ArticlePDF Available

Wake-Promoting Agents, Insights into Clinical Use and Molecular Perspectives

Authors:

Abstract

Wake-Promoting Agents (WPAs) such as amphetamine-like stimulants or modafinil, armodafinil, methyl phenidate, caffeine and nicotine reinforce the level of vigilance through an stimulated release of neurotransmitters implicated in the arousal threshold maintenance, hence shift the drive from the sleep-promoting to wake-promoting system. The modulatory effects of WPAs on cortical activation pathways give rise to enhanced vigilance. For example, cholinergic neurons of the basal forebrain and the adenosine receptors on these neurons are agonized and antagonized by nicotine andcaffeine, respectively. Caffeine similarly antagonizes adenosine receptors on the GABAergic neurons and intensifies the inhibitory drive in preoptic/anterior hypothalamus which involve in sleep induction. Modafinil however exerts its wake promoting effects through stimulating the tuberomammillary nucleus and the hypocretinergic neurons which activate the ascending reticular activating system. Although many neurotransmitter systems such as dopamine are thought to be involved upon the effects of WPAs, the empirical evidence to explain the exact mechanisms need to gain strength.
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
Review Article
Wake-Promoting Agents, Insights into
Clinical Use and Molecular Perspectives
Bijan Zare1, Behrooz Moosavi2,3, Mohammad Torabi-Nami4,5*
1Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz
University of Medical Sciences, Shiraz, Iran
2Key Laboratory of Pesticide & Chemical Biology, Ministry of Education, College of Chemistry, Central China
Normal University, Wuhan, China
3Laboratory of Organometallics, Catalysis and Ordered Materials, State Key Laboratory of Advanced Technology
for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
4Department of Neuroscience, School of Advanced Medical Sciences and Technologies,
Shiraz University of Medical Sciences, Shiraz, Iran
5Shiraz Neuroscience Research Center,
Shiraz University of Medical Sciences, Shiraz, Iran
The Burden of Excessive Daytime
Sleepiness
Sleepiness is in fact resulted from the less-
maintained arousal threshold which relieves the
inhibition exerted on the sleep-promoting system
upon wakefulness. Daytime somnolence,
characterized by inability to keep awake and vigilant
during the typical waking hours of the day, can turn
into persistent drowsiness or unintended sleep.
Moreover, sleepiness may significantly vary in
intensity and tend to occur in situations which
require minimal or no active participation (1).
The practice of general medicine encounters
frequent cases with sleep-related complaints
including excessive daytime sleepiness (EDS). EDS
is shown to leave strongly negative effects on
individuals’ quality of life, mood, interpersonal
communications, and functionality. Despite such a
significance, EDS is often under-diagnosed in
clinical practice. While EDS is known to significantly
interfere with patients’ health and functional status,
surveys have revealed that almost in 60% of
Abstract
Wake-Promoting Agents (WPAs) such as amphetamine-like stimulants or modafinil,
armodafinil, methyl phenidate, caffeine and nicotine reinforce the level of vigilance
through an stimulated release of neurotransmitters implicated in the arousal threshold
maintenance, hence shift the drive from the sleep-promoting to wake-promoting
system. The modulatory effects of WPAs on cortical activation pathways give rise to
enhanced vigilance. For example, cholinergic neurons of the basal forebrain and the
adenosine receptors on these neurons are agonized and antagonized by nicotine and
caffeine, respectively. Caffeine similarly antagonizes adenosine receptors on the
GABAergic neurons and intensifies the inhibitory drive in preoptic/anterior
hypothalamus which involve in sleep induction. Modafinil however exerts its wake-
promoting effects through stimulating the tuberomammillary nucleus and the
hypocretinergic neurons which activate the ascending reticular activating system.
Although many neurotransmitter systems such as dopamine are thought to be involved
upon the effects of WPAs, the empirical evidence to explain the exact mechanisms
need to gain strength.
