reducing binge eating days per week at 50 or 70 mg/day doses.

2. Search criteria weight loss [15], which may be beneficial for obese BED
patients.
In July 2017, the US National Library of Medicine’s PubMed
resource (http://www.ncbi.nlm.nih.gov/pubmed/) and the
EMBASE (Elsevier) commercial database were queried with 3.2. LDX chemistry and mechanism of action
the terms ‘lisdexamfetamine’ OR ‘LDX’ OR ‘SPD489’ to identify
literature describing the pharmacodynamic, pharmacokinetic, LDX is an inactive pro-drug of d-amphetamine that is covalently
pharmacogenomic, or chemical properties of LDX. The term bonded to L-lysine [15]. D-amphetamine is a non-catecholamine
‘binge’ was included with filters for articles with human clinical sympathomimetic amine, the more potent dextrorotatory isomer
trials to identify primary literature describing the efficacy and of amphetamine [16], formed from LDX when peptidases in red
safety of LDX for the treatment of BED. Concurrently, clinical blood cells hydrolyze the parent compound [17,18]. In vitro
trial registries http://www.clinicaltrials.gov and http://www.clin studies suggest this activation process is efficient even in the
icaltrialsregister.eu were searched with the filters ‘lisdexamfe- setting of low hematocrit and sickle cell disease [18,19].
tamine’ and ‘binge’ to identify all clinical trials involving LDX The exact mechanism by which LDX improves symptoms
for the treatment of BED. This amounted to six completed, and of BED is unknown, but is likely related to d-amphetamine’s
one in-progress, clinical trials. Finally, the manufacturer of LDX ability to increase brain interstitial concentrations of norepi-
(Shire, Dublin, Ireland) was contacted to obtain copies of nephrine and dopamine through action at dopamine and
posters that have been presented at scientific congresses. norepinephrine membrane transporters [20–23] involving
factors such as extracellular sodium (Na+) binding [24] and
intracellular calcium (Ca2+) levels [25]. Dopamine concentra-
3. Overview of LDX tions may also be increased through non-dopamine trans-
porter-related mechanisms involving norepinephrine
3.1. Introduction to drug product
transmission [26]. Amphetamine also binds with less affinity
LDX is a central nervous system (CNS) stimulant that first to the serotonin transporter [21]. An in vitro study with
received FDA approval in 2007, as a schedule II medication, amphetamine and human monoamine transporters identi-
for the treatment of attention-deficit/hyperactivity disorder fied the following binding affinities for the dopamine, nor-
(ADHD) in children [8]. This was followed by approval for epinephrine, and serotonin transporters (inhibitory constant
the treatment of ADHD in adults (2008) and adolescents [KI] in µM ± standard error of the mean [SEM]), respectively:
(2010), and then for the maintenance treatment of ADHD 0.64 ± 0.14, 0.07 ± 0.01, and 38.46 ± 3.84 [21].
in adults in 2012 [8]. LDX was originally investigated for use Another potential mechanism of action of LDX for the
in managing BED following the observation that BED and treatment of BED is through d-amphetamine’s effect at the
ADHD share neurobiological and psychological features, trace amine-associated receptor 1 (TAAR1) [27]. TAAR1 is a
including dysregulation in dopamine signaling [9] and G-protein-coupled receptor that is activated by endogenous
impulsivity [10–12]. For a review of the neurobiological trace amines, such as tyramine, tryptamine, and octopamine;
changes underlying BED, see Kessler 2016 [13]. LDX influ- regulates dopamine; and moderates response to ampheta-
ences the concentration of some of the neurotransmitters mine and other psychostimulants [28]. TAAR1 agonism as a
that are associated with BED, decreases impulsivity in mechanism of improving BED symptoms was supported in a
patients with BED [10,14], and has a propensity to cause recent rat study that demonstrated a TAAR1 agonist reduced
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 231

the consumption of palatable food, without reducing regular LDX does not appear to significantly inhibit or induce CYP1A2,
chow intake, in normal and aversive settings [29]. 2D6, or 3A activity in vivo [41,42], but competitively inhibits
monoamine oxidase (MAO-A Ki 20µM [43]), leading to potentially
significant drug interactions with monoamine oxidase inhibitors
3.3. Pharmacokinetics such as selegiline [15]. There is also potential for 2D6 inhibitors to
increase d-amphetamine concentrations, and LDX may contri-
3.3.1. Absorption
bute to serotonin syndrome risk in patients with multiple med-
Table 1 provides a summary of pharmacokinetic parameters of
ications inhibiting serotonin reuptake, in addition to enhancing
LDX that were measured in adults with ADHD who had been
the activity of medications with sympathomimetic activity [15].
taking an average LDX dose (SD) of 62.5 (14.2) for 5 weeks
Serotonin syndrome occurs in the setting of elevated synaptic
[30]. LDX is available as capsules and chewable tablets, and
serotonin concentrations, with central and peripheral effects, and
the capsule can be opened and dissolved in water without
can be mild (gastrointestinal upset, tremors, agitation) to life-
decreasing the overall amount absorbed, as measured by area
threatening (seizures, hyperthermia, coma, and death) [44].
under the curve (AUC) [31]. Absorption of LDX from the gas-
trointestinal tract is rapid [18], and when taken with food, the
time to maximum serum concentration (tmax) is delayed by 3.3.4. Excretion
approximately one hour [31]. Additionally, gastric pH does not In adults with ADHD, the plasma half-life of LDX is approximately
appear to impact the absorption of d-amphetamine from LDX, 0.5 h, and the half-life of d-amphetamine is approximately 17.0 h
as measured by the maximum serum concentration (Cmax) and [30], which allows for once-daily dosing [15]. The majority (96%)
AUC in patients who were exposed to LDX with, and without, of LDX is excreted renally, mostly as d-amphetamine (41.5%) and
the proton pump inhibitor omeprazole [32]. hippuric acid (24.8%), but also 2.2% as intact LDX [45]. Several
dose adjustments are recommended by the manufacturer
related to renal excretion [15]. In patients with glomerular filtra-
3.3.2. Distribution tion rates (GFR) 15–30 mL/min/1.73 m2, the dose should be no
There is little data on the distribution of LDX. A small pharmaco-
more than 50 mg/day, and if GFR is less than 15 mL/min/1.73 m2,
kinetic study of healthy participants, the majority male, described
the dose should be no more than 30 mg/day. LDX and d-amphe-
d-amphetamine as following a one-compartment model of dis-
tamine do not appear to be recoverable in dialysate, and there-
tribution with a plasma volume (95% CI) of 377 L (326–428) [34].
fore LDX should not be recommended in patients on dialysis [46].
