Novel Pharmacological

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

HHS Public Access

Author manuscript
Depress Anxiety. Author manuscript; available in PMC 2021 August 01.
Author Manuscript

Published in final edited form as:


Depress Anxiety. 2020 August ; 37(8): 747–759. doi:10.1002/da.23038.

Novel Pharmacological Treatments for Generalized Anxiety


Disorder: Pediatric Considerations
A. Irem Sonmez, MD1, Ammar Almorsy, MBBCh1, Laura B. Ramsey, PhD2, Jeffrey R.
Strawn, MD3, Paul E. Croarkin, DO, MSCS1,*
1Department of Psychiatry and Psychology Mayo Clinic, Rochester, Minnesota, USA;
2Divisionof Research in Patient Services and Clinical Pharmacology, Cincinnati Children’s
Author Manuscript

Hospital Medical Center, University of Cincinnati College of Medicine Cincinnati, Ohio, USA;
3Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati,
Ohio, USA;

Abstract
Pediatric anxiety disorders such as generalized anxiety disorder (GAD) are common, impairing,
and often undertreated. Moreover, many youth do not respond to standard, evidence-based
psychosocial or psychopharmacologic treatment. An increased understanding of the gamma-
aminobutyric acid (GABA) and glutamate neurotransmitter systems has created opportunities for
novel intervention development for pediatric GAD. This review examines potential candidates for
pediatric GAD: eszopiclone, riluzole, Eglumegad (LY354740), pimavanserin, agomelatine. The
Author Manuscript

pharmacology, preclinical data, clinical trial findings and known side effects of these compounds
are reviewed, particularly with regard to their potential therapeutic relevance to pediatric GAD.
Notwithstanding numerous challenges, some of these agents represent potential candidate drugs
for pediatric GAD. Further treatment development studies of agomelatine, eszopiclone,
pimavanserin and riluzole for pediatric GAD also have the prospect of informing the
understanding of GABAergic and glutamatergic function across development.

Keywords
Agomelatine; Anxiety; Drug Development; Eglumegad (LY354740); Eszopiclone; Generalized
Anxiety Disorder; Pediatric; Pimavanserin; Riluzole
Author Manuscript

Introduction
Pediatric anxiety disorders affect up to 20% of youth and contribute to social impairment,
poor academic performance, and family stress (Essau et al., 2000). Anxiety disorders

*
Reprints: Paul E. Croarkin, DO, MSCS, Department of Psychiatry and Psychology, Mayo Clinic, 200 First St SW, Rochester, MN
55905, croarkin.paul@mayo.edu, Telephone: (507) 293-2557, Fax: (507) 293-3933.
Data Sharing:
Data Sharing is not applicable to this article as no new data were created or analyzed in this study.
Conflicts of Interest
The authors report no financial or other relationship relevant to the subject of this article.
Sonmez et al. Page 2

increase the risk of developing depressive disorders (Pine, 1999), substance use disorders,
Author Manuscript

and suicidality (Asselmann et al., 2018; Husky et al., 2012). Generalized anxiety disorder
(GAD) is one of the more common anxiety disorders in childhood and is characterized by
diffuse, difficult-to-control anxiety that typically emerges during adolescence (Beesdo-Baum
& Knappe, 2012).

Putative variations in the serotonin transporter gene (5-HTT) may predispose children and
adolescents to developing GAD (Tsuang et al., 2004). However, the neurochemical basis of
GAD in children and adolescents is likely more wide-ranging and complex with
contributions from serotonin, norepinephrine, glutamate, and GABA systems. This
complexity creates clinical challenges in precision medicine approaches to tailoring
treatment in youth with GAD but also presents immense research opportunities for
developing new pharmacological treatments. Neurodevelopmental considerations are
important in considering the pathophysiology of anxiety as the glutamatergic and
Author Manuscript

GABAergic neurotransmitter systems also undergo substantial changes, potentially


facilitating more heterogeneity in the clinical picture, pathophysiology, and pharmacological
response in childhood GAD (Pine, 1999).

Current, evidence-based pharmacologic treatments for GAD as main diagnosis in children


and adolescents are limited to selective serotonin reuptake inhibitors (SSRIs) and serotonin
norepinephrine reuptake inhibitors (SNRIs) (Dobson et al., 2019). Duloxetine is the only
antidepressant approved by the FDA for pediatric GAD (Jeffrey R Strawn et al., 2015).

Despite the promise of novel therapeutics, there are significant challenges to developing
more effective treatments. These challenges relate not only to the fluctuating course of
anxiety disorders but to developmental factors and inherent limitations of clinical trials
Author Manuscript

design. In youth, anxiety symptoms—like depressive symptoms—tend to fluctuate over time


(Costello et al., 2005). Thus, a group of anxious youth may have symptoms and variance of
symptom measures as well as the autocorrelation of symptoms that are determined by non-
treatment. From a clinical trial design standpoint, even with current statistical approaches,
some degree of stationarity is necessary to detect treatment-related improvement. Treatments
are also largely borrowed by those that have demonstrated efficacy in adults with anxiety
disorders (J. R. Strawn et al., 2018).This approach assumes that the pathophysiology and by
extension the neuropharmacology of these disorders is the same in pediatric and adult
populations. However, differences in neurocircuitry have been observed in anxious adults
relative to anxious youth. Measures of anxiety symptoms that have been used in pediatric
(and adult studies) are encumbered by issues related to unidimensionality and over
representation of some symptoms (e.g., somatic symptoms) relative to psychic symptoms or
Author Manuscript

functional impairment (e.g., relationship dysfunction and avoidant behavior). Importantly,


problems introduced by the unidimensionality of these measures are amplified in the
pediatric population that often has greater co-morbidity (Kendall et al., 2010).

This review examined potential investigational pharmacologic treatments for pediatric GAD
with the objective of informing related, clinical research, and drug development. Embase
and Medline from 2008 to 2019 were searched with the assistance of a research librarian
using keywords listed for this review. Studies and manuscripts that were published in

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 3

English were included. The reference list was finalized in March 2019 through review of
Author Manuscript

initial literature search and discussion among all authors. The reference list of each
manuscript was reviewed for additional potential references. Additional manuscripts were
reviewed and included by PEC and JS as deemed necessary for a comprehensive manuscript.

(1) Rationale for GABA and glutamate neurotransmitter systems for pediatric GAD
In pre-clinical models, stress enhances excitatory neurotransmission, mediated by elements
of the glutamatergic neurotransmission system and chronic stress increases vulnerability to
developing anxiety disorders (Simon & Gorman, 2006). Glutamatergic tone in the anterior
cingulate cortex correlates with the severity of anxiety symptoms in adolescents with GAD
(Jeffrey R Strawn et al., 2014). Thus, glutamate-modulating agents may have a role in
treating severe anxiety disorders. Acute administration of N-Methyl-D-Aspartic acid
(NMDA) receptor antagonists has anxiolytic effects in preclinical and clinical models (Javitt,
Author Manuscript

2004; Plaznik et al., 1994). Conventional antidepressants and NMDA antagonists decrease
glutamatergic neurotransmission without histologic changes in the neuron which may be
associated with remission from depression or anxiety disorders via restoration of a basal rate
of neurogenesis (Duman, 2002; Jacobs et al., 2000; Manji et al., 2003; Santarelli et al.,
2003). Notably, prior work suggests that glutamatergic and GABAergic balances may
regulate the expression of BDNF (Marmigère et al., 2003).

(2) Promising Drugs


Eszopiclone—Eszopiclone, a γ-aminobutyric acid (GABAA) receptor agonist, is rapidly
absorbed after oral administration and extensively metabolized by CYP3A4 and CYP2E1.
N-desmethylzopiclone, the pharmacologically active primary metabolite, is distributed in
brain and among other tissues and then eliminated via urine and saliva. In pediatric studies
Author Manuscript

of eszopiclone, body weight significantly affects clearance and volume of distribution


(Maier et al., 2011; Sunovian Pharmaceuticals Inc., 2012). With a 7.5 mg dose, the time to
peak concentration (TMAX) is 1 hour and the t½ is 6 hours. As eszopiclone is weakly protein
bound, drug interactions related to protein binding are not concerning. CYP3A4 and
CYP2E1 play a role in the metabolism of eszopiclone. Eszopiclone does not inhibit CYP
1A2, 2A6, 2C9, 2C19, 2D6, 2E1, or 3A4 (Sunovion Pharmaceuticals Inc., 2014).

In pre-clinical rodent models of insomnia, eszopiclone decreases acetylcholine release in


sleep promoting regions of pontine reticular formation (Hambrecht-Wiedbusch et al., 2010)
and in the amygdala suggesting its potential in regulation of fear and anxiety as well as
learning, memory, sleep and autonomic control (Hambrecht-Wiedbusch et al., 2014). In
terms of anxiety, low dose eszopiclone (1–10 mg/kg) had promising effects controlling
anxiety and anxiety related insomnia on rats (Huang et al., 2010). Eszopiclone may also
Author Manuscript

facilitate neurogenesis. Co-administration of eszopiclone with fluoxetine significantly


increased survival of marked newborn cells in the adult rat dorsal hippocampus (Su et al.,
2009). In another trial twice daily eszopiclone treatment strongly enhanced the survival of
proliferating cells in hippocampal dentate gyrus cells in rats (Methippara et al., 2010). This
effect may be due to responsiveness of adult born hippocampal cells GABAergic stimulation
(Nakamichi et al., 2009). Neurogenesis involves proliferation of neural progenitors, survival

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 4

of newly proliferating cells, and differentiation to neural phenotypes, all of which may be
Author Manuscript

involved in the pathophysiology and treatment of anxiety (Revest et al., 2009).

Eszopiclone is currently indicated for insomnia in adults and 6 randomized controlled trials
supporting this indication (Sunovion Pharmaceuticals Inc., 2014). In one of these trials add-
on eszopiclone to escitalopram resulted in greater improvement in HAM-A scores at each
week, even when the insomnia item was removed, and also enhanced the possibility of
response and remission in a 10 week long insomnia trial. Despite discontinuation of
eszopiclone, treatment differences in anxiety measures were maintained (Pollack et al.,
2008).