*Corresponding author:
M. Torabi-Nami
Department of Neuroscience,
School of Advanced Medical
Sciences and Technologies,
Shiraz University of Medical
Sciences, Shiraz, Iran
torabinami@sums.ac.ir
Received: 10.03.2016
Revised: 26.03.2016
Accepted: 02.04.2016
Keywords:
Sleepiness; Wake-promoting
agents; Modafinil;
Stimulants Molecular
Use your device to scan and
read the article online
129
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
instances, the primary physicians do not track this
in patients (2, 3).
Clinically, when subjects obtain the score of 10
and more in a self-rated questionnaire known as
Epworth Sleepiness Scale (ESS), they will be
regarded as cases of EDS. On the other hand, the
Psychomotor Vigilance Task (PVT) or steer clear test
are utilized as objective assessments of sustained
vigilant attention in the practice of sleep medicine
in cases who present with the complaint of
excessive sleepiness. Some more comprehensive
sleep laboratory-based tests to confirm the
diagnosis of EDS include multiple-sleep latency test
(MSLT) and maintenance of wakefulness test
(MWT) (4-8).
When EDS is a chronic condition secondary to
continued sleep deprivation (intense shift work, for
instance) and especially when subjects are critical
job holders who need to deal with sensitive vehicles,
catastrophic accidents may occur. Whatever the
cause of EDS might be, drowsy driving is known to
result in many traffic road accidents and this has
lately turned to be among the foremost health
priorities in many countries (9-12).
EDS may result from numerous sleep-related
insufficiencies and long-lasting disorders including
obstructive or central sleep apnea/hypopnea
syndrome, circadian rhythm disorder and shift work,
chronic insomnia, parasomnias, restless-leg
syndrome, drug effects, narcolepsy, idiopathic
hypersomnia (with or without long sleep time) and
several related medical disorders. These conditions
not only potentially give rise to EDS but also affect
cardiovascular, neurological and psychiatric status
of the sufferers (13-15).
In general, EDS may present in various qualities.
For instance, narcolepsy and idiopathic
hypersomnia may both represent EDS, however
unlike narcolepsy which is characterized by sleep
attacks and propensity to fall sleep during the day,
idiopathic hypersomnia with long sleep time is
mainly described by sleep inertia or inability to
terminate sleep(16-18).
With regard to the prevalence of EDS, overall
standardized prevalence is shown to range from 10-
15% of general population in different studies.
However, EDS is more prevalent among older-age
strata with almost involving 35% of the subjects
over the age of 80 (19-27).
The management of EDS largely depends of
targeting and overcoming the underlying cause of
sleep inefficiency and related disorders. However, in
the event of unexplained EDS, wake-promoting
agents (WPAs) are prescribed and expected to
assist patient’s performance. Howbeit, these
agents have always had a true potential of abuse
(28).
WPAs such as amphetamine-like stimulants or
modafinil, armodafinil, methylphenidate, caffeine
and nicotine reinforce the level of vigilance through
an stimulated release of neurotransmitters
implicated in the arousal threshold maintenance,
hence shift the drive from the sleep-promoting to
wake-promoting system. The modulatory effects of
WPAs on cortical activation pathways give rise to
enhanced vigilance. For example, cholinergic
neurons of the basal forebrain and the adenosine
receptors on these neurons are agonized and
antagonized by nicotine and caffeine, respectively.
Caffeine similarly antagonizes adenosine receptors
on the GABAergic neurons and intensifies the
inhibitory drive in preoptic/anterior hypothalamus
which involve in sleep induction.
Modafinil however exerts its wake-promoting
effects through stimulating the tuberomammillary
nucleus and the hypocretinergic neurons which
activate the ascending reticular activating system.
Although many neurotransmitter systems such as
dopamine are thought to be involved upon the
effects of WPAs, the empirical evidence to explain
the exact mechanisms remain thin (28-35).
Understanding how wake-promoting drugs interact
with different components of the dopamine system
to induce arousal remains a challenge for future
research to establish new stimulant treatments
(31).
This paper provides an overview on different drugs
used as WPAs.