This is a relatively large volume of distribution, signifying that the
Additionally, renal excretion appears to be dependent on urine
drug leaves the intravascular space and distributes to most
pH, with higher blood levels of amphetamine observed with
tissues in the body. In a population of patients with high rates
higher urine pH, and may require dose adjustments if a patient
of obesity, this may have implications for dose requirements, but
is taking additional medication that changes urine pH [15].
it is difficult to predict because lipophilic drugs do not always
require dosage adjustments in obesity [35]. The extent of LDX
plasma protein binding is not known; however, an in vitro equili- 3.4. Pharmacogenetics
brium dialysis study of amphetamine noted a mean percentage
Currently, there are no published studies examining the relation-
of bound amphetamine in humans (±SEM) to be 14.5 ± 0.9, these
results were independent of drug concentration and did not ship between genetic variability and response to LDX as a treat-
differ significantly when the plasma was uremic [36]. This indi- ment for BED. A small study (N = 32) in 2007 noted that a dopamine
transporter gene (DAT1) variant was associated with more appetite
cates that protein binding is unlikely to be clinically significant,
with respect to impact on free amphetamine in the plasma. suppression in BED subjects who took the stimulant methylpheni-
date, when compared to control participants with the same geno-
type [47]. Additionally, a recent review highlights a number of
3.3.3. Metabolism studies suggesting that genetic variability in genes coding for
As mentioned previously, LDX is transformed by peptidases in monoamine transporters, and enzymes that impact monoamine
red blood cells to form d-amphetamine. D-amphetamine is concentrations, may be associated with response to amphetamine
further metabolized in the liver to hippuric acid, benzoic as it pertains to ADHD response or abuse or dependence potential
acid, norephedrine, 4-hydroxynorephedrine, and benzyl [48]. At this point, none of the associations reviewed are supported
methyl ketone [15,37]. Although LDX itself is not metabolized by sufficient evidence to warrant dosing recommendation
by cytochrome (CYP) P450 enzymes, amphetamine is primarily changes for amphetamine products used to treat ADHD or BED.
metabolized by CYP2D6 [38–40].

Table 1. Summary of lisdexamfetamine and d-amphetamine pharmacokinetics.a


4. Clinical trials investigating the use of LDX for the
treatment of BED
Pharmacokinetic parameter Lisdexamfetamine D-amphetamine
Cmax (ng/mL) 25.0 67.9 4.1. Efficacy
Tmax (hours) 1.5 4.4
AUC (ng/mL × hours) 45.9 641.6 Clinical trials involving the use of LDX for the treatment of BED
T1/2 0.5 17 are summarized in Table 2. Three randomized, placebo-con-
a
Data summarized from Adler et al. 2017 [33] trolled trials, one phase II and two phase III, were pivotal for
AUC: area under the curve, Cmax: maximum serum concentration, Tmax: time to
maximum serum concentration, T1/2: time when half of the concentration of demonstrating the efficacy and safety of LDX for the treatment
drug in the body is eliminated. of BED [49,50], and ultimately resulted in FDA approval for this
232

Table 2. Clinical trials determining the efficacy and safety of lisdexamfetamine for binge eating disorder in adults.a
Length (weeks), dose Phase,
Study title (mg/day) Nb Primary outcome Primary outcome results Study period Sponsor, trial IDc, resultsd
A phase 2, multicenter, randomized, double-blind, 11 II Change in log-transformed binge eating ● Placebo: −1.23 May 2011–January 2012 Shire
parallel-group, placebo-controlled, forced-dose 30, 50, or 70 255 days per week, as measured between (SE 0.069) NCT01291173
titration Study to evaluate the efficacy, safety, baseline and week 11 and compared ● 30 mg/day: −1.24 Results on ClinicalTrials.gov and
and tolerability of SPD489 in adults aged to placebo (SE 0.067) published: PMID 25587645
18–55 years with binge eating disorder [49] ● 50 mg/day: −1.46
(SE 0.066), p = 0.008
● 70 mg/day: −1.57
(SE 0.067), p = 0.002
K. WARD AND L. CITROME

The SPD489343, phase 3, multicenter, randomized, 12 III Change from baseline to week 11 or 12 ● Placebo: −2.51 November 2012– Shire
double-blind, parallel-group, placebo-controlled, 50 or 70 374 (visit 8) in number of binge eating (SE 0.125) September 2013 NCT01718483
dose-optimization study to evaluate the efficacy, days per week ● 50 or 70 mg/day: 2012–003309-91
safety and tolerability of SPD489 in adults aged -3.87 (SE 0.124), Results on ClinicalTrials.gov and
18–55 years with moderate to severe binge ● p < 0.001 published: PMID 26346638
eating disorder [50]
The SPD489344, phase 3, multicenter, randomized, 12 III Change from baseline to week 11 or 12 ● Placebo: −2.26 November 2012– Shire
double-blind, parallel-group, placebo-controlled, 50 or 70 350 (visit 8) in number of binge eating (SE 0.137) September 2013 NCT01718509
dose-optimization study to evaluate the efficacy, days per week ● 50 or 70 mg/day: 2012–003310-14
safety, and tolerability of SPD489 in adults aged −3.92 (SE 0.135), Results on ClinicalTrials.gov and
18–55 years with moderate to severe binge p < 0.001 published: PMID 26346638
eating disorder [50]
Lisdexamfetamine in binge eating disorder of 12 III Rate of reduction of mean binge eating ● LDX change from January 2010 – October Linder Center of Hope with
moderate or greater severity [51] 20 to 70 50 days per week baseline – placebo 2012 collaborators Shire and University
change from baseline of Cincinnati
(95% CI) = −0.7 (0.5– NCT01090713
1.0), p = 0.08 Published results:
PMID 27650406
A phase 3, multicenter, open-label, 12 month 53 III 1) Percentage of patients with TEAEs ●Any TEAE: 84.5% August 2012 – October Shire
extension safety and tolerability study of SPD489 50 or 70 599e (week 52) ●Serious TEAE: 2.8% 2014 NCT01657019
in the treatment of adults with binge eating 2) Number with a positive response on ●Active SI: 2 2012–003313-34
disorder [57] the C-SSRS (week 53) ●Non-suicidal self- Results on ClinicalTrials.gov and
injurious behavior: 3 published: PMID 28383364
A phase 3, multicenter, double-blind, placebo- 26 III Time to relapse (2 or more binge days/ ● Relapse criteria was January 2014–April 2015 Shire
controlled, randomized-withdrawal study to 50 or 70 275 week for 2 consecutive weeks with an met for 3.7% of NCT02009163
evaluate the maintenance of efficacy of SPD489 increase in CGI-S ≥ 2) measured from participants on LDX, 2012-004457-88
in adults aged 18–55 years with moderate to 26 weeks after randomization when and 32.1% of the Results on ClinicalTrials.gov and
severe binge eating disorder [52] compared to placebo participants receiving published: PMID 28700805
placebo,
p < 0.001
Effects of lisdexamfetamine on prefrontal brain 12 II Measurement of brain activation in the N/A September 2015– Lindner Center of HOPE with
dysfunction in binge eating disorder 50 or 70 40f ventral prefrontal, striatal, and December 2017 collaborator University of
amygdala with fMRI following food (estimated) Cincinnati
cues NCT02659488
No results available, still recruiting
a
Records identified on ClinicalTrials.gov and clinicaltrialsregister.eu in June 2017.
b
Number of participants included in primary outcome analysis.
c
ClinicalTrials.gov and EU Clinical Trials Register identifiers, as appropriate.
d
PubMed ID provided for published study results, if applicable.
e
N was 599 for the primary outcome of any TEAE, N was 597 for the primary outcome of participants with a positive response on the C-SSRS.
f
Estimated enrollment, trial is still recruiting.