In a more recent trial comparing cognitive behavioral therapy (CBT) and eszopiclone
combined therapy versus eszopiclone alone in adults with sleep disorder (N=29), greater
improvement in self-reported anxiety (self-rating anxiety scale, SAS) was observed in the
Author Manuscript

combined group (Zhang et al., 2018). Another study of perimenopausal women with
impaired daytime functioning due to insomnia as well as some depressive and/or anxiety
symptoms demonstrated improvement in insomnia as well as depression and anxiety
symptoms in eszopiclone group compared to placebo (Joffe et al., 2010).

There are no pediatric trials of eszopiclone in GAD. One study of youth with attention
deficit hyperactivity disorder (ADHD) and insomnia (age 6–17 years, N=486) failed to
detect differences in efficacy for insomnia and safety between eszopiclone and placebo
(Sangal et al., 2014).

In general, eszopiclone is well tolerated (Joffe et al., 2010; Pollack et al., 2008; Zhang et al.,
2018). Most frequently reported adverse event was metallic taste (Joffe et al., 2010; Pollack
et al., 2008); other adverse events were reported as somnolence, headache, dry mouth,
Author Manuscript

dizziness, jitteriness and heart palpitations (Greenblatt & Zammit, 2012). There may be an
impairment of driving in the initial waking hours(Greenblatt & Zammit, 2012). Among
youth with ADHD, the most frequent adverse events were headache, dysgeusia and
dizziness (Sangal et al., 2014).

In terms of safety concerns for pediatric GAD studies, eszopiclone is a schedule IV


controlled substance. However eszopiclone may still be a promising candidate for study and
development in pediatric GAD, particularly given concerns about tolerability limitations
associated with benzodiazepines in pediatric GAD and other anxiety disorders (Dobson et
al., 2019).

Riluzole—The exact mechanism of action of riluozole is unknown. Theories postulate that


Author Manuscript

riluzole inhibits glutamate release considering its role in amyotrophic lateral sclerosis
(ALS). But there might be other processes involved. Riluzole inhibits glutamic acid release
in cultured neurons, from brain slices, and from corticostriatal neurons in vivo (Doble, 1996)
and decreases release of glutamate from synaptic terminals throughout the CNS (Prakriya &
Mennerick, 2000). Riluzole also blocks some of the postsynaptic effects of glutamic acid by
noncompetitive blockade of N-methyl-D-aspartate (NMDA) receptors(Doble, 1996),
increases glutamate reuptake (Frizzo et al., 2004), include inhibition of voltage-dependent

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 5

sodium channels in CNS neurons (Urbani & Belluzzi, 2000), enhances of hippocampal
Author Manuscript

alpha-amino-3-hydrosy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit


(GluR1 and GluR2) expression (Du et al., 2007). Riluzole stimulates growth factor
synthesis, including BDNF (Katoh-Semba et al., 2002; Mizuta et al., 2001); promotes
neurogenesis, neurite branching, and neurite outgrowth (Shortland et al., 2006).

Riluzole is well-absorbed and its bioavailability is approximately 60%. Riluzole is highly


protein-bound (90%) and has a t½ 12 hours. Metabolism is mostly hepatic and consists of
hydroxylation mainly and glucuronidation. CYP1A2 is the principal isoenzyme responsible
for N-hydroxylation. Primary excretion pathways are urine (90%) and feces (5%)(ITF
Pharma Inc., 2018; Sanofi-Aventis U.S. LLC., 2008).

In preclinical models of anxiety (elevated plus maze, light/dark and open field tests), the
anxiolytic effects of riluzole are comparable to diazepam (Sugiyama et al., 2012). Riluzole
Author Manuscript

infusion into the basolateral amygdala in rats improved recognition memory and resulted in
anxiolytic effect in the elevated plus-maze test and decreased freezing time in contextual
fear conditioning test with foot shocks (Sugiyama et al., 2017; Sugiyama et al., 2018).
Riluzole attenuated anxiety-like behaviors in mice that were pre-treated with veratrine, a
sodium channel activator and anxiety inducer, in the open field test (Ohashi et al., 2015).

Other preclinical studies are available in various models including traumatic brain injury,
spinal cord injury, Huntington’s disease, ischemia and ALS showing neuroprotective effects
of riluzole(Wahl & Stutzmann, 1999). In another trial, riluzole stimulated human platelets to
release BDNF, which is a contributor molecule in brain development, neuronal plasticity and
neuronal survival (Turck & Frizzo, 2015).

One open-label study suggested that riluzole (100 mg/day) in adults with GAD may reduce
Author Manuscript

anxiety and is associated with increased hippocampal N-acetylaspartate increase (as a


marker of neuronal integrity) (Mathew et al., 2005). Eighteen GAD patients (seven of the
participants with comorbid panic disorder) received riluzole monotherapy and 8 patients
(44%) remitted based on HAM-A score. Most of comorbid panic disorder patients (5 out of
7) completed the trial and all had significant improvement in PDSS (Abdallah et al., 2013;
Mathew et al., 2008).

Riluzole’s side effect profile was favorable and preliminary results regarding riluzole for the
treatment of severe mood, anxiety and impulsive disorders were encouraging (Zarate &
Manji, 2008). In trials of adults with GAD, the most common adverse events during the trial
were insomnia/sleep disturbance, nausea, abdominal discomfort, sedation, and dry mouth.
No serious adverse events were noted. One patient who completed the trial required a dose
Author Manuscript

reduction of riluzole due to sedation. Transient increase in alanine aminotransferase ranging


from 1.1 to 1.8 times the upper limit of normal was observed; for some patients, it
normalized within 2 weeks while receiving riluzole and remained normal at endpoint; and
for the remaining patients, aminotransferase values normalized after discontinuation of
riluzole. No patient exhibited symptoms of hepatic toxicity (Mathew et al., 2005; Mathew et
al., 2008).

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 6

The safety of riluzole in pediatric patients has been examined in two OCD trials. Six
Author Manuscript

treatment resistant OCD patients received riluzole for 12 weeks in an open label trial. All
subjects reached a target dose of 100 mg daily without limitation of adverse events aside
from one patient who experienced drowsiness. Increases in lactate dehydrogenase and liver
transaminases were observed with riluzole and both resolved on their own despite continued
riluzole treatment (Grant et al., 2007). An RCT with 60 pediatric OCD patients (mean age
14 years) did not demonstrate a significant difference between riluzole and placebo with
respect to adverse events(Grant et al., 2014). One serious adverse event (pancreatitis) was
recorded in the riluzole group which resolved. Five other patients with asymptomatic
elevations of transaminases were recorded. There were no differences in primary and
secondary outcome measures among the riluzole and placebo groups. This patient sample
was treatment refractory and taking a variety of other medications concurrently (Grant et al.,
2014).
Author Manuscript

In considering safety, riluzole may be promising agent to study in pediatric GAD. The safety
data from the prior pediatric OCD clinical trial are promising. Although efficacy was not
demonstrated for OCD, this sample was highly treatment refractory and taking variable
concomitant medications (Grant et al., 2014). Further study as monotherapy in pediatric
GAD may be worth considering.

Eglumegad (LY354740)—Eglumegad (LY354740) is a selective agonist of Group II


mGluRs (Schoepp et al., 2003). The mGlu2/3 belongs to Group II mGluRs that are located
mainly presynaptically. Their major role is to inhibit neurotransmitter release (Nicoletti et
al., 2011). They have an established role in synaptic plasticity regulation and in drug
addiction (Lindsley et al., 2016).
Author Manuscript

The clinical use of Eglumegad (LY354740) is limited by its poor gastrointestinal absorption
and poor bioavailability (3–5%) (Rorick-Kehn et al., 2006). To improve Eglumegad
(LY354740) absorption and further assess the therapeutic effects of mGlu2/3 agonists, a
pharmacologically inactive peptidyl prodrug (L-alanine) form of the active compound
Eglumegad (LY354740), named LY544344, was developed. LY544344 is an Eglumegad
(LY354740) prodrug that increases Eglumegad (LY354740) bioavailability. In rodents, oral
administration of LY544344 improves its bioavailability by 10-fold compared to Eglumegad
(LY354740) and produces comparable behavioral effects observed with parenteral
administration of Eglumegad (LY354740) (Rorick-Kehn et al., 2006). Pharmacokinetic data
are currently available for Eglumegad (LY354740) in animals (Johnson et al., 2002). In rats
and dogs, oral Eglumegad (LY354740) has a linear pharmacokinetic profile from 30 to 1000
mg/kg. The bioavailability is 10% and 45 % respectively. The primary elimination route was
Author Manuscript

urine in both animals. Metabolism of Eglumegad (LY354740) was evaluated in vitro using
rat and dog liver microsomes and rat liver slices. Neither rats nor dogs metabolized
Eglumegad (LY354740). In summary, Eglumegad (LY354740) is poorly absorbed in rats,
moderately absorbed in dogs and rapidly excreted as unchanged drug in the urine (Johnson
et al., 2002; Pilc, 2003).