Wake Promoting Agents (WPAs)
Problems of ‘wakefulness’, including states of
impaired alertness, vigilance and attention affect
millions of individuals (36). Several drugs belong to
different chemical classes are used as wake-
promoting medications. Direct-acting
sympathomimetics (e.g. phenylephrine), indirect-
acting sympathomimetics (e.g., methylphenidate,
amphetamine), and non-sympathomimetic
stimulants (e.g. caffeine, modafinil and armodafinil)
are pharmacologic interventions to treat excessive
sleepiness. Hypothalamic neuropeptide (hypocretin
or orexin) are recently reported to have an important
role in the regulation of sleep and arousal states
(37, 38).
Some well-known untoward effects of psycho-
stimulants including increased feelings of anxiety
and agitation, erectile dysfunction, insomnia,
decreased libido, and in some cases, mania are
reported. Below, the main drugs used as wake
promoting agents are reviewed.
130
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
-
Modafinil
Modafinil, holds the chemical name, molecular
formula and the molecular weight 2-
[(diphenylmethyl) sulfinyl] acetamide,
C15H15NO2S and 273.35 g/mol, respectively. It is
a white to off-white, crystalline powder, practically
insoluble in water and cyclohexane while sparingly
to slightly soluble in methanol and acetone.
Modafinil is shown to improve the activity of wake-
promoting neurons through increasing dopamine
extracellular concentration partly through the
blockade of dopamine transporters (39-41). While
many studies have predetermined its possible
action on dopamine, adrenalin, noradrenalin,
serotonin and GABA systems, the precise
mechanism of action of modafinil has remained
unclear (41, 42). Although the dopaminergic and
norepinephrinergic systems appear to be crucial
targets for modafinil action, they seem not to be
exclusive and modafinil’s mechanism of action is
found to be far more complex. According to earlier
reports, modafinil directly or indirectly modulates
several neurotransmitters other than
catecholamine such as histamine and orexin (43).
As reported in a double-blind placebo-controlled
trials, 200400 mg per day modafinil significantly
reduced sleepiness, hence the medication is
recommended as a first-line therapy for sleepiness
(44, 45).
Modafinil with an elimination half-life of 13.8
hours is a long-acting compound. Its maximum
concentration is achieved in 24 hours after intake.
Generally, response to modafinil depends on the
catechol-O-methyltransferase genotype (46). The
drug is initially prescribed at 100 mg twice daily for
12 weeks and then increased to 200 mg twice
daily. Most common adverse events are mild and
comprise headache (13%), nervousness (8%) and
nausea (5%), with no evidence of tolerance and low
potential for abuse. Modafinil can raise hepatic
cytochrome P450 enzyme concentrations and
increase the metabolism of different drugs such as
oral contraceptives (47, 48).
A systematic review and meta-analysis of the
efficacy of modafinil in narcolepsy has proposed
significant benefits of modafinil for the treatment of
EDS as assessed by ESS (ESS declined by 2.73
points), multiple sleep latency test (MSLT prolonged
by 1.11 min) and MWT(MWT increased by 2.82 min)
(48, 49). Modafinil appears to lack the similar
addiction potential of other dopamine transporter
(DAT) inhibitors, such as amphetamine,
methylphenidate and cocaine. Modafinil is also
shown to reduce the excessive sleepiness observed
in patients with shift work disorder (SWD) and
results in a notably improved performance. Shift
workers with EDS underwent treatment with 200
mg of modafinil or placebo before the start of each
shift with their symptoms studied for 3 months (50).
Although modafinil reduced lapses of attention in
tests, it resulted in no loss in daytime sleepiness as
compared to placebo (51).