CI: confidence interval; CGI-S: Clinical Global Impression-Severity scale; C-SSRS: Columbia Suicide Severity Rating Scale; SE: standard error; TEAE: treatment emergent adverse event.;
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 233

indication (summarized in Table 2, rows 1–3). All three trials day, 30 mg/day, and placebo groups, respectively. Among
share several similarities: (1) recruited adults 18–55 years old significant secondary end point outcomes, CGI-I score was
with moderate to severe BED based on DSM-IV-TR criteria and significantly favorable for all dosages of LDX when compared
experiencing ≥3 BE days/week for at least two consecutive to placebo (scores of 1 or 2 at week 11).
weeks before baseline, (2) measured change in BE days per The two, 12-week, phase III trials investigating LDX efficacy,
week from baseline to weeks 11 or 12 of treatment, (3) the safety, and tolerability were structured to have matching
majority of participants were white, obese, females, with an designs and methods (listed in Table 2, with internal assign-
average age of approximately 40 years, (4) adherence was ment classifiers SPD489343 and SPD489344 in their titles) [50].
tracked by counting remaining pills at study visits, (5) efficacy For ease of reference they will be referred to as study 343 and
and safety analyses were completed for all participants who 344 when they are discussed separately. Both were interna-
took one dose of study drug and completed one post-baseline tional, randomized, double-blind, placebo-controlled trials.
assessment, and (6) were sponsored by Shire (manufacturer of Inclusion/exclusion criteria that are different from the phase
branded LDX product). II trial include a CGI-S score of at least 4 at baseline (indicating
The Phase II trial was conducted at multiple sites in the moderate illness, with higher numbers corresponding to more
United States [49]. Patients were excluded if (1) their body severe illness), and a modification of the exclusionary BMI
mass index (BMI) was <25 or >45 kg/m2; (2) they had used a range to <18 kg/m2 (maintaining >45 kg/m2 exclusionary
medication for weight loss, or a psychostimulant recently (3 criterion). Eligible participants were randomized (1:1) to pla-
and 6 months, respectively); (3) had a history of personal or cebo or dose-optimized LDX. As compared to the phase II trial,
familial cardiovascular disease (CVD) that may make them 4 weeks, instead of 3, were provided for dose titration. During
more sensitive to stimulant side effects; (4) those with a dose optimization, patients assigned to LDX began treatment
recent history of substance abuse (not nicotine depen- at 30 mg/day, and after 1 week of treatment were subse-
dence); or (5) had a Montgomery–Asberg Depression Score quently titrated to 50 mg/day. Additional increases to
(MADRS) of at least 18 at baseline. Additional exclusion 70 mg/day were made as tolerated and clinically indicated.
criteria included comorbid anorexia or bulimia nervosa, Participants could decrease their dose from 70 to 50 mg/day
ADHD or another psychiatric disorder, and any history of during the last 2 weeks of the optimization phase. The main-
psychostimulant abuse or dependence in addition to recent tenance period lasted 8 weeks.
substance abuse (except nicotine). Patients who had used As in the phase II trial, the primary end point measure was
antidepressants, sedatives, anxiolytics, antipsychotics, benzo- change from baseline to the end of the maintenance period in
diazepines, antihistamines, herbals preparations, or over-the- BE days/week, but this value was not log-transformed. A
counter medications within the last 30 days were also mixed effect model was used in the primary analysis.
excluded. Female participants were required to have nega- Approximately 60% of participants in the LDX groups of
tive pregnancy tests and be willing to use contraception. each study tolerated 70 mg/daily, and the average daily dose
Eligible participants were randomized (1:1:1:1) into placebo, (SD) of LDX was 56.9 (9.72) and 57.6 (9.24) mg/day for study
30 mg, 50 mg, or 70 mg LDX daily groups. All capsules were 343 and 344, respectively. A total of 374 participants from
visually identical, and participants randomized to study drug study 343 and 350 participants from study 344 were included
were started at 30 mg daily, and increased by 20 mg weekly in the primary analysis. Results from both studies identified
until they reached their assigned dose, at which time main- significant improvement in LDX’s ability to decrease BE days/
tenance continued for another 8 weeks. The primary efficacy week when compared to placebo.
measure was number of BE days per week, and was ana- An additional phase III, placebo-controlled, double-blind,
lyzed after log-transformation of the number of BE days per flexible-dose study was conducted at the Lindner Center of
week (+1). There were an extensive number of secondary HOPE in Mason, Ohio [51]. This study was designed before the
efficacy measures used in this study, including number of BE previously discussed clinical trials. The flexible dose design
episodes per week, 4-week cessation (no binge episodes), allowed participants to receive doses as low as 20 mg/day
score on the Clinical Global Impressions-Improvement scale based on tolerability. The primary outcome measure in this
(CGI-I, dichotomized as ratings of 1–2, or 3–7, with lower trial was the longitudinal rate of change of BE days/week, and
numbers denoting improvement in symptoms), the Yale– all 50 participants were included in the primary outcome
Brown Obsessive Compulsive Scale modified for BE (Y- analysis. The mean (SD) LDX dose at end point was 59.6
BOCS-BE), the Barratt Impulsiveness Scale (BIS-11), the (14.9) mg/day. Results of the primary outcome measure
MADRS, and the Hamilton Anxiety Rating Scale. demonstrated that rate of reduction in BE days/week was
The primary end point was least squares mean change in not significantly different between the LDX group and the
BE days per week from baseline to week 11 (or early termina- placebo group (Table 1), but the secondary end points of
tion). Mixed-effects modeling for repeated measures was used change in BE days/week and episodes/week were different
to analyze data for the primary outcome. About 255 partici- between the two groups (p = 0.03 and 0.005, respectively).
pants were included in the efficacy analysis. As shown in An additional phase III trial was constructed with the aim of
Table 2, the primary end point demonstrated significant assessing the maintenance of efficacy of LDX in an interna-
improvement over placebo with the 50 and 70 mg/day tional, double-blind, placebo-controlled randomized withdra-
doses. The non-transformed BE days per week (SD) changed wal study [52]. Inclusion/exclusion criteria were similar to
from 4.6 (1.25) to 0.5 (1.25), 4.5 (1.28) to 0.4 (0.86), 4.5 (1.44) to those in studies 343 and 344. Following a 12-week open-
1.0 (1.69), and 4.3 (1.38) to 1.1 (1.45) in the 70 mg/day, 50 mg/ label phase that was modeled after trials 343 and 344,
234 K. WARD AND L. CITROME

participants who were classified as responders (≤1 BE day per Table 3. Treatment-emergent adverse events occurring in at least 5% of parti-
cipants taking lisdexamfetamine in clinical trials for binge eating disorder,
week for 4 consecutive weeks and CGI-S scores ≤2 [borderline separated by trial design.