Rat models of anxiety have supported a mechanistic role for an imbalance between
glutamatergic and GABAergic tone in panic attacks(Johnson & Shekhar, 2012). Group II

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 7

metabotropic glutamate (mGlu) receptors are located in areas that are thought to be
Author Manuscript

implicated in panic networks such as dorsomedial hypothalamic regions and dorsal


periaqueductal gray areas (Johnson et al., 2013). Eglumegad (LY354740) has anxiolytic
activity in many rodent stress and anxiety models, including fear-potentiated startle, elevated
plus maze, lactate-induced panic, and stress-induced hyperthermia (Helton et al., 1998;
Kłodzińska et al., 1999; Shekhar & Keim, 2000; Spooren et al., 2002). Eglumegad
(LY354740) had similar efficacy to benzodiazepines in animal models, but had no evidence
for central nervous system (CNS) depression of motor function, learning, and memory
(Helton et al., 1998; Tizzano et al., 2002). Pretreatment with Eglumegad (LY354740)
inhibits panic-like response without producing sedation in rats (Shekhar & Keim, 2000).
Neuroprotective characteristics of mGlu2/3 receptor agonists were also shown in animal
models of cerebral ischemia(Bond et al., 2000; Bond et al., 1999) and mGlu2/3 knock out
mice(Corti et al., 2007).
Author Manuscript

A double-blind trial in patients with panic disorder revealed no statistically significant


differences between Eglumegad (LY354740) and placebo in reducing Panic Disorder
Severity Scale (PDSS) scores and the clinical global impression (CGI) (Bergink &
Westenberg, 2005). However, in infusion studies with cholecystokinin tetrapeptide (CCK-4)
—a reliable panicogen—suggested benefit for the pro-drug LY544344 (which has higher
bioavailability than Eglumegad [LY354740])(Kellner et al., 2005).

One double-blind trial for adults with GAD (N=738)(Michelson et al., 2005) that
randomized patients to Eglumegad (LY354740) (100 or 200 mg BID), placebo, or lorazepam
(4–5 mg daily) demonstrated that Eglumegad (LY354740) had greater efficacy compared to
placebo and comparable reductions to lorazepam over 5 weeks. The tolerability profile of
Eglumegad (LY354740) was superior to lorazepam (Michelson et al., 2005). Reported side
Author Manuscript

effects can be found in Table 1. A subsequent study of LY544344 in adults with GAD
(Dunayevich et al., 2008) randomized patients to LY544344 (8 or 16 mg BID) or placebo;
however, secondary to a preclinical concern for treatment-emergent seizures (dog at 60
mg/kg, and in rat and mouse studies at doses 70 mg/kg) the study was terminated and active
patients were discontinued after only 29 patients (26.1%) completed the double-blind
treatment phase and entered the single-blind placebo, drug discontinuation assessment
phase. Primary efficacy analysis showed a positive treatment effect for patients in the
LY544344 treatment groups relative to placebo treatment (p=0.027). Patients who received
LY544344 16 mg b.i.d., but not 8 mg b.i.d., had greater mean improvement from baseline on
the HAM-A total score than those who received placebo (Dunayevich et al., 2008).

There are no trials of LY544344 in pediatric patients. Further study in children and
Author Manuscript

adolescents could prove challenging as the adult clinical trial was halted for preclinical
concerns related to seizure risk.

(3) Other Potential Drugs


Agomelatine—Agomelatine, a selective melatonergic MT1 and MT2 receptor agonist may
have hypnotic effects (Fornaro et al., 2010). Agomelatine is also an antagonist at
serotonergic 5-hydroxytryptamine 2C (5-HT2C) receptors that are implicated in underlying
neurobiology of anxiety related behaviors(De Berardis et al., 2013). Additionally, the

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 8

anxiolytic activity of agomelatine may be related to its modulation of GABAergic pathways


Author Manuscript

and 5-HT2C antagonism (De Berardis et al., 2013; Fornaro et al., 2010; Loiseau et al., 2006).

It is rapidly absorbed (>75%) after oral administration, although, extensive first-pass


metabolism lowers its absolute bioavailability to <5% (Zupancic & Guilleminault, 2006).
Thus, intranasal and transdermal formulations that have been developed may circumvent its
extensive first-pass metabolism (Fatouh et al., 2017; Said et al., 2017). Agomelatine is
mainly metabolized by hepatic CYP450 CYP1A2 and CYP2C9 to its main metabolites, 3-
hydroxy-agomelatine and 7-desmethyl-agomelatine. Agomelatine is primarily eliminated by
urinary excretion of metabolites and has a mean half-life (t½) of approximately 2 h.

Preclinical studies demonstrated anxiolytic effects of agomelatine under basal conditions


and in fear conditioning models (Guardiola‐Lemaitre et al., 2014). Agomelatine has
demonstrated anxiolytic effects in a wide variety of preclinical models of anxiety including
Author Manuscript

prenatal restraint stress (Morley-Fletcher et al., 2011), hypercortisolemic mice (Rainer et al.,
2012), social defeat (Tuma et al., 2005), and social isolation rearing [SIR] model (Regenass
et al., 2018). Additionally, agomelatine increased hippocampal brain-derived neurotrophic
factor (BDNF) levels in another rat model (Lu et al., 2018; Marmigère et al., 2003). Chronic
(3 weeks) agomelatine administration increased cell proliferation and neurogenesis in the
ventral dentate gyrus, a region implicated in fear-related behaviors. Chronic agomelatine
treatment facilitated neuronal survival throughout the dentate gyrus, although this effect
appears to be restricted to developing neurons (Banasr et al., 2006). Another trial with light
stress-exposed rat brains revealed that agomelatine stimulated neurogenesis and prevented
apoptosis (Yucel et al., 2016). Agomelatine appeared to protect the rat brain from cerebral
ischemia and reperfusion injury by suppressing apoptosis (Chumboatong et al., 2017). Also,
chronic agomelatine treatment prevented transcription of the interleukin–1β and
Author Manuscript

metabotropic glutamatergic receptor (mGluR) genes (Rossetti et al., 2018).

In three randomized, placebo-controlled, clinical trials, agomelatine was superior to placebo


in adults with GAD (Stein et al., 2017; Stein et al., 2014; Stein et al., 2008) and, in another
RCT, was more tolerable, but equally effective, compared to escitalopram in adults with
GAD (Stein et al., 2018). High and low-dose (10 and 25 mg/day) agomelatine groups
outperformed the placebo group, although only the high dose group showed a robust
difference in remission rate compared to placebo (40.2 versus 12.5%) (Stein et al., 2017).
When escitalopram and agomelatine compared over the course of 12 weeks, both
significantly decreased Hamilton Anxiety Rating Scale (HAM-A) scores, however non-
inferiority of agomelatine versus escitalopram was not established (Stein et al., 2018).
Agomelatine (25–50 mg/kg) has been reported to be clinically effective and well tolerated in
Author Manuscript

adults with GAD (Buoli et al., 2017; Stein et al., 2018) but is not approved by the FDA. At
present there are no studies of agomelatine in pediatric patients.

In regards to safety, agomelatine, escitalopram, and placebo have similar tolerability in


adults with GAD (Stein et al., 2017; Stein et al., 2014; Stein et al., 2008; Stein et al., 2018).
The most frequently reported adverse events included dizziness, nausea, headache,
nasopharyngitis, diarrhea and somnolence. Adverse events leading to discontinuation were
less frequent in the agomelatine-treated patients compared to those who received placebo

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 9

and escitalopram. The most recent agomelatine study (Stein et al., 2018) reported that
Author Manuscript

agomelatine had superior tolerability compared to escitalopram with respect to headache,


nausea, insomnia, dizziness, anxiety, hyperhidrosis, and diarrhea. There were no differences
between agomelatine and escitalopram in terms of other adverse events. No suicidal
behavior was reported during the study.

Rare cases of severe and life-threatening liver injury have been reported (Friedrich et al.,
2016; Štuhec, 2013). Agomelatine is contraindicated in patients with impaired liver function
(Freiesleben & Furczyk, 2015) and pre-treatment transaminases should be evaluated with
subsequent hepatic profiles being obtained after 3, 6, 12, and 24 weeks(European Medicines
Agency, 2016). If an elevation of transaminases is detected, these exams should be repeated
within 48 h and agomelatine should be discontinued if it increases more than three times the
higher limit of normal value (European Medicines Agency, 2016).
Author Manuscript

There are safety concerns to consider for future study in children and adolescents. Liver
toxicity is a challenge to consider in further development of agomelatine for pediatric GAD.
Agomelatine-induced hepatotoxicity appears to be rare and unpredictable (Gahr et al.,
2013). Analysis of 9,234 patients who were treated with agomelatine showed that serum
transaminases increased three fold higher than the upper normal limit in 1.3 and 2.5 % of
patients treated with 25 and 50 mg of agomelatine, respectively, compared with 0.5 % for
placebo. The incidence of abnormal transaminases was low and dose dependent. No patient-
specific factors were identified regarding potential risk factors and withdrawal of
agomelatine led to rapid recovery (Perlemuter et al., 2016).

Pimavanserin—Pimavanserin, is a potent selective serotonin 5-HT2A inverse agonist with


a combination of inverse agonist and antagonist activity to a lesser extent for 5-HT2C and
Author Manuscript

sigma-1 receptors (σ1R) (ACADIA Pharmaceuticals Inc, 2016). Pimavanserin has no


affinity for the dopaminergic, adrenergic, histaminergic, or muscarinic receptors which
separates it from other antipsychotic drugs (Vanover et al., 2006).

The mean t½ for pimavanserin and its active metabolite (N-desmethylated metabolite) are
approximately 60 hours and 200 hours, respectively. Pimavanserin demonstrates dose-
proportional pharmacokinetics and is predominantly metabolized by CYP3A4 and CYP3A5
and to a lesser extent by CYP2J2, CYP2D6, and various other CYP and FMO enzymes.
CYP3A4 is responsible for the formation of its major active metabolite and in vitro data
suggest that pimavanserin does not irreversibly inhibit major hepatic and intestinal human
CYP enzymes. Elimination is both via urine and feces (ACADIA Pharmaceuticals Inc,
2016).
Author Manuscript

In rodents, activation of 5-HT2C receptors in the basolateral amygdala complex induces


anxiogenic behaviors (Campbell & Merchant, 2003; Hackler et al., 2007)and 5-HT2C
receptor antagonists have shown to have anxiolytic properties (Aloyo et al., 2009). The 5-
HT2C and 5-HT2A receptors have been implicated in the etiology and treatment of various
psychiatric disorders. Activation of 5-HT2C receptors, with agonists results in feelings of
anxiety and panic in humans (Gatch, 2003). Pimavanserin’s activity on 5-HT2A and 5-HT2C
receptors may suggest its potential on anxiety and fear conditions. Serotonin 2A receptors

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 10

are concentrated in the limbic system and are important for mediating fear. There is evidence
Author Manuscript

showing that the processing of emotionally loaded information is modulated by the 5HT2A
receptor and that the expression of these receptors related to anxiety traits (Frokjaer et al.,
2008). Individuals with higher serotonin activity tend to have more fearful personalities, and
interestingly, animals with a deficiency of 5-HT2A receptors lack normal fear reactions
(Moresco et al., 2002).