A review of several aspects of modafinil, focusing
on its use for ES in patients with SWD, narcolepsy
and residual sleepiness in the syndrome of
obstructive sleep apnea is published by Schwartz
(51). Generally with modafinil treatment, there was
an objective reduction in sleepiness and
improvement in general clinical conditions related
to the severity of sleepiness. The improvement in
wakefulness was associated with an improvement
in both behavioral alertness and functional status
as well as in health and quality of life. In patients
with SWD, there were decreases in the maximum
level of sleepiness during night work, in the level of
sleepiness during the travel to home and in the
incidence of accidents. Modafinil as a drug was well-
tolerated with no impairment in sleep or
cardiovascular parameters. Long-term studies
suggest that the efficacy is maintained with little
likelihood of tolerance and there were no adverse
effects on scheduled sleep, demonstrating the
beneficial effect of modafinil on daily-life and well-
being (51).
-
Armodafinil
The R-enantiomer of modafinil known as
armodafinil, has recently been approved to treat
excessive sleepiness associated with OSA, SWD,
and narcolepsy. Armodafinil has a longer half-life
and is approved for the treatment of ES associated
with SWD in some countries. Armodafinil has been
shown to improve alertness and performance (52).
Armodafinil is used similarly in the treatment of
EDS associated with the narcoleptic syndrome,
obstructive sleep apnea, and shift-work sleep
disorder. Plasma concentration following
armodafinil administration lasts longer than that
following modafinil administration, resulting in a
more prolonged effect during the day and potential
improvement in sleepiness in the late afternoon in
patients with narcolepsy. A single dose of 150 or
250 mg armodafinil is given orally in the morning for
treatment of the narcoleptic syndrome or
obstructive sleep apnea and a single dose (150 mg)
is prescribed 1 hour before starting work for the
management of shift-work sleep disorder. Reduced
doses are recommended in the elderly and in
patients with severe hepatic impairment (52, 53).
131
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
-
Amphetamines
Stimulants include naturally occurring substances
such as cocaine and caffeine or synthetic drugs for
example amphetamine and dexamphetamine,
methamphetamine, pemoline, bupropion,
ephedrine/pseudoephedrine and methylphenidate
broadly acting as both dopaminergic and
noradrenergic reuptake inhibitors at varying
degrees (54).
Amphetamines including L-amphetamine, D-
amphetamine and methamphetamine were coined
from amphetamine that was initially synthesized as
1-methyl-2-phenethylamine and initially used to
treat narcolepsy (55, 56).
Amphetamine was initially synthesized in Berlin in
1887 as 1-methyl-2-phenethylamine (55).
Amphetamines have been used for narcolepsy
since the 1930s (53). At low doses, the main effect
of amphetamine is to release dopamine and to a
lesser degree, norepinephrine through reverse
efflux via monoamine, dopamine and
norepinephrine transporters. At higher doses,
monoaminergic depletion and inhibition of reuptake
occurs. Similar to modafinil, amphetamines are
racemic compounds with their D-isomers more
potent on dopamine transmission than L-isomers
and subsequently have a greater stimulating effect
(57).
The methylated form of amphetamine named
methamphetamine exerts a potent wake-promoting
effect because it more efficiently crosses the blood
brain barrier.
Beside the effectiveness of amphetamines in
reducing sleepiness, they showed adverse effects
including disturbances of mood and behavior in
addition to cardiac and gastrointestinal effects. The
most common drug related effects are loss of
appetite, insomnia, emotional labiality,
nervousness, fever, irritability and impulsivity and at
worst, psychotic reactions. On the other hand,
insomnia, hypertension and abnormal movements
can also occur at large doses (60 mg/day and
beyond) (53, 55).
Amphetamine is effective at the dose of 10- 60
mg/day. The therapeutic effects begin within
45−60 min after ingestion of an immediate-release
tablet, with peak effect in 2 to 3 hours, and a total
duration of 4−6 h. Effects peak about 4−7 h after
ingestion of extended-release doses, and last about
12 h, depending on the dose (57).
-
Methylphenidate
Methylphenidate, another N-methyl derivative of
amphetamine, with a shorter half-life, milder side
effects, and low abuse potential has been used
since 1970 instead of amphetamine (58). It is a
potent stimulant that primarily acts by blocking of
the reuptake of monoamines (mainly dopamine)
and, unlike amphetamines, does not inhibit the
vesicular monoamine transporter. It improves
daytime sleepiness in patients with narcolepsy at
daily doses of 1060 mg, going up to 100 mg daily
at most for severe cases (59).