ill or less]) at week 12 were randomized to placebo, or con- A. Acute, double-blind randomized controlled trialsa
tinued LDX, during a 26-week, double-blind randomized with- TEAE Lisdexamfetamine Placebo
drawal phase. The primary outcome variable was time to N (% out of 594) N (% out of 460)
relapse (≥2 BE days per week for 2 consecutive weeks and Dry mouth 219 (36.9) 32 (7.0)
Headache 86 (14.5) 44 (9.6)
≥2-point CGI-S score increase) from randomized withdrawal Insomnia 90 (15.2) 23 (5.0)
baseline, and was analyzed with a log-rank test. The primary Decreased appetite 70 (11.8) 13 (2.8)
outcome was assessed using participants who had received at Nausea 49 (8.2) 25 (5.4)
Constipation 35 (5.9) 8 (1.7)
least one dose LDX and completed one CGI-S assessment Upper RTI 22 (3.7) 17 (3.7)
post-randomization. Log-rank testing was used to calculate Feeling jittery 37 (6.2) 2 (0.4)
the primary end point, and it was stratified based on 4-week Irritability 36 (6.1) 24 (5.2)
B. Acute, open-label phase of maintenance studyb
cessation status. TEAE Lisdexamfetamine
A total of 418 participants were enrolled in the open-label N (% out of 411)
phase, and 275 responders from the open-label phase entered Dry mouth 139 (33.8)
Headache 66 (16.1)
the randomized-withdrawal phase. Out of the 143 participants Insomnia 46 (11.2)
who withdrew from the open-label phase, 22 were reportedly Decreased appetite 38 (9.2)
due to adverse events, and 48 failed to meet criteria for the Nausea 35 (8.5)
Anxiety 29 (7.1)
randomized phase. Another six participants withdrew from the Constipation 28 (6.8)
LDX randomized group (none from the placebo group) due to Hyperhidrosis 23 (5.6)
adverse events. Results from the primary outcome analysis Feeling jittery 21 (5.1)
Diarrhea 21 (5.1)
indicate that the risk of relapse over 26 weeks was lower in C. 26-week double-blind, randomized controlled withdrawal phase of
those continuing LDX than those randomized to placebo, with maintenance studyc
32.1% of participants on placebo relapsing, as compared to TEAE Lisdexamfetamine Placebo
N (% out of 136) N (% out of 134)
3.7% of participants on LDX (p < 0.001) (Table 2). Nasopharyngitis 13 (9.6) 9 (6.7)
Although the results of the secondary analyses are not Headache 12 (8.8) 9 (6.7)
described in detail here, highlights from the secondary analyses Upper RTI 11 (8.1) 5 (3.7)
Dry mouth 7 (5.0) 2 (1.5)
across all available studies are suggestive of the ability of LDX to D. Open-label, 12-month extension studyd
decrease obsessive-compulsive thoughts and behavior (measured TEAE Lisdexamfetamine
by Y-BOCS-BE) and impulsive features of BED (measured by BIS- N (% out of 599)
Dry mouth 163 (27.2)
11), as well as symptom improvement (CGI-I), and that response to Headache 79 (13.2)
LDX appears as early as week 1 after treatment. McElroy and Insomnia 74 (12.4)
colleagues have provided a more thorough discussion on these Upper RTI 68 (11.4)
Nasopharyngitis 53 (8.8)
secondary outcomes and the advantages and limitations of the Nausea 41 (6.8)
selected scales in two recent publications [14,53]. Constipation 41 (6.8)
Decreased appetite 36 (6.0)
Irritability 36 (6.0)
Bruxism 35 (5.8)
Sinusitis 35 (5.8)
4.2. Safety and tolerability Feeling jittery 30 (5.0)
Anxiety 30 (5.0)
In clinical trials utilizing LDX for the treatment of BED, the
a
profile of treatment-emergent adverse events (TEAEs) was One phase II [49] and three phase III trials [50,51] were included in this
summary; all were 11–12 weeks in duration.
consistent with those seen in adults with ADHD [54–56]. b
Adverse events reported during the 12-week open-label phase preceding the
Approximately 80–85% of participants taking LDX experienced 26-week, double-blind randomized withdrawal period of the maintenance
any adverse event, but dropouts due to TEAEs in participants study [52].
c
Adverse events reported during the 26-week, double-blind, randomized with-
on LDX were relatively low in all trials (<10% of participants). drawal phase of the maintenance study.
d
Table 3 provides a list of common adverse events (occurring in Adverse events reported during the 12-month extension [57] study, in which
≥5% of participants receiving LDX) reported in the LDX for participants were recruited from previously described double-blind rando-
mized controlled trials [49,50].
BED clinical trials [49–52,57]. They are stratified by study type: N: number of participants, RTI: respiratory tract infection, TEAE: Treatment-
acute (3A) and maintenance (3C) double-blind randomized emergent adverse event.
controlled trials, and open-label study phases (3B and 3D).
When appropriate, pooled values were reported with TEAE
data from all participants who completed the safety analysis phase II and phase III studies: 343 and 344 [57]. The primary
and were receiving LDX (either open-label or blinded), and objectives of this study were to assess occurrence of TEAEs and
dividing the total number of participants reporting the side positive responses on the C-SSRS. The study consisted of a 4-
effect by the total number of participants taking LDX. A similar week dose optimization phase that was the same as in trials
calculation was used for patients receiving placebo [49–51]. 343 and 344, followed by a 48-week maintenance phase.
Long-term safety and tolerability of LDX for BED was Inclusion criteria required no clinically significant TEAEs during
assessed through an open-label extension trial that was con- participation in the previous qualifying trial, and otherwise had
ducted by recruiting participants from the previously discussed to meet inclusion criteria for the studies 343 and 344. If the
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 235

participants were enrolled 30 or more days after they com- substance abuse, although one participant in the 52-week
pleted the previous study, they could be excluded if they clinical trial discontinued the study due to periodic drug crav-
started a lipid-lowering medication <3 months before screen- ings [57]. There was also a death in the phase II trial consistent
ing. Measures of safety and tolerability included TEAEs, vital with methamphetamine overdose in a participant who denied
signs, weight changes, ECGs, laboratory evaluations, and a history substance abuse during the screening [49].