Pimavanserin has not yet been studied in clinical trials for GAD. In adults, pimavanserin and
other 5-HT2A receptor inverse agonists attenuated insomnia in few trials through increases in
slow wave sleep (Ancoli-Israel et al., 2011; Monti, 2010; Rosenberg et al., 2008; Teegarden
et al., 2008). There are no pediatric trials of pimavanserin in pediatric patients with GAD, or
any other disorder.

Overall, clinical trials for pimavanserin in Parkinson’s demonstrated an acceptable safety


Author Manuscript

profile (Bozymski et al., 2017). Prescription information reported most frequent side effects
in Parkinson patients in the pimavanserin arms in Parkinson’s disease trials and in healthy
volunteers as drowsiness, nausea, peripheral edema, confusion, hallucinations, constipation,
and gait disturbance (Vanover et al., 2007). Pimavanserin may prolong QT intervals and
concomitant administration of medications that prolong the QT interval should be avoided
(ACADIA Pharmaceuticals Inc, 2016).

The primary safety concern for development of pimavanserin is the knowledge gaps related
to its safety profile in children and adolescents. Initial and ongoing studies would need to
examine this in a systematic fashion (ACADIA Pharmaceuticals Inc, 2016). A secondary
challenge is the cost of pimavanserin—approximately $3,000/month.
Author Manuscript

Conclusion
Many children and adolescents with GAD do not respond completely to standard
interventions such as CBT, SSRIs, and SNRIs (Bushnell et al., 2019). There is an urgent
need to expand pharmacologic treatment options for pediatric GAD. Moreover, GABAergic
and glutamatergic dysfunction are implicated in the pathophysiology of GAD in children,
adolescents and adults, yet in youth, relatively few treatment studies have targeted these
systems (Figure 1). These neurotransmitter systems undergo substantial shifts in childhood
and adolescence (Benarroch, 2012; Chugani et al., 2001). This trajectory is poorly
understood in GAD but could present strategic opportunities for pharmacological
intervention, thereby sparing the need for lifelong treatment. In this regard, agomelatine,
eszopiclone, and riluzole are promising agents to consider in for clinical development in
pediatric GAD. Given that an adult clinical trial of Eglumegad (LY354740) was
Author Manuscript

discontinued in adults due to safety concerns it is unlikely that this drug will have promise
for clinical development in children (Dunayevich et al., 2008).

Previous anxiolytic drug discovery has largely focused on single targets (highly selective
agents acting on specific molecular targets). However, the notion of better and safety drugs
for GAD has not been realized. Multimodal drug candidates in development, like

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 11

agomelatine, with the intention of re-equilibrating perturbed circuits, may potentially lead to
Author Manuscript

more effective treatments (Figure 2).

Each of these candidates has unique strengths and barriers to consider for future study.
Broadly, eszopiclone and riluzole may have the most promise in the short-term for
development in pediatric GAD as there are existing safety data in children. Preclinical work
with eszopiclone and riluzole also are encouraging. Eszopiclone and riluzole also have a
favorable cost profile compared to other agents. Agomelatine may be another promising
agent to consider for pediatric GAD with respect to prior preclinical work and adult clinical
trials. Unfortunately at present there are substantial barriers to developing LY354740 and
pimavanserin for pediatric GAD related to safety, preclinical seizure evidence and cost.
Further preclinical studies of agomelatine, eszopiclone, pimavanserin and riluzole in the
context of neurodevelopment are critical to advance mechanistic understanding of anxiety in
children and adolescents and to facilitate drug discovery.
Author Manuscript

Acknowledgments:
Preparation of this manuscript was supported by the National Institutes of Health under awards R01 MH113700
(PEC) and R01 HD098757 (JRS). The content of this publication is solely the responsibility of the authors and does
not necessarily represent the official views of the National Institutes of Health. The supporters had no role in the
literature review, interpretation, or publication. We thank Anosha Zanjani for her expertise and assistance with the
creation of Figure 1. We thank Danielle Gerberi for her help with literature search.

Financial Disclosures

Dr. Croarkin receives research support from the National Institutes of Health (NIH). Dr. Croarkin has received
research grant support from Pfizer, Inc.; equipment support from Neuronetics, Inc.; and received supplies and
genotyping services from Assurex Health, Inc. for investigator-initiated studies. He is the primary investigator for a
multicenter study funded by Neuronetics, Inc. and a site primary investigator for a study funded by NeoSync, Inc.
Dr. Croarkin serves as a paid consultant for Procter & Gamble Company and Myriad Neuroscience. Dr. Ramsey has
Author Manuscript

received research support from the National Institutes of Health (NICHD) and BTG, International Ltd. Dr. Strawn
has received research support from the National Institutes of Health (NIMH/NIEHS), Patient-Centered Outcomes
Research (PCORI) , Allergan, Otzuka,and Neuronetics. He has received material support from Myriad Genetics and
receives royalties from the publication of two texts (Springer). Dr. Strawn serves as an author for UpToDate. He is
an Associate Editor for Current Psychiatry.,He has received honoraria from CMEology and Neuroscience
Educational Institute. The other authors have no disclosures or potential conflicts of interest to declare.

Abbreviations
ACh acetylcholine

ADHD attention deficit hyperactivity disorder

AMPA alpha-amino-3-hydrosy-5-methyl-4-isoxazolepropionic acid

ALS amyotrophic lateral sclerosis


Author Manuscript

BDNF brain-derived neurotrophic factor

CBT cognitive behavioral therapy

CCK-4 cholecystokinin tetrapeptide

CNS central nervous system

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 12

CYP cytochrome P450


Author Manuscript

EPS extrapyramidal symptoms

h hours

HAM-A Hamilton anxiety rating scale

HDRS-17 Hamilton depression rating scale

5-HT 5-hydroxytryptamine (serotonin) receptor

5-HTT serotonin transporter gene

5-HT2C 5-hydroxytryptamine c receptor

GABA gamma-aminobutyric acid


Author Manuscript

GAD generalized anxiety disorder

mGluR metabotropic glutamatergic receptor

M1 and M2 melatonin receptors

NMDA N-Methyl-D-aspartic acid

OCD obsessive compulsive disorder

σ1R sigma-1 receptors

SIR social isolation rearing


Author Manuscript

SNRIs serotonin norepinephrine reuptake inhibitors

SSRIs selective serotonin reuptake inhibitors

t½, half-life

References
Abdallah CG, Coplan JD, Jackowski A, Sato JR, Mao X, Shungu DC, & Mathew SJ (2013). A pilot
study of hippocampal volume and N-acetylaspartate (NAA) as response biomarkers in riluzole-
treated patients with GAD. Eur Neuropsychopharmacol, 23(4), 276–284. 10.1016/
j.euroneuro.2012.05.009 [PubMed: 22739126]
ACADIA Pharmaceuticals Inc. (2016). HIGHLIGHTS OF PRESCRIBING INFORMATION
NUPLAZID™ (pimavanserin) tablets, ACADIA Pharmaceuticals Inc., San Diego, CA 92130 USA
Retrieved from https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/207318lbl.pdf
Author Manuscript

Aloyo VJ, Berg KA, Spampinato U, Clarke WP, & Harvey JA (2009). Current status of inverse
agonism at serotonin2A (5-HT2A) and 5-HT2C receptors. Pharmacol Ther, 121(2), 160–173.
10.1016/j.pharmthera.2008.10.010 [PubMed: 19109993]
Ancoli-Israel S, Vanover KE, Weiner DM, Davis RE, & van Kammen DP (2011). Pimavanserin
tartrate, a 5-HT2A receptor inverse agonist, increases slow wave sleep as measured by
polysomnography in healthy adult volunteers. Sleep medicine, 12(2), 134–141. 10.1016/
j.sleep.2010.10.004 [PubMed: 21256805]

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 13

Asselmann E, Wittchen HU, Lieb R, Perkonigg A, & Beesdo-Baum K (2018). Incident mental
disorders in the aftermath of traumatic events: A prospective-longitudinal community study. Journal
Author Manuscript

of Affective Disorders, 227, 82–89. 10.1016/j.jad.2017.10.004 [PubMed: 29053980]


Banasr M, Soumier A, Hery M, Mocaër E, & Daszuta A (2006). Agomelatine, a New Antidepressant,
Induces Regional Changes in Hippocampal Neurogenesis. Biological Psychiatry, 59(11), 1087–
1096. 10.1016/j.biopsych.2005.11.025 [PubMed: 16499883]
Beesdo-Baum K, & Knappe S (2012). Developmental Epidemiology of Anxiety Disorders. Child and
Adolescent Psychiatric Clinics of North America, 21(3), 457–478. 10.1016/j.chc.2012.05.001
[PubMed: 22800989]
Benarroch EE (2012). GABAB receptors: structure, functions, and clinical implications. Neurology,
78(8), 578–584. 10.1212/WNL.0b013e318247cd03 [PubMed: 22351795]
Bergink V, & Westenberg HGM (2005). Metabotropic glutamate II receptor agonists in panic disorder:
a double blind clinical trial with LY354740. Int Clin Psychopharmacol, 20(6), 291–293. [PubMed:
16192835]
Bond A, Jones NM, Hicks CA, Whiffin GM, Ward MA, O’Neill MF, … Schoepp DD (2000).
Neuroprotective effects of LY379268, a selective mGlu2/3 receptor agonist: investigations into
Author Manuscript

possible mechanism of action in vivo. Journal of Pharmacology and Experimental Therapeutics,