The effect of methylphenidate is thought be
similar to effect of amphetamines. However, there
is an argue for a slight superiority of amphetamines.
Adverse effects of methylphenidate are similar to
amphetamines adverse effect, but to a lesser
degree. Methylphenidate probably has a better
therapeutic index than D-amphetamine and showed
less reduction of appetite or increase in blood
pressure. Methylphenidate with relatively short
duration of action (34 h) supposed to be useful in
cases that needs to maximum alertness or at a
specific time of day (59, 60).
-
Gamma hydroxybutyrate
Gamma-hydroxybutyrate (GHB) (sodium
oxybate®) is a short-chain fatty acid derivative of
gamma-aminobutyric acid (GABA). This compound
readily crosses the blood-brain barrier to enter the
central nervous system (57, 61). It is a GABAB
receptor agonist at a pharmacological doseand
leaves a positive effect on EDS. Gamma-
hydroxybutyrate, is approved by the EMA for the
treatment of narcolepsy and by the US FDA for the
treatment of cataplexy and EDS in patients with
narcolepsy (58, 59). The frequency of inadvertent
daytime naps and night-time awakenings reduced
with GABA (58). However, common side effects of
gamma aminobutyrate are dizziness, headache,
nausea, pain, somnolence, sleep disorder,
confusion, infection, vomiting, and enuresis
(58).The major problem with GHB is its non-medical
use. GHB is misused in athletes for its metabolic
effects (growth hormone-releasing effects).
However, post marketing follow-up studies indicate
that abuse potential in patients with narcolepsy
receiving sodium oxybate is low (59, 61). Overall,
gamma hydroxybutyrate is effective, and
recommended for treatment of cataplexy, EDS, and
disrupted sleep due to narcolepsy (58).
-
Orexin (Hypocretin)
The neuropeptides orexin produced in
hypothalamic neurons also known as hypocretin. In
this system two neuropeptide was recognized orexin
A and orexin B. Recently, the critical role of orexin
system in regulation of arousal and maintenance of
wakefulness as well as other behavioral traits such
132
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
as feeding and reward processes have been proven
in several studies.
On the other hand, orexin deficiency is seen in
narcolepsy in humans, suggesting that the orexin
system is particularly important for maintenance of
wakefulness.
Orexin stimulates waking active monoaminergic
and cholinergic neurons in the hypothalamus and
brainstem regions to maintain a long, consolidated
waking period. Also orexin system effects complex
interactions between monoaminergic/cholinergic
wakepromoting and GABAergic sleeppromoting
neuronal systems.
Research for the orexin agonist and antagonist for
the treatment of sleep disorders has forcefully
increased over the past decades. As a result, this
system may be a potentially important therapeutic
target for the treatment of sleep disorders, and
orexin replacement therapy would probably be an
alternative promising strategy for both sleepiness
and cataplexy (62-64).
Since a drug should be delivered to the site of
action to show its effect, orexin with peptide
structure encounter blood brain barriers as an
obstacle. It is the main reason for defeat in
peripherally and systemically administration
approach. Intra-cerebro-ventricular route for orexin
administration is possibly the most effective, but
unsuitable for the treatment of patients. Other
noninvasive method such as intranasal delivery that
targets drugs to the brain along olfactory and
trigeminal neural pathways, bypassing the BBB and
minimizing systemic exposure and side effects (62).
Orexin-producing cell transplantation might
theoretically provide a cure for patients with
narcolepsy. Recent improvements in stem cell
technology open a new insight to overcome the
problems of cell therapy approach such as graft
rejection, low survival rate of implant and lack of
supply for graft availability (57).
-
Thyrotropin-releasing hormone
Thyrotropin-releasing hormone (TRH) and its
agonists have alerting properties and could help in
improving the waking system. TRH at high doses
and TRH agonists increase alertness and have been
showed to be wake-promoting and anti-cataplectic
in the canine narcoleptic model.