C-SSRS scores. When changes from baseline were described, Additional warnings and precautions from the product
baseline was set as the prior study baseline, or the extension label include (1) serious cardiovascular reactions such as sud-
study screening visit (if enrolled > 30 days from previous study). den death, stroke, and myocardial infarctions, and recom-
Out of the 604 enrolled, 599 were included in the safety mends avoiding use in patients with serious heart problems;
analysis, and 61% completed the study (369 participants). Over (2) blood pressure and heart rate increases; and (3) psychiatric
the course of the study 9% (54 participants) left due to adverse reactions that include exacerbating preexisting psy-
adverse events, but only 0.5% left due to lack of efficacy (3). chosis and potentiating new psychotic or manic symptoms.
The majority of participants were maintained at the 70 mg/day Serious adverse events involving these drug warnings were
dose (64.9%, 389 participants), and average length (SD) of rare in clinical trials of LDX for BED, except for increased heart
drug exposure was 284.3 (118.84) days. Serious TEAEs deter- rate and blood pressure that was observed in 5–10 percent of
mined to be due to study drug included (one instance of each patients taking LDX in these clinical trials. When described in
in the same participant upon taking 100 mg LDX the day of the 12-month safety study, the authors noted that <5% of
the event) were acute coronary syndrome and supraventricu- participants had changes in blood pressure or pulse that
lar tachycardia. Severe TEAEs considered related to study drug were clinically significant [57].
were irritability (3), dry mouth (2), and insomnia (2). When the investigators of the safety study presented
The LDX product label includes a boxed warning for abuse change in blood pressure and pulse graphically over the year
potential [15], although animal studies suggest LDX may have and to the week of follow-up visit, it is observable that pulse
less reinforcing effects than immediate release d-amphetamine increase occurred within the first week of therapy and roughly
[58]. This was supported in a 2009 human study, with a pla- plateaued by week 8. The authors report that heart rate
cebo-controlled, double-blind, randomized crossover design, peaked with an increase of 8.85 (SD 10.24) beats per minute
that compared the abuse potential of single doses of oral on week 28. Heart rate elevation above baseline dropped to
d-amphetamine (40 mg), LDX (50, 100, and 150 mg), and approximately 5 bpm at the 1-week posttreatment follow-up
diethylpropion (200mg) to placebo in 36 participants with a visit. Blood pressure increased slowly from baseline to a peak
history of stimulant abuse [59]. The primary measure in this increase of 2.78 (SD 10.56) mm Hg systolic (week 28) and 2.09
study was change on the Liking Scale of the Drug Rating (SD 8.215) mm Hg diastolic (week 16) over baseline, but also
Questionnaire-Subject from stimulant to placebo, and this mea- followed the same approximate pattern of plateauing at week
sure was significant for d-amphetamine and diethylpropion, but 8. At the follow-up visit 1-week posttreatment, blood pressure
was insignificant for all doses of LDX except 150 mg, which is had returned to approximately baseline. In comparison, the
equivalent to 50% more amphetamine product as compared to results of the two large, placebo-controlled randomized phase
d-amphetamine 40 mg [59]. Jasinski et al. also compared abuse III trials noted an increase in blood pressure of 0.2–1.45 mm
potential of intravenous d-amphetamine (20 mg) or LDX (25 or Hg (systolic) and 1.06–1.83 mm Hg (diastolic), and a heart rate
50 mg) to intravenous placebo in a randomized, double-blind, increase of 4.41–6.31 bpm after 12 weeks of treatment [50].
crossover study of adult stimulant abusers [60]. The authors Finally, weight loss is a known side effect of LDX treatment
found that abuse liability did not change significantly for LDX that may be considered beneficial in this population, but
when compared to placebo at either dose, but that it did for should be carefully monitored because the safety of LDX for
d-amphetamine. As attempting to change the pharmacokinetic weight loss has not been established, and past use of sym-
profile could impact its abuse potential, Ermer et al. also com- pathomimetic agents for weight loss has resulted in serious
pared intranasal and oral pharmacokinetics of 50 mg LDX in adverse cardiovascular side effects [15]. When weight loss was
healthy adult men, and found no significant differences in LDX monitored as a safety outcome in the phase III studies 343 and
or d-amphetamine with respect to maximum concentration, 344, it was found to be significantly higher than weight loss in
time to maximum concentration, or absorption [61]. patients taking placebo [50]. In the extension study, weight
It is important to note that conflicting results in abuse liability loss peaked at week 44, with an average loss (SD) of 8.21 kg
have been reported in a recent sample of 24 healthy adults (7.73), corresponding to a percent decrease in body weight
administered placebo, and equivalent concentrations of amphe- (SD) of 8.67% (7.848).
tamine base as d-amphetamine (40 mg) and LDX (100mg) in a
double-blind, placebo-controlled, randomized crossover study
[62]. Although pharmacokinetic differences were observed as
5. Regulatory issues
anticipated (e.g. delayed time to maximum concentration of
d-amphetamine with LDX, but similar d-amphetamine AUC), As discussed, LDX has been approved for the treatment of BED
Visual Analog Scale measures of abuse liability (drug liking, in adults in the United States based on the results of the phase
well-being, and more) were not different between d-ampheta- II and two phase III 11–12-week studies that described the
mine and LDX when compared to placebo [62]. efficacy and safety of LDX for this indication [49,50]. LDX has
The clinical studies on the efficacy of LDX for the treatment also been approved in Canada for the treatment of BED [63],
of BED discussed here excluded patients with a history of as well as in Israel [64]. Other agents are being commercially
236 K. WARD AND L. CITROME

developed for the management of BED, including dasotraline, NNT and NNH are measures of effect size and indicate how
currently in Phase III (NCT03107026). many patients would need to be treated with one agent
instead of the comparator to encounter one additional out-
come of interest. Using data pooled from three available
6. Conclusions
studies, NNT was calculated for the dichotomized CGI-I score
LDX is a CNS stimulant, and the first drug with FDA approval of improved (scores 1 and 2) versus not improved (3–7).
to treat BED. It is transformed from an inactive pro-drug to Highlights from this analysis include a NNT (95% CI) of 4 (4–
d-amphetamine by enzymes in red blood cells [17,18]. The 6) for doses of 30–70 mg/day to result in one more person
mechanism of action for the treatment of BED is likely related meeting CGI-I improvement as compared to placebo. When
to increased concentrations of dopamine, norepinephrine, and calculated for only the phase III trials (50 and 70 mg/day LDX
to a lesser extent, serotonin, in the synapse after d-ampheta- doses), the effect size is more robust: 3 (3–4). NNH (95% CI)
mine binds to monoamine transporters [21]. LDX is only avail- calculations of the most common potential adverse events
able in oral formulations, allows for once-daily dosing, and include: 4 (3–5), 11 (8–17), 11 (8–17), and 19 (11–75) for dry
needs to be dose-adjusted in settings of renal dysfunction mouth, decreased appetite, insomnia, and headache, respec-
[15]. The primary study outcomes for five out of six phase II tively [66]. In addition, results from the 26-week maintenance
and III clinical studies support the ability of LDX, at doses of 50 study evidenced lower rates of relapse for LDX than for pla-
and 70 mg/day, to decrease the number of BE days per week cebo, 3.7% versus 32.1% [44], for a NNT of 4 (95% CI 3–6).