294(3), 800–809. [PubMed: 10945827]
Bond A, Ragumoorthy N, Monn JA, Hicks CA, Ward MA, Lodge D, & O’Neill MJ (1999). LY379268,
a potent and selective Group II metabotropic glutamate receptor agonist, is neuroprotective in
gerbil global, but not focal, cerebral ischaemia. Neuroscience letters, 273(3), 191–194. 10.1016/
S0304-3940(99)00663-1 [PubMed: 10515191]
Bozymski KM, Lowe DK, Pasternak KM, Gatesman TL, & Crouse EL (2017). Pimavanserin: A Novel
Antipsychotic for Parkinson’s Disease Psychosis. Ann Pharmacother, 51(6), 479–487.
10.1177/1060028017693029 [PubMed: 28375643]
Buoli M, Grassi S, Serati M, & Altamura AC (2017). Agomelatine for the treatment of generalized
anxiety disorder. Expert opinion on pharmacotherapy, 18(13), 1373–1379.
10.1080/14656566.2017.1359257 [PubMed: 28730851]
Bushnell GA, Dusetzina SB, Compton SN, Gaynes BN, Brookhart MA, & Sturmer T (2019).
Psychotherapy Claims Surrounding Pharmacotherapy Initiation in Children and Adolescents with
Anxiety Disorders. J Child Adolesc Psychopharmacol, 29(2), 100–106. 10.1089/cap.2018.0108
Author Manuscript

[PubMed: 30565956]
Campbell BM, & Merchant KM (2003). Serotonin 2C receptors within the basolateral amygdala
induce acute fear-like responses in an open-field environment. Brain research, 993(1–2), 1–9.
10.1016/S0006-8993(03)03384-5 [PubMed: 14642825]
Chugani DC, Muzik O, Juhasz C, Janisse JJ, Ager J, & Chugani HT (2001). Postnatal maturation of
human GABAA receptors measured with positron emission tomography. Ann Neurol, 49(5), 618–
626. 10.1002/ana.1003 [PubMed: 11357952]
Chumboatong W, Thummayot S, Govitrapong P, Tocharus C, Jittiwat J, & Tocharus J (2017).
Neuroprotection of agomelatine against cerebral ischemia/reperfusion injury through an
antiapoptotic pathway in rat. Neurochemistry international, 102, 114–122. 10.1016/
j.neuint.2016.12.011 [PubMed: 28012846]
Corti C, Battaglia G, Molinaro G, Riozzi B, Pittaluga A, Corsi M, … Bruno V (2007). The use of
knock-out mice unravels distinct roles for mGlu2 and mGlu3 metabotropic glutamate receptors in
mechanisms of neurodegeneration/neuroprotection. Journal of Neuroscience, 27(31), 8297–8308.
Author Manuscript

10.1523/JNEUROSCI.1889-07.2007 [PubMed: 17670976]


Costello EJ, Egger HL, & Angold A (2005). The Developmental Epidemiology of Anxiety Disorders:
Phenomenology, Prevalence, and Comorbidity. Child and Adolescent Psychiatric Clinics, 14(4),
631–648. 10.1016/j.chc.2005.06.003
De Berardis D, Marini S, Fornaro M, Srinivasan V, Iasevoli F, Tomasetti C, … Martinotti G (2013).
The melatonergic system in mood and anxiety disorders and the role of agomelatine: implications
for clinical practice. International journal of molecular sciences, 14(6), 12458–12483. 10.3390/
ijms140612458 [PubMed: 23765220]

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 14

Doble A (1996). The pharmacology and mechanism of action of riluzole. Neurology, 47(6 Suppl 4),
S233–241. 10.1212/WNL.47.6_Suppl_4.233S [PubMed: 8959995]
Author Manuscript

Dobson ET, Bloch MH, & Strawn JR (2019). Efficacy and Tolerability of Pharmacotherapy for
Pediatric Anxiety Disorders: A Network Meta-Analysis. J Clin Psychiatry, 80(1). 10.4088/
JCP.17r12064
Du J, Suzuki K, Wei Y, Wang Y, Blumenthal R, Chen Z, … Manji HK (2007). The anticonvulsants
lamotrigine, riluzole, and valproate differentially regulate AMPA receptor membrane localization:
relationship to clinical effects in mood disorders. Neuropsychopharmacology, 32(4), 793 10.1038/
sj.npp.1301178 [PubMed: 16936714]
Duman RS (2002). Synaptic plasticity and mood disorders. Molecular psychiatry, 7(S1), S29 10.1038/
sj.mp.4001016
Dunayevich E, Erickson J, Levine L, Landbloom R, Schoepp DD, & Tollefson GD (2008). Efficacy
and tolerability of an mGlu2/3 agonist in the treatment of generalized anxiety disorder.
Neuropsychopharmacology, 33(7), 1603. doi:10.1038/sj.npp.1301531 [PubMed: 17712352]
Essau CA, Conradt J, & Petermann F (2000). Frequency, Comorbidity, and Psychosocial Impairment
of Anxiety Disorders in German Adolescents. Journal of Anxiety Disorders, 14(3), 263–279.
Author Manuscript

10.1016/S0887-6185(99)00039-0 [PubMed: 10868984]


European Medicines Agency. (2016). European Medicines Agency: Evaluation of medicines for
human use CHMP assessment report for Valdoxan. Retrieved from https://www.ema.europa.eu/
documents/variation-report/valdoxan-h-c-915-p46-0027-epar-assessment-report_en.pdf
Fatouh AM, Elshafeey AH, & Abdelbary A (2017). Intranasal agomelatine solid lipid nanoparticles to
enhance brain delivery: formulation, optimization and in vivo pharmacokinetics. Drug Des Devel
Ther, 11, 1815–1825. 10.2147/dddt.s102500
Fornaro M, Prestia D, Colicchio S, & Perugi G (2010). A systematic, updated review on the
antidepressant agomelatine focusing on its melatonergic modulation. Current Neuropharmacology,
8(3), 287–304. [PubMed: 21358978]
Freiesleben SD, & Furczyk K (2015). A systematic review of agomelatine-induced liver injury. Journal
of molecular psychiatry, 3(1), 4 10.1186/s40303-015-0011-7 [PubMed: 25932327]
Friedrich M-E, Akimova E, Huf W, Konstantinidis A, Papageorgiou K, Winkler D, … Kasper S
(2016). Drug-induced liver injury during antidepressant treatment: results of AMSP, a drug
surveillance program. International Journal of Neuropsychopharmacology, 19(4).
Author Manuscript

Frizzo ME, Dall’Onder LP, Dalcin KB, & Souza DO (2004). Riluzole enhances glutamate uptake in rat
astrocyte cultures. Cell Mol Neurobiol, 24(1), 123–128. [PubMed: 15049516]
Frokjaer VG, Mortensen EL, Nielsen FÅ, Haugbol S, Pinborg LH, Adams KH, … Paulson OB (2008).
Frontolimbic serotonin 2A receptor binding in healthy subjects is associated with personality risk
factors for affective disorder. Biological Psychiatry, 63(6), 569–576. 10.1016/
j.biopsych.2007.07.009 [PubMed: 17884017]
Gahr M, Freudenmann R, Connemann B, Hiemke C, & Schönfeldt-Lecuona C (2013). Agomelatine
and hepatotoxicity: implications of cumulated data derived from spontaneous reports of adverse
drug reactions. Pharmacopsychiatry, 46(06), 214–220. 10.1055/s-0033-1353156 [PubMed:
23966266]
Gatch MB (2003). Discriminative stimulus effects of m-chlorophenylpiperazine as a model of the role
of serotonin receptors in anxiety. Life Sciences, 73(11), 1347–1367. 10.1016/
S0024-3205(03)00422-3 [PubMed: 12850497]
Grant P, Joseph LA, Farmer CA, Luckenbaugh DA, Lougee LC, Zarate CA Jr, & Swedo SE (2014).
Author Manuscript

12-week, placebo-controlled trial of add-on riluzole in the treatment of childhood-onset obsessive–


compulsive disorder. Neuropsychopharmacology, 39(6), 1453. doi:10.1038/npp.2013.343
[PubMed: 24356715]
Grant P, Lougee L, Hirschtritt M, & Swedo SE (2007). An open-label trial of riluzole, a glutamate
antagonist, in children with treatment-resistant obsessive-compulsive disorder. Journal of Child
and Adolescent Psychopharmacology, 17(6), 761–767. 10.1089/cap.2007.0021 [PubMed:
18315448]

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 15

Greenblatt DJ, & Zammit GK (2012). Pharmacokinetic evaluation of eszopiclone: clinical and
therapeutic implications. Expert Opinion on Drug Metabolism & Toxicology, 8(12), 1609–1618.
Author Manuscript

10.1517/17425255.2012.741588 [PubMed: 23126273]


Guardiola‐Lemaitre B, De Bodinat C, Delagrange P, Millan M, Munoz C, & Mocaër E (2014).
Agomelatine: mechanism of action and pharmacological profile in relation to antidepressant
properties. Br J Pharmacol, 171(15), 3604–3619. [PubMed: 24724693]
Hackler EA, Turner GH, Gresch PJ, Sengupta S, Deutch AY, Avison MJ, … Sanders-Bush E (2007). 5-
Hydroxytryptamine2C receptor contribution to m-chlorophenylpiperazine and N-methyl-β-
carboline-3-carboxamide-induced anxiety-like behavior and limbic brain activation. Journal of
Pharmacology and Experimental Therapeutics, 320(3), 1023–1029. doi:10.1124/jpet.106.113357
[PubMed: 17138863]
Hambrecht-Wiedbusch VS, Gauthier EA, Baghdoyan HA, & Lydic R (2010). Benzodiazepine receptor
agonists cause drug-specific and state-specific alterations in EEG power and acetylcholine release
in rat pontine reticular formation. Sleep, 33(7), 909–918. 10.1093/sleep/33.7.909 [PubMed:
20614851]
Hambrecht-Wiedbusch VS, Mitchell MF, Firn KA, Baghdoyan HA, & Lydic R (2014).
Author Manuscript