However, older clinical trials, mainly on
depression, reported little efficacy on mood and on
alerting effects (57,59).
-
Caffeine/paraxanthine
Caffeine is a widely an earliest used stimulant
and often used in the treatment against daytime
sleepiness and narcolepsy (59). Caffeine as a
natural alkaloid, rapidly absorbed through the
gastrointestinal tract. It is primarily metabolized in
the liver, by demethylation, to 1,7-dimethylxanthine
via cytochrome P-450 1A2. The variable effect of
caffeine within a population may be explained by
differing cytochrome P450 activities between
individuals (56). Because of low potency of wake-
promoting effect of caffeine high dose is need to
reach the best effect, where cardiovascular side
effect is the bottleneck (58, 59). However, caffeine
is known as an adenosine A1 and A2a receptor
antagonist. Zhi-Li et al used knockout mouse to
show that caffeine-related increase in wakefulness
depends on adenosine A2a receptors (65).
Paraxanthine, is a metabolite of caffeine with
greater stimulant properties and longer-lasting
wake promoting potency, lower toxicity and lesser
anxiogenic effects (56, 59).
Pitolisant
Pitolisant is a histamine H3 inverse agonist which
has passed phase-III clinical trials for the treatment
of EDS and narcolepsy. Histamine H3 receptors
reduce the synthesis and release of histamine (66).
In a double-blind randomized trial, Dauvilliers et al.
showed that pitolisant at doses up to 40 mg was
efficacious on EDS compared with placebo and well-
tolerated compared with modafinil. They suggested
that pitolisant could offer a new treatment option for
patients with narcolepsy and EDS (67).
-
Other wake-promoting drugs
Mazindol, an imidazole derivative, has similar
pharmacological effects to the amphetamines.
However, it is a weak releasing agent for dopamine,
it also blocks dopamine and adrenaline reuptake
with high affinity and holds some efficacy on
sleepiness. Adverse events are frequent, including
weight-loss, dry mouth, nervousness, constipation
and, less frequently, nausea, vomiting, headache,
dizziness and tachycardia. A careful cardiology
follow-up is recommended. Mazindol has less
potential for abuse and tolerance than
amphetamines (57).
The challenges of studying
wakefulness at molecular level
Studying arousal has been facing several
challenges including involvement of various neural
circuits and neurotransmitter systems. Sometimes
arousal-inducing drugs may not be specific for a
particular target resulting in other effects.
Additionally, target proteins might be distributed
133
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
throughout the brain raising the question “whether
all the targets required for the drug effect to be
observed?” Understanding the wakefulness and
sleep phenomena at molecular level in eukaryotes
such as zebra fish, fruit fly, and mouse can solve at
least some of the issues raised. Conservation of
sleep genetics and pharmacology, simplicity of the
organism and the control which can be posed over
the interfering factors has provided the opportunity
to decipher molecular events occurring during
wakefulness and sleep. This particularly has
advanced as a result of development of molecular
biology tools such as conditional knockout
technology, virus-mediated gene transfer and RNAi
technology using the model organisms (66).
Pharmacogenetics of sleep-wake
therapeutics
Over years, sleep medicine has been witnessing a
great deal of advancement in therapeutics
discovered against EDS disorder. Modafinil as a
novel non-amphetamine wake-promoting drug
replaced benzodiazepines which themselves have
been initiated subsequent to the emergence of
barbiturates (molecular genetic advances). Here,
we discuss the molecular mechanisms of action of
the wake promoting therapeutics used against EDS.
The focus will be on modafinil, metamphetamine
and orexin (68, 69).
-
Modafinil
The precise mechanism of action of modafinil
remains to be established. However several lines of
evidence provide clues on how it may act. Modafinil
has been reported to increase HA, NE, 5-HE, and DA
levels in the brain, though some of these effects
might be indirect. At least parts of the modafinil
effect might be exerted through DAT and NET.