in adult patients with moderate to severe BED [49,50,52,57]. Ultimately, clinicians will need to weigh the efficacy of LDX for
Secondary outcome analyses from these efficacy studies sug- improving BED outcomes against the potential for adverse
gest that LDX may also improve obsessive-compulsive, and events. Of note, adverse events may differ in terms of impact,
impulsive, aspects of BED in a time frame shorter than and may not be clinically relevant if the adverse event is mild,
11–12 weeks [14]. An additional phase III trial supports the time-limited, or easily managed. For example, the average
ability of LDX to decrease relapse rates over a 6-month period pulse rate and blood pressure increases noted in the
when compared to placebo [52]. In clinical trials, the most described studies can be more clinically significant in patients
common TEAEs were dry mouth, insomnia, headache, and with hypertension or congestive heart failure. A caveat is that
decreased appetite, and are reflective of the adverse event patients carry variable propensities to experience different
profile of LDX in adults with ADHD [54–56]. This pattern adverse events or to achieve a therapeutic response.
continued in a safety and tolerability study that was 1 year As mentioned previously, there are a large percentage of
in duration [57]. Future studies will need to assess the efficacy, BED patients with comorbid psychiatric diagnoses [3] and
safety, and tolerability of LDX for the treatment of BED in metabolic syndrome [67–70]. Physician willingness to pre-
studies with that are more inclusive of typical BED patients scribe LDX for the treatment of BED will likely be impacted
with respect to comorbid diagnoses. by concern over the lack of safety studies in BED patient
populations with controlled comorbid psychiatric (including
substance use disorders) or CVD. More inclusive clinical trials
7. Expert opinion
comparing the efficacy and safety of LDX in BED treatment
Prior to FDA approval of LDX as the first drug with an indica- will be important for making study results more general-
tion for the treatment of BED, SSRIs and topiramate had been izable. Additional studies comparing current treatments
considered among the best pharmacologic treatment options used for BED, and if LDX provides added benefit to CBT in
for this eating disorder [5]. The mechanism of action of LDX is BED treatment, will help providers determine the best treat-
unique when compared to these agents, and may represent a ment options for their patients.
more direct approach to balancing the underlying neuro-
chemical abnormalities in patients with BED. A currently
Funding
recruiting study aims to measure brain activation following
food cues in BED patients, before and after receiving 12 weeks This paper was not funded
LDX treatment that may better characterize the effects of LDX
on BED (Table 2). Unlike antidepressants and topiramate, LDX
also has the added benefit of not contributing to weight gain Declaration of Interest
(that can be seen with antidepressants) or cognitive impair- L Citrome has engaged in collaborative research with, or received con-
ment (that can be associated with the use of topiramate). A sulting fees from Acadia, Alexza, Alkermes, Allergan, AstraZeneca, Avanir,
potential downside of LDX therapy is acquisition cost, as it is Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, Forum, Genentech,
Intra-Cellular Therapeutics, Janssen, Jazz, Lundbeck, Merck, Medivation,
still available only as a branded product, while topiramate and
Mylan, Neurocrine, Novartis, Noven, Otsuka, Pfizer, Reckitt Benckiser,
many of the SSRIs are available as generic products. Reviva, Shire, Sunovion, Takeda, Teva, Valeant, and Vanda. The authors
Nonetheless, a recent study comparing 12 weeks LDX treat- have no other relevant affiliations or financial involvement with any
ment to placebo suggests that LDX provides cost-effective organization or entity with a financial interest in or financial conflict
improvements in quality-adjusted life years [65]. with the subject matter or materials discussed in the manuscript apart
from those disclosed. A reviewer on this manuscript has disclosed receiv-
To interpret the clinical utility of LDX treatment, Citrome
ing grants/research support, consulting fees and/or honoraria within the
completed a review on the use of LDX for BED that included last three years from Angelini, AOP Orphan Pharmaceuticals AG,
calculating the number needed to treat (NNT) and number AstraZeneca, Eli Lilly, Janssen, KRKA-Pharma, Lundbeck, Neuraxpharm,
needed to harm (NNH) when comparing LDX to placebo [66]. Pfizer, Pierre Fabre, Schwabe and Servier.
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 237

References 20. Volkow ND, Fowler JS, Wang G-J, et al. Dopamine in drug abuse
and addiction: results of imaging studies and treatment implica-
Papers of special note have been highlighted as either of interest (•) or of tions. Arch Neurol. 2007;64:1575–1579.
considerable interest (••) to readers. 21. Han DD, Gu HH. Comparison of the monoamine transporters from
1. American Psychiatric Association. Diagnostic and statistical manual human and mouse in their sensitivities to psychostimulant drugs.
of mental disorders. 5th ed. Washington (DC): American Psychiatric BMC Pharmacol. 2006;6:6.
Publishing; 2013. 22. Kahlig KM, Galli A. Regulation of dopamine transporter function
2. American Psychiatric Association. Diagnostic and statistical manual and plasma membrane expression by dopamine, amphetamine,
of mental disorders. 4th ed. Washington (DC): American Psychiatric and cocaine. Eur J Pharmacol. 2003;479:153–158.
Association; 2000. 23. Jones SR, Gainetdinov RR, Wightman RM, et al. Mechanisms of
3. Hudson JI, Hiripi E, Pope HG, et al. The prevalence and correlates of amphetamine action revealed in mice lacking the dopamine trans-
eating disorders in the national comorbidity survey replication. Biol porter. J Neurosci. 1998;18:1979–1986.
Psychiatry. 2007;61:348–358. 24. Pifl C, Singer EA. Ion dependence of carrier-mediated release in
4. Grucza RA, Przybeck TR, Cloninger CR. Prevalence and correlates of dopamine or norepinephrine transporter-transfected cells ques-
binge eating disorder in a community sample. Compr Psychiatry. tions the hypothesis of facilitated exchange diffusion. Mol
2007;48:124–131. Pharmacol. 1999;56:1047–1054.
5. American Psychiatric Association. Treatment of patients with eating 25. Gnegy ME, Khoshbouei H, Berg KA, et al. Intracellular Ca2+ regu-
disorders, third edition. Am Psychiatr Assoc Am J Psychiatry. lates amphetamine-induced dopamine efflux and currents
2006;163:4–54. mediated by the human dopamine transporter. Mol Pharmacol.
6. Claudino AM, de Oliveira IR, Appolinario JC, et al. Double-blind, 2004;66:137–143.
randomized, placebo-controlled trial of topiramate plus cognitive- 26. Dela Peña I, Gevorkiana R, Shi W-X. Psychostimulants affect dopa-
behavior therapy in binge-eating disorder. J Clin Psychiatry. mine transmission through both dopamine transporter-dependent
2007;68:1324–1332. and independent mechanisms. Eur J Pharmacol. 2015;764:562–570.