Benzodiazepine site agonists differentially alter acetylcholine release in rat amygdala. Anesth
Analg, 118(6), 1293–1300. 10.1213/ane.0000000000000201 [PubMed: 24842176]
Helton DR, Tizzano JP, Monn JA, Schoepp DD, & Kallman MJ (1998). Anxiolytic and side-effect
profile of LY354740: a potent, highly selective, orally active agonist for group II metabotropic
glutamate receptors. Journal of Pharmacology and Experimental Therapeutics, 284(2), 651–660.
Retrieved from http://jpet.aspetjournals.org/content/jpet/284/2/651.full.pdf [PubMed: 9454811]
Huang MP, Radadia K, Macone BW, Auerbach SH, & Datta S (2010). Effects of eszopiclone and
zolpidem on sleep-wake behavior, anxiety-like behavior and contextual memory in rats. Behav
Brain Res, 210(1), 54–66. 10.1016/j.bbr.2010.02.018 [PubMed: 20153782]
Husky MM, Olfson M, He J, Nock MK, Swanson SA, & Merikangas KR (2012). Twelve-Month
Suicidal Symptoms and Use of Services Among Adolescents: Results From the National
Comorbidity Survey. Psychiatric Services, 63(10), 989–996. 10.1176/appi.ps.201200058
[PubMed: 22910768]
ITF Pharma Inc. (2018). TIGLUTIK, HIGHLIGHTS OF PRESCRIBING INFORMATION, ITF
Pharma, Inc 850 Cassatt Road, Suite 350 Berwyn, PA 19312 USA, 2018 ITF Pharma. Retrieved
Author Manuscript

from https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/209080s000lbl.pdf
Jacobs BL, Van Praag H, & Gage FH (2000). Adult brain neurogenesis and psychiatry: a novel theory
of depression. Molecular psychiatry, 5(3), 262 10.1038/sj.mp.4000712 [PubMed: 10889528]
Javitt D (2004). Glutamate as a therapeutic target in psychiatric disorders. Molecular psychiatry, 9(11),
984 10.1038/sj.mp.4001551 [PubMed: 15278097]
Joffe H, Petrillo L, Viguera A, Koukopoulos A, Silver-Heilman K, Farrell A, … Cohen LS (2010).
Eszopiclone improves insomnia and depressive and anxious symptoms in perimenopausal and
postmenopausal women with hot flashes: a randomized, double-blinded, placebo-controlled
crossover trial. Am J Obstet Gynecol, 202(2), 171.e171–171.e111. 10.1016/j.ajog.2009.10.868
[PubMed: 20035910]
Johnson, Fitz SD, Engleman EA, Svensson KA, Schkeryantz JM, & Shekhar A (2013). Group II
metabotropic glutamate receptor type 2 allosteric potentiators prevent sodium lactate-induced
panic-like response in panic-vulnerable rats. Journal of Psychopharmacology, 27(2), 152–161.
10.1177/0269881112454230 [PubMed: 22914798]
Author Manuscript

Johnson, Mattiuz EL, Chay SH, Herman JL, Wheeler WJ, Kassahun K, … Phillips DL (2002). The
disposition, metabolism, and pharmacokinetics of a selective metabotropic glutamate receptor
agonist in rats and dogs. Drug Metab Dispos, 30(1), 27–33. 10.1124/dmd.30.1.27 [PubMed:
11744608]
Johnson, & Shekhar A (2012). An animal model of panic vulnerability with chronic disinhibition of
the dorsomedial/perifornical hypothalamus. Physiology & behavior, 107(5), 686–698. 10.1016/
j.physbeh.2012.03.016 [PubMed: 22484112]
Katoh-Semba R, Asano T, Ueda H, Morishita R, Takeuchi IK, Inaguma Y, & Kato K (2002). Riluzole
enhances expression of brain-derived neurotrophic factor with consequent proliferation of granule

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 16

precursor cells in the rat hippocampus. The FASEB Journal, 16(10), 1328–1330. 10.1096/
fj.02-0143fje [PubMed: 12154010]
Author Manuscript

Kellner M, Muhtz C, Stark K, Yassouridis A, Arlt J, & Wiedemann K (2005). Effects of a


metabotropic glutamate 2/3 receptor agonist (LY544344/LY354740) on panic anxiety induced by
cholecystokinin tetrapeptide in healthy humans: preliminary results. Psychopharmacology (Berl),
179(1), 310–315. 10.1007/s00213-004-2025-1 [PubMed: 15821951]
Kendall PC, Compton SN, Walkup JT, Birmaher B, Albano AM, Sherrill J, … Piacentini J (2010).
Clinical characteristics of anxiety disordered youth. Journal of Anxiety Disorders, 24(3), 360–365.
10.1016/j.janxdis.2010.01.009 [PubMed: 20206470]
Kłodzińska A, Chojnacka-Wojcik E, Pałucha A, Brański P, Popik P, & Pilc A (1999). Potential anti-
anxiety, anti-addictive effects of LY 354740, a selective group II glutamate metabotropic receptors
agonist in animal models. Neuropharmacology, 38(12), 1831–1839. 10.1016/
S0028-3908(99)00066-0 [PubMed: 10608278]
Lindsley CW, Emmitte KA, Hopkins CR, Bridges TM, Gregory KJ, Niswender CM, & Conn PJ
(2016). Practical Strategies and Concepts in GPCR Allosteric Modulator Discovery: Recent
Advances with Metabotropic Glutamate Receptors. Chem Rev, 116(11), 6707–6741. 10.1021/
Author Manuscript

acs.chemrev.5b00656 [PubMed: 26882314]


Loiseau F, Le Bihan C, Hamon M, & Thiébot M-H (2006). Effects of melatonin and agomelatine in
anxiety-related procedures in rats: interaction with diazepam. European
Neuropsychopharmacology, 16(6), 417–428. [PubMed: 16376525]
Lu Y, Ho CS, McIntyre RS, Wang W, & Ho RC (2018). Agomelatine-induced modulation of brain-
derived neurotrophic factor (BDNF) in the rat hippocampus. Life Sciences, 210, 177–184.
10.1016/j.lfs.2018.09.003 [PubMed: 30193943]
Maier G, Lu Q, Blumer J, Robertson B, Zummo J, & Ludwig E (2011). A Pharmacokinetic and Safety
Evaluation of Single Oral Doses of Eszopiclone in Pediatric Subjects from 6 to 17 Years of Age
with Attention Deficit Hyperactivity Disorder and Insomnia. Paper presented at the American
Academy of Child & Adolescent Psychiatry Annual Meeting, (Toronto, Canada) https://
www.simulations-plus.com/assets/115_poster106_AACAP_eszopiclone_2011oct18.pdf
Manji HK, Quiroz JA, Sporn J, Payne JL, Denicoff K, Gray NA, … Charney DS (2003). Enhancing
neuronal plasticity and cellular resilience to develop novel, improved therapeutics for difficult-to-
treat depression. Biological Psychiatry, 53(8), 707–742. [PubMed: 12706957]
Author Manuscript

Marmigère F, Rage F, & Tapia-Arancibia L (2003). GABA–glutamate interaction in the control of


BDNF expression in hypothalamic neurons. Neurochemistry international, 42(4), 353–358.
10.1016/S0197-0186(02)00100-6 [PubMed: 12470709]
Mathew SJ, Amiel JM, Coplan JD, Fitterling HA, Sackeim HA, & Gorman JM (2005). Open-label trial
of riluzole in generalized anxiety disorder. Am J Psychiatry, 162(12), 2379–2381. 10.1176/
appi.ajp.162.12.2379 [PubMed: 16330605]
Mathew SJ, Price RB, Mao X, Smith EL, Coplan JD, Charney DS, & Shungu DC (2008).
Hippocampal N-acetylaspartate concentration and response to riluzole in generalized anxiety
disorder. Biol Psychiatry, 63(9), 891–898. 10.1016/j.biopsych.2007.09.012 [PubMed: 18028881]
Methippara M, Bashir T, Suntsova N, Szymusiak R, & McGinty D (2010). Hippocampal adult
neurogenesis is enhanced by chronic eszopiclone treatment in rats. J Sleep Res, 19(3), 384–393.
10.1111/j.1365-2869.2010.00833.x [PubMed: 20408925]
Michelson D, Levine L, Dellva M, Mesters P, Schoepp D, Dunayevich E, & Tollefson G (2005).
Clinical studies with mGluR2/3 agonists: LY354740 compared with placebo in patients with
Author Manuscript

generalized anxiety disorder. Paper presented at the Neuropharmacology.


Mizuta I, Ohta M, Ohta K, Nishimura M, Mizuta E, & Kuno S (2001). Riluzole stimulates nerve
growth factor, brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor
synthesis in cultured mouse astrocytes. Neuroscience letters, 310(2–3), 117–120. [PubMed:
11585581]
Monti JM (2010). Serotonin 5-HT(2A) receptor antagonists in the treatment of insomnia: present status
and future prospects. Drugs Today (Barc), 46(3), 183–193. 10.1358/dot.2010.46.3.1437247
[PubMed: 20467592]

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 17

Moresco FM, Dieci M, Vita A, Messa C, Gobbo C, Galli L, … Fazio F (2002). In Vivo Serotonin
5HT2A Receptor Binding and Personality Traits in Healthy Subjects: A Positron Emission
Author Manuscript

Tomography Study. NeuroImage, 17(3), 1470–1478. 10.1006/nimg.2002.1239 [PubMed:


12414286]
Morley-Fletcher S, Mairesse J, Soumier A, Banasr M, Fagioli F, Gabriel C, … Nicoletti F (2011).
Chronic agomelatine treatment corrects behavioral, cellular, and biochemical abnormalities
induced by prenatal stress in rats. Psychopharmacology (Berl), 217(3), 301. [PubMed: 21503609]
Nakamichi N, Takarada T, & Yoneda Y (2009). Neurogenesis mediated by GABA and glutamate
signaling. Journal of Pharmacological Sciences, 0905290246–0905290246. 10.1254/
jphs.08R03CR
Nicoletti F, Bockaert J, Collingridge G, Conn P, Ferraguti F, Schoepp D, … Pin J (2011). Metabotropic
glutamate receptors: from the workbench to the bedside. Neuropharmacology, 60(7–8), 1017–
1041. [PubMed: 21036182]
Ohashi M, Saitoh A, Yamada M, Oka J, & Yamada M (2015). Riluzole in the prelimbic medial
prefrontal cortex attenuates veratrine-induced anxiety-like behaviors in mice.
Psychopharmacology (Berl), 232(2), 391–398. 10.1007/s00213-014-3676-1 [PubMed: 25127925]
Author Manuscript

Perlemuter G, Cacoub P, Valla D, Guyader D, Saba B, Batailler C, & Moore K (2016).


Characterisation of agomelatine-induced increase in liver enzymes: frequency and risk factors
determined from a pooled analysis of 7605 treated patients. CNS Drugs, 30(9), 877–888. 10.1007/
s40263-016-0351-6 [PubMed: 27342740]
Pilc A (2003). LY-354740 (Eli Lilly). IDrugs, 6(1), 66–71. Retrieved from (3) (PDF) LY-354740 Eli
Lilly. Available from: https://www.researchgate.net/publication/10722185_LY-354740_Eli_Lilly
[accessed Apr 01 2019]. [PubMed: 12789623]
Pine DS (1999). Pathophysiology of childhood anxiety disorders. Biological Psychiatry, 46(11), 1555–
1566. 10.1016/S0006-3223(99)00115-8 [PubMed: 10599483]
Plaznik A, Palejko W, Nazar M, & Jessa M (1994). Effects of antagonists at the NMDA receptor
complex in two models of anxiety. European Neuropsychopharmacology, 4(4), 503–512.
10.1016/0924-977X(94)90299-2 [PubMed: 7894261]
Pollack M, Kinrys G, Krystal A, McCall WV, Roth T, Schaefer K, … Krishnan R (2008). Eszopiclone
coadministered with escitalopram in patients with insomnia and comorbid generalized anxiety
disorder. Archives of General Psychiatry, 65(5), 551–562. doi:10.1001/archpsyc.65.5.551
Author Manuscript

[PubMed: 18458207]
Prakriya M, & Mennerick S (2000). Selective Depression of Low–Release Probability Excitatory
Synapses by Sodium Channel Blockers. Neuron, 26(3), 671–682. 10.1016/
S0896-6273(00)81203-9 [PubMed: 10896162]
Rainer Q, Xia L, Guilloux J-P, Gabriel C, Mocaë r, E., … David DJ (2012). Beneficial behavioural and
neurogenic effects of agomelatine in a model of depression/anxiety. International Journal of
Neuropsychopharmacology, 15(3), 321–335. 10.1017/S1461145711000356 [PubMed: 21473810]
Regenass W, Möller M, & Harvey BH (2018). Studies into the anxiolytic actions of agomelatine in
social isolation reared rats: Role of corticosterone and sex. Journal of Psychopharmacology, 32(2),
134–145. 10.1177/0269881117735769 [PubMed: 29082818]
Revest J, Dupret D, Koehl M, Funk-Reiter C, Grosjean N, Piazza P, & Abrous D (2009). Adult
hippocampal neurogenesis is involved in anxiety-related behaviors. Molecular psychiatry, 14(10),
959 10.1038/mp.2009.15 [PubMed: 19255582]
Rorick-Kehn LM, Perkins EJ, Knitowski KM, Hart JC, Johnson BG, Schoepp DD, & McKinzie DL
Author Manuscript

(2006). Improved bioavailability of the mGlu2/3 receptor agonist LY354740 using a prodrug
strategy: in vivo pharmacology of LY544344. Journal of Pharmacology and Experimental
Therapeutics, 316(2), 905–913. doi:10.1124/jpet.105.091926 [PubMed: 16223873]
Rosenberg R, Seiden DJ, Hull SG, Erman M, Schwartz H, Anderson C, … Roth T (2008). APD125, a
selective serotonin 5-HT(2A) receptor inverse agonist, significantly improves sleep maintenance in
primary insomnia. Sleep, 31(12), 1663–1671. 10.1093/sleep/31.12.1663 [PubMed: 19090322]
Rossetti AC, Paladini MS, Racagni G, Riva MA, Cattaneo A, & Molteni R (2018). Genome-wide
analysis of LPS-induced inflammatory response in the rat ventral hippocampus: Modulatory

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 18

activity of the antidepressant agomelatine. The world journal of biological psychiatry, 19(5), 390–
401. 10.1080/15622975.2017.1298839 [PubMed: 28337940]
Author Manuscript

Said M, Elsayed I, Aboelwafa AA, & Elshafeey AH (2017). Transdermal agomelatine microemulsion
gel: pyramidal screening, statistical optimization and in vivo bioavailability. Drug Deliv, 24(1),
1159–1169. 10.1080/10717544.2017.1365392 [PubMed: 28831842]
Sangal RB, Blumer JL, Lankford DA, Grinnell TA, & Huang H (2014). Eszopiclone for insomnia
associated with attention-deficit/hyperactivity disorder. Pediatrics, 134(4), e1095–e1103.
doi:10.1542/peds.2013-4221 [PubMed: 25266438]
Sanofi-Aventis U.S. LLC. (2008). RILUTEK (riluzole) Tablets, LABEL, Sanofi-Aventis U.S. LLC
Bridgewater, NJ 08807 Retrieved from https://www.accessdata.fda.gov/drugsatfda_docs/label/
2009/020599s011s012lbl.pdf
Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S, … Arancio O (2003). Requirement of
hippocampal neurogenesis for the behavioral effects of antidepressants. science, 301(5634), 805–
809. 10.1126/science.1083328 [PubMed: 12907793]
Schoepp DD, Wright RA, Levine LR, Gaydos B, & Potter WZ (2003). LY354740, an mGlu2/3
Receptor Agonist as a Novel Approach to Treat Anxiety/Stress. Stress, 6(3), 189–197.
Author Manuscript

10.1080/1025389031000146773 [PubMed: 13129812]


Shekhar A, & Keim S (2000). LY354740, a potent group II metabotropic glutamate receptor agonist
prevents lactate-induced panic-like response in panic-prone rats. Neuropharmacology, 39(7),
1139–1146. [PubMed: 10760357]
Shortland PJ, Leinster VH, White W, & Robson LG (2006). Riluzole promotes cell survival and
neurite outgrowth in rat sensory neurones in vitro. European Journal of Neuroscience, 24(12),
3343–3353. 10.1111/j.1460-9568.2006.05218.x [PubMed: 17229083]
Simon AB, & Gorman JM (2006). Advances in the treatment of anxiety: targeting glutamate.
NeuroRx, 3(1), 57–68. 10.1016/j.nurx.2005.12.005 [PubMed: 16490413]
Spooren WP, Schoeffter P, Gasparini F, Kuhn R, & Gentsch C (2002). Pharmacological and
endocrinological characterisation of stress-induced hyperthermia in singly housed mice using
classical and candidate anxiolytics (LY314582, MPEP and NKP608). Eur J Pharmacol, 435(2–3),
161–170. 10.1016/S0014-2999(01)01562-X [PubMed: 11821022]
Stein DJ, Ahokas A, Jarema M, Avedisova AS, Vavrusova L, Chaban O, … de Bodinat C (2017).
Efficacy and safety of agomelatine (10 or 25 mg/day) in non-depressed out-patients with
Author Manuscript

generalized anxiety disorder: A 12-week, double-blind, placebo-controlled study. Eur


Neuropsychopharmacol, 27(5), 526–537. 10.1016/j.euroneuro.2017.02.007 [PubMed: 28298261]
Stein DJ, Ahokas A, Marquez MS, Hoschl C, Oh KS, Jarema M, … Olivier V (2014). Agomelatine in
generalized anxiety disorder: an active comparator and placebo-controlled study. Journal of
Clinical Psychiatry, 75(4), 362–368. doi: 10.4088/JCP.13m08433 [PubMed: 24569045]
Stein DJ, Ahokas AA, & de Bodinat C (2008). Efficacy of agomelatine in generalized anxiety disorder:
a randomized, double-blind, placebo-controlled study. J Clin Psychopharmacol, 28(5), 561–566.
10.1097/JCP.0b013e318184ff5b [PubMed: 18794654]
Stein DJ, Khoo JP, Ahokas A, Jarema M, Van Ameringen M, Vavrusova L, … de Bodinat C (2018).
12-week double-blind randomized multicenter study of efficacy and safety of agomelatine (25–
50mg/day) versus escitalopram (10–20mg/day) in out-patients with severe generalized anxiety
disorder. Eur Neuropsychopharmacol, 28(8), 970–979. 10.1016/j.euroneuro.2018.05.006
[PubMed: 30135032]
Strawn JR, Dominick KC, Patino LR, Doyle CD, Picard LS, & Phan KL (2014). Neurobiology of
Author Manuscript

Pediatric Anxiety Disorders. Curr Behav Neurosci Rep, 1(3), 154–160. 10.1007/
s40473-014-0014-1 [PubMed: 25745592]
Strawn JR, Geracioti L, Rajdev N, Clemenza K, & Levine A (2018). Pharmacotherapy for generalized
anxiety disorder in adult and pediatric patients: an evidence-based treatment review. Expert Opin
Pharmacother, 19(10), 1057–1070. 10.1080/14656566.2018.1491966 [PubMed: 30056792]
Strawn JR, Welge JA, Wehry AM, Keeshin B, & Rynn MA (2015). Efficacy and tolerability of
antidepressants in pediatric anxiety disorders: a systematic review and meta‐analysis. Depression
and Anxiety, 32(3), 149–157. [PubMed: 25449861]

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 19

Štuhec M (2013). Agomelatine-induced hepatotoxicity. Wiener klinische Wochenschrift, 125(7–8),