7. McElroy SL. Pharmacologic treatments for binge-eating disorder. J 27. Sotnikova TD, Caron MG, Gainetdinov RR. Trace amine-associated
Clin Psychiatry. 2017;78:14–19. receptors as emerging therapeutic targets. Mol Pharmacol.
8. U.S. Food and Drug Organization. Drugs@FDA: FDA approved drug 2009;76:229–235.
products [Internet]. 2017 [cited 2017 Sep 3]. Available from: https:// 28. Borowsky B, Adham N, Jones KA, et al. Trace amines: identification
www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=over of a family of mammalian G protein-coupled receptors. Proc Natl
view.process&ApplNo=021977. Acad Sci USA. 2001;98:8966–8971.
9. Wang G-J, Geliebter A, Volkow ND, et al. Enhanced striatal dopa- 29. Ferragud A, Howell AD, Moore CF, et al. The trace amine-associated
mine release during food stimulation in binge eating disorder. receptor 1 agonist RO5256390 blocks compulsive, binge-like eating
Obesity. 2011;19:1601–1608. in rats. Neuropsychopharmacology. 2017;42:1458–1470.
10. Vickers SP, Goddard S, Brammer RJ, et al. Investigation of impulsiv- 30. Adler LA, Alperin S, Leon T, et al. Pharmacokinetic and pharmaco-
ity in binge-eating rats in a delay-discounting task and its preven- dynamic properties of lisdexamfetamine in adults with attention-
tion by the d-amphetamine prodrug, lisdexamfetamine. J deficit/hyperactivity disorder. J Child Adolesc Psychopharmacol.
Psychopharmacol. 2017;31:784–797. 2017;27:196–199.
11. Galanti K, Gluck ME, Geliebter A. Test meal intake in obese binge 31. Krishnan S, Zhang Y. Relative bioavailability of lisdexamfetamine
eaters in relation to impulsivity and compulsivity. Int J Eat Disord. 70-mg capsules in fasted and fed healthy adult volunteers and in
2007;40:727–732. solution: a single-dose, crossover pharmacokinetic study. J Clin
12. Schag K, Teufel M, Junne F, et al. Impulsivity in binge eating Pharmacol. 2008;48:293–302.
disorder: food cues elicit increased reward responses and disinhibi- 32. Haffey MB, Buckwalter M, Zhang P, et al. Effects of omeprazole on
tion. Tomé D, editor. PLoS One. 2013;8:e76542. the pharmacokinetic profiles of lisdexamfetamine dimesylate and
13. Kessler RM, Hutson PH, Herman BK, et al. The neurobiological basis extended-release mixed amphetamine salts in adults. Postgrad
of binge-eating disorder. Neurosci Biobehav Rev. 2016;63:223–238. Med. 2009;121:11–19.
•• A review of the known neurobiology underlying binge eating 33. Adler LA, Alperin S, Leon T, et al. Pharmacokinetic and pharmaco-
disorder. dynamic properties of lisdexamfetamine in adults with attention-
14. McElroy SL, Mitchell JE, Wilfley D, et al. Lisdexamfetamine dimesy- deficit/hyperactivity disorder.
late effects on binge eating behaviour and obsessive-compulsive 34. Roberts JK, Cook SF, Stockmann C, et al. A population pharmaco-
and impulsive features in adults with binge eating disorder. Eur Eat kinetic analysis of dextroamphetamine in the plasma and hair of
Disord Rev. 2016;24:223–231. healthy adults. Clin Drug Investig. 2015;35:633–643.
• Secondary outcome results from double-blind randomized 35. De Baerdemaeker LE, Mortier EP, Struys MM. Pharmacokinetics in obese
controlled trials. patients. Contin Educ Anaesthesia Crit Care Pain. 2004;4:152–155.
15. National Institutes of Health, U.S. National Library of Medicine. 36. Baggot JD, Davis LE, Neff CA. Extent of plasma protein binding of
VYVANSE- lisdexamfetamine dimesylate capsule VYVANSE- lisdex- amphetamine in different species. Biochem Pharmacol.
amfetamine dimesylate tablet, chewable [Internet]. 2017 [cited 1972;21:1813–1816.
2017 Sep 3]. Available from: https://dailymed.nlm.nih.gov/dai 37. Asatoor AM, Galman BR, Johnson JR, et al. The excretion of dex-
lymed/drugInfo.cfm?setid=704e4378-ca83-445c-8b45- amphetamine and its derivatives. Br J Pharmacol Chemother.
3cfa51c1ecad. 1965;24:293–300.
16. Heal DJ, Smith SL, Gosden J, et al. Amphetamine, past and present– 38. Stein MA, McGough JJ. The pharmacogenomic era: promise for
a pharmacological and clinical perspective. J Psychopharmacol. personalizing attention deficit hyperactivity disorder therapy.
2013;27:479–496. Child Adolesc Psychiatr Clin N Am. 2008;17:475–490,xi–xii.
17. Sharman J, Pennick M. Lisdexamfetamine prodrug activation by 39. Moody DE, Ruangyuttikarn W, Law MY. Quinidine inhibits in vivo
peptidase-mediated hydrolysis in the cytosol of red blood cells. metabolism of amphetamine in rats: impact upon correlation
Neuropsychiatr Dis Treat. 2014;10:2275–2280. between GC/MS and immunoassay findings in rat urine. J Anal
18. Pennick M. Absorption of lisdexamfetamine dimesylate and its Toxicol. 14:311–317.
enzymatic conversion to d-amphetamine. Neuropsychiatr Dis 40. Markowitz JS, Patrick KS. Pharmacokinetic and pharmacodynamic
Treat. 2010;6:317–327. drug interactions in the treatment of attention-deficit hyperactivity
19. Pennick M. Metabolism of the prodrug lisdexamfetamine dimesy- disorder. Clin Pharmacokinet. 2001;40:753–772.
late in human red blood cells from normal and sickle cell disease 41. Ermer J, Corcoran M, Martin P. Lisdexamfetamine dimesylate effects
donors. J Drug Assess. 2013;2:17–20. on the pharmacokinetics of cytochrome P450 substrates in healthy
238 K. WARD AND L. CITROME

adults in an open-label, randomized, crossover study. Drugs R D. week clinical trial in adults with attention-deficit/hyperactivity dis-
2015;15:175–185. order. J Clin Psychiatry. 2009;70:1652–1661.