225–226. 10.1007/s00508-013-0344-0 [PubMed: 23519541]
Author Manuscript

Su XW, Li XY, Banasr M, & Duman RS (2009). Eszopiclone and fluoxetine enhance the survival of
newborn neurons in the adult rat hippocampus. Int J Neuropsychopharmacol, 12(10), 1421–1428.
10.1017/s1461145709990629 [PubMed: 19775501]
Sugiyama A, Saitoh A, Iwai T, Takahashi K, Yamada M, Sasaki-Hamada S, … Yamada M (2012).
Riluzole produces distinct anxiolytic-like effects in rats without the adverse effects associated
with benzodiazepines. Neuropharmacology, 62(8), 2489–2498. 10.1016/
j.neuropharm.2012.02.012 [PubMed: 22377384]
Sugiyama A, Saitoh A, Yamada M, Oka JI, & Yamada M (2017). Administration of riluzole into the
basolateral amygdala has an anxiolytic-like effect and enhances recognition memory in the rat.
Behav Brain Res, 327, 98–102. 10.1016/j.bbr.2017.03.035 [PubMed: 28359884]
Sugiyama A, Yamada M, Saitoh A, Oka JI, & Yamada M (2018). Administration of riluzole to the
basolateral amygdala facilitates fear extinction in rats. Behav Brain Res, 336, 8–14. 10.1016/
j.bbr.2017.08.031 [PubMed: 28843863]
Sunovian Pharmaceuticals Inc. (2012). Eszopiclone 21476 Clinpharm BPCA, Clinical Pharmacology
Author Manuscript

Review. Retrieved from https://www.fda.gov/files/drugs/published/Eszopiclone-21476-


Clinpharm-BPCA.pdf
Sunovion Pharmaceuticals Inc. (2014). LUNESTA™ [HIGHLIGHTS OF PRESCRIBING
INFORMATION] Sunovion Pharmaceuticals Inc., Marlborough, MA. . Retrieved from https://
www.accessdata.fda.gov/drugsatfda_docs/label/2014/021476s030lbl.pdf
Teegarden BR, Shamma HA, & Xiong Y (2008). 5-HT2A inverse-agonists for the treatment of
insomnia. Current topics in medicinal chemistry, 8(11), 969–976. Retrieved from http://
www.eurekaselect.com/82936/article [PubMed: 18673166]
Tizzano JP, Griffey KI, & Schoepp DD (2002). The anxiolytic action of mGlu2/3 receptor agonist,
LY354740, in the fear-potentiated startle model in rats is mechanistically distinct from diazepam.
Pharmacology Biochemistry and Behavior, 73(2), 367–374.
Tsuang MT, Bar JL, Stone W, & Faraone SV (2004). Gene-environment interactions in mental
disorders. World psychiatry : official journal of the World Psychiatric Association (WPA), 3(2),
73–83. [PubMed: 16633461]
Tuma J, Strubbe JH, Mocaër E, & Koolhaas JM (2005). Anxiolytic-like action of the antidepressant
Author Manuscript

agomelatine (S 20098) after a social defeat requires the integrity of the SCN. European
Neuropsychopharmacology, 15(5), 545–555. 10.1016/j.euroneuro.2005.02.004 [PubMed:
16139172]
Turck P, & Frizzo ME (2015). Riluzole stimulates BDNF release from human platelets. Biomed Res
Int, 2015, 189307 10.1155/2015/189307 [PubMed: 25629040]
Urbani A, & Belluzzi O (2000). Riluzole inhibits the persistent sodium current in mammalian CNS
neurons. European Journal of Neuroscience, 12(10), 3567–3574. [PubMed: 11029626]
Vanover, Robbins-Weilert D, Wilbraham DG, Mant TG, van Kammen DP, Davis RE, & Weiner DM
(2007). Pharmacokinetics, tolerability, and safety of ACP-103 following single or multiple oral
dose administration in healthy volunteers. J Clin Pharmacol, 47(6), 704–714.
10.1177/0091270007299431 [PubMed: 17473118]
Vanover, Weiner DM, Makhay M, Veinbergs I, Gardell LR, Lameh J, … Ott TR (2006).
Pharmacological and behavioral profile of N-(4-fluorophenylmethyl)-N-(1-methylpiperidin-4-
yl)-N′-(4-(2-methylpropyloxy) phenylmethyl) carbamide (2R, 3R)-dihydroxybutanedioate (2: 1)
Author Manuscript

(ACP-103), a novel 5-hydroxytryptamine2A receptor inverse agonist. Journal of Pharmacology


and Experimental Therapeutics, 317(2), 910–918. 10.1124/jpet.105.097006 [PubMed: 16469866]
Wahl F, & Stutzmann J (1999). Neuroprotective effects of riluzole in neurotrauma models: a review In
Current Progress in the Understanding of Secondary Brain Damage from Trauma and Ischemia
(pp. 103–110): Springer.
Yucel A, Yucel N, Ozkanlar S, Polat E, Kara A, Ozcan H, & Gulec M (2016). Effect of agomelatine on
adult hippocampus apoptosis and neurogenesis using the stress model of rats. Acta Histochemica,
118(3), 299–304. 10.1016/j.acthis.2016.02.007 [PubMed: 26970810]

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 20

Zarate CA, & Manji HK (2008). Riluzole in psychiatry: a systematic review of the literature. Expert
Opin Drug Metab Toxicol, 4(9), 1223–1234. 10.1517/17425255.4.9.1223 [PubMed: 18721116]
Author Manuscript

Zhang Y, Su J, Wang J, Tang G, Hu W, Mao J, … Yu Z (2018). Cognitive behavioral therapy for


insomnia combined with eszopiclone for the treatment of sleep disorder patients transferred out
of the intensive care unit: A single-centred retrospective observational study. Medicine
(Baltimore), 97(37), e12383 10.1097/md.0000000000012383 [PubMed: 30213004]
Zupancic M, & Guilleminault C (2006). Agomelatine: a preliminary review of a new antidepressant.
CNS Drugs, 20(12), 981–992. 10.2165/00023210-200620120-00003 [PubMed: 17140278]
Author Manuscript
Author Manuscript
Author Manuscript

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 21
Author Manuscript
Author Manuscript
Author Manuscript

Figure 1.
GABAergic and Glutamatergic Modulators. Agomelatine modulates (GABAergic receptors
and pathways broadly. Eglumegad (LY354740) is a selective agonist of presynaptic Group II
MGluRs, Eszopiclone is GABA A receptor agonist. Riluzole inhibits glutamate release and
Author Manuscript

blocks the postsynaptic effects by noncompetitive antagonism of N-methyl-D-aspartate


(NMDA) receptors. Further, riluzole increases glutamate uptake and enhances hippocampal
alpha-amion-3–3hydrosy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit
(GluR1 and GluR2) expression.

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Sonmez et al. Page 22
Author Manuscript
Author Manuscript
Author Manuscript

Figure 2.
Pharmacodynamic Targets of Novel Agents for Anxiety. Pharmacodynamic targets of novel
agents for anxiety. Agomelatine is a selective melatonin 1 and 2 receptor agonist.
Pimavanserin is a selective serotonin 5-HT2A inverse agonist. Pimavanserin also has inverse
Author Manuscript

agonist and antagonist activity for 5-HT2C and sigma-1 receptors. Riluzole inhibits voltage-
dependent sodium channels.

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Table 1.

Summary of the mechanism of action, pharmacokinetics, and pharmacodynamics of the potential drugs for GAD

Mechanism of action Bioavailability Half-life Metabolism Excretion


Sonmez et al.

Metabolized by hepatic CYP450, CYP1A2,


MT1 and MT2 receptor agonist. Rapidly absorbed after oral administration Eliminated by urinary
and CYP2C9 to 3-hydroxy-agomelatine
Agomelatine (>75%), but absolute bioavailability after 2.3 hours. excretion of its
5-HT2C receptor antagonist. (inactive), and 7-desmethyl-agomelatine
first-pass metabolism is (<5%). metabolites.
(inactive).

Oral bioavailability in humans is only 3–


5% due to limited gastrointestinal No human studies Primarily eliminated via
Eglumegad Selective mGlu2/3 agonist. No human studies for metabolism.
absorption. The oral bioavailability of the for half-life. renal excretion.
drug was ~ 10% in rats and 45% in dogs.

Unknown, likely interactions


Extensively metabolized by CYP3A4 Eliminated via excretion in
with GABA-receptor complexes
isoforms and CYP2E1 into its active urine (up to 75% as
Eszopiclone binding domains near or linked Rapidly absorbed after oral administration. ~6 hours.
metabolite, N-desmethylzopiclone, which is metabolite), and saliva
allosterically to benzodiazepine
then distributed in brain and other tissues. (<10% as parent drug).
receptors.

Pimavanserin has dose-proportional


Pimavanserin: ~57
5-HT2A antagonist/inverse pharmaco-kinetics after single oral doses Feces (<2% as parent
hours. N- Primarily metabolized via CYP3A4 and
agonist. Some affinity for 5- from 17 to 255 mg (0.5–7.5 times the drug); urine (<1% as
Pimavanserin desmethylated CYP3A5 to form the active N-
HT2C and sigma-1 receptors dosage recommendation). The parent drug; <1% as
metabolite: ~200 desmethylated metabolite.
(σ1R). bioavailability of pimavanserin oral tablets metabolites).
hours.
and oral solution is identical.

Exact mechanism is unknown. Almost 88% of the drug is metabolized by


Likely glutamate inhibitor and either oxidation (via CYP1A2) or Feces (5%); urine (90%,
Riluzole Approximately 60% by oral administration. ~12 hours.
noncompetitive NMDA receptor glucoronidation (via UGT-HP4). Some 2% as unchanged).
blocker. metabolites are pharmacologically active.

Note: NMDA N-Methyl-D-aspartic acid, 5-HT 5-hydroxytryptamine (serotonin) receptor, MT melatonergic receptor, CYP cytochrome p enzyme, UGT-HP4 UDP-glucuronosyltransferase enzyme.

Depress Anxiety. Author manuscript; available in PMC 2021 August 01.


Page 23

You might also like