42. Comiran E, Kessler FH, Fröehlich PE, et al. Lisdexamfetamine: a 56. Wigal T, Brams M, Gasior M, et al. Randomized, double-blind,
pharmacokinetic review. Eur J Pharm Sci. 2016;89:172–179. placebo-controlled, crossover study of the efficacy and safety of
• A recent review of lisdexamfetamine pharmacokinetics. lisdexamfetamine dimesylate in adults with attention-deficit/hyper-
43. Weyler W, Salach JI. Purification and properties of mitochondrial activity disorder: novel findings using a simulated adult workplace
monoamine oxidase type A from human placenta. J Biol Chem. environment design. Behav Brain Funct. 2010;6:34.
1985;260:13199–13207. 57. Gasior M, Hudson J, Quintero J, et al. A phase 3, multicenter, open-
44. Volpi-Abadie J, Kaye AM, Kaye AD. Serotonin syndrome. Ochsner J. label, 12-month extension safety and tolerability trial of lisdexam-
2013;13:533–540. fetamine dimesylate in adults with binge eating disorder. J Clin
45. Krishnan SM, Pennick M, Stark JG. Metabolism, distribution and Psychopharmacol. 2017;37:315–322.
elimination of lisdexamfetamine dimesylate: open-label, single-cen- • Results from a 12-month open-label extension study.
tre, phase I study in healthy adult volunteers. Clin Drug Investig. 58. Heal DJ, Buckley NW, Gosden J, et al. A preclinical evaluation of the
2008;28:745–755. discriminative and reinforcing properties of lisdexamfetamine in
46. Ermer J, Corcoran M, Lasseter K, et al. A single-dose, open-label comparison to d-amfetamine, methylphenidate and modafinil.
study of the pharmacokinetics, safety, and tolerability of lisdexam- Neuropharmacology. 2013;73:348–358.
fetamine dimesylate in individuals with normal and impaired renal 59. Jasinski DR, Krishnan S. Abuse liability and safety of oral lisdexam-
function. Ther Drug Monit. 2016;38:546–555. fetamine dimesylate in individuals with a history of stimulant
47. Davis C, Levitan RD, Kaplan AS, et al. Dopamine transporter gene (DAT1) abuse. J Psychopharmacol. 2009;23:419–427.
associated with appetite suppression to methylphenidate in a case– 60. Jasinski DR, Krishnan S. Human pharmacology of intravenous lis-
control study of binge eating disorder. Neuropsychopharmacology. dexamfetamine dimesylate: abuse liability in adult stimulant abu-
2007;32:2199–2206. sers. J Psychopharmacol. 2009;23:410–418.
48. Hart AB, de Wit H, Palmer AA. Genetic factors modulating the 61. Ermer JC, Dennis K, Haffey MB, et al. Intranasal versus oral admin-
response to stimulant drugs in humans. Curr Top Behav Neurosci. istration of lisdexamfetamine dimesylate. Clin Drug Investig.
2012;12:537–577. 2011;31:357–370.
49. McElroy SL, Hudson JI, Mitchell JE, et al. Efficacy and safety of 62. Dolder PC, Strajhar P, Vizeli P, et al. Pharmacokinetics and pharma-
lisdexamfetamine for treatment of adults with moderate to severe codynamics of lisdexamfetamine compared with D-amphetamine
binge-eating disorder. JAMA Psychiatry. 2015;72:235. in healthy subjects. Front Pharmacol. 2017;8:617.
•• Results from the phase II trial pivotal for FDA approval to 63. Shire LLC. Product Monograph Vyvanse [Internet]. 2017 [cited 2017
expand lisdexamfetamine indications to include binge eating Sep 13]. Available from: http://www.shirecanada.com/-/media/
disorder. shire/shireglobal/shirecanada/pdffiles/productinformation/
50. McElroy SL, Hudson J, Ferreira-Cornwell MC, et al. Lisdexamfetamine vyvanse-pm-en.pdf.
dimesylate for adults with moderate to severe binge eating disorder: 64. Ministry of Health Israel. ‫ מאגר תרופות‬- ‫[ משרד הבריאות‬Internet].
results of two pivotal phase 3 randomized controlled trials. 2017 [cited 2017 Sep 13]. Available from: https://www.old.health.
Neuropsychopharmacology. 2016;41:1251–1260. gov.il/units/pharmacy/trufot/PerutTrufa.asp?Reg_Number=15320
•• Results from the phase III trials pivotal for FDA approval to 34001 00&safa=h.
expand lisdexamfetamine indications to include binge eating 65. Ágh T, Pawaskar M, Nagy B, et al. The cost effectiveness of lisdex-
disorder. amfetamine dimesylate for the treatment of binge eating disorder
51. Guerdjikova AI, Mori N, Blom TJ, et al. Lisdexamfetamine dimesy- in the USA. Clin Drug Investig. 2016;36:305–312.
late in binge eating disorder: a placebo controlled trial. Hum 66. Citrome L. Lisdexamfetamine for binge eating disorder in adults: a
Psychopharmacol Clin Exp. 2016;31:382–391. systematic review of the efficacy and safety profile for this newly
52. Hudson JI, McElroy SL, Ferreira-Cornwell MC, et al. Efficacy of approved indication - what is the number needed to treat, number
lisdexamfetamine in adults with moderate to severe binge-eating needed to harm and likelihood to be helped or harmed? Int J Clin
disorder. JAMA Psychiatry. 2017;74:903–910. Pract. 2015;69:410–421.
• Results from the maintenance study comparing lisdexamfeta- 67. Sheehan DV, Herman BK. The psychological and medical factors
mine vs placebo for a 26-week, double-blind, randomized associated with untreated binge eating disorder. Prim Care
withdrawal period following a 12-week open label phase. Companion CNS Disord. 2015;17. doi: 10.4088/PCC.14r01732
53. McElroy SL, Hudson JI, Gasior M, et al. Time course of the effects of 68. Hudson JI, Lalonde JK, Coit CE, et al. Longitudinal study of the
lisdexamfetamine dimesylate in two phase 3, randomized, double- diagnosis of components of the metabolic syndrome in indivi-
blind, placebo-controlled trials in adults with binge-eating disorder. duals with binge-eating disorder. Am J Clin Nutr. 2010;91:1568–
Int J Eat Disord. 2017;50:884–892. 1573.
54. Adler LA, Goodman DW, Kollins SH, et al. Double-blind, placebo- 69. Bulik CM, Reichborn-Kjennerud T. Medical morbidity in binge eat-
controlled study of the efficacy and safety of lisdexamfetamine ing disorder. Int J Eat Disord. 2003;34(Suppl):S39–S46.
dimesylate in adults with attention-deficit/hyperactivity disorder. J 70. Barnes RD, Boeka AG, McKenzie KC, et al. Metabolic syndrome in
Clin Psychiatry. 2008;69:1364–1373. obese patients with binge-eating disorder in primary care clinics: a
55. Adler LA, Weisler RH, Goodman DW, et al. Short-term effects of cross-sectional study. Prim Care Companion CNS Disord. 2011;13.
lisdexamfetamine dimesylate on cardiovascular parameters in a 4- pii: PCC.10m01050. doi: 10.4088/PCC.10m01050

You might also like