Additionally, although orexin similar to modafinil
has the wake-promoting effect, modafinil does not
act through orexin and its receptors. Modafinil
increases fos expression and HAergic tone in TMN,
but this effect was not observed when the drug was
administered directly into TMN. Modafinil also
affects the glutamate level and this appears to be
dependent on the brain region. However its effect
on the GABA level is more consistent with no effect
in the thalamus and hippocampus and a reduction
in the cortex, medial preoptic area of the
hypothalamus, posterior hypothalamus, nucleus
accumbens, pallidum, and striatum. This effect
presumably is mediated by serotonin (70).
Modafinil has neuroprotective (antioxidant)
effects as well, but this might not be totally
irrelevant to its wake-promoting effect. The target of
antioxidant activity of modafinil is still not clear, but
it may be the enzymes responsible for free-radical
scavenging system. Since mitochondrion is the
main source of the generation of free radicals, it is
also possible that modafinil acts on the enzymes in
this organ such as cytochrome c which affects the
levels of free radicals and ATP. Similarly, modafinil
may reduce the activity of the inhibitory K-ATP
channels the suppress neurotransmitter release
with the end result of increased neurotransmitter
release. Furthermore modafinil may suppress other
enzymes such as cytochrome p450 which has been
shown to be a source of reactive oxygen species in
coronary artery ischemia and reperfusion injury.
CYP2C a member of C p450 family may also be
involved in the metabolism of arachidonic acid in
the brain and altering the effect of
neurotransmitters. Therefore the inhibitory effect of
modafinil might be exerted through this pathway.
Modafinil may suppress CYP2C either directly or
through the release of serotonin and epinephrine
(40, 42, 71-75).
At least parts of the effects of free radicals might
be exerted through an increase in extracellular level
of adenosine; a sleep promoting factor throughout
the brain. Although the antioxidant effect of
modafinil might be scattered throughout the brain,
since adenosine exert its sleep-inducing effect in
the basal forebrain, this region might be mainly
influenced by modafinil (40, 42, 71).
An alternative target of modafinil might be a
receptor or an intracellular protein. One such
candidate is sodium or calcium channels as
changes in sodium homeostasis and calcium influx
can affect neurotransmitter release. However this
does not explain its neuroprotective effects.
Accumulating evidence suggest that
neurodegeneration and sleep may have a common
or related mechanisms, thus there may be a single
site of action for these phenomena. Sine
mitochondria, oxidative stress and calcium
homeostasis have also been implicated in the
pathogenesis of a number of neurodegenerative
diseases, it seems plausible that mitochondria is
the main culprit both for neurodegeneration and
sleep-inducing factors (76-83).
-
Metamphetamines (MAs)
The vesicular monoamine transporter-2 (VMAT-2)
and the plasmalemmal dopamine transporter (DAT)
are the two main substrates of amphetamine on
dopamine neuronal terminals. MAs are substrates
of the Na+/Cl- dependent dopamine transporters.
"Exchange diffusion model" predicts that
metamphetamines compete with extracellular
134
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
synaptic dopamine on the DAT. Binding of
extracellular MA to the DAT leads to the cytosolic
dopamine to be reverse transported to the outside
the cell. At higher concentrations, amphetamines
can diffuse directly through the plasmalemmal
membrane. According to this model, the DAT activity
is regulated by cell signaling mechanisms such as
calmodulin-dependent protein kinase-II and
phosphotydyl inositol 3-kinase. Through another
mechanism, DAT can assume a channel-like
conformation, enabling brief release of dopamine
as well. Furthermore MAs in-vitro can promote
internalization of DAT through endocytosis which
ablates the DAT capacity to decrease synaptic levels
of dopamine(84). The VMAT-2, a membrane protein,
transports monoamines from the cytosol into
synaptic vesicles. This activity is coupled to a
vacuolar type H+-pumping ATPase. Amphetamines
at concentrations higher than 100 µM can disrupt
the proton gradient between inside and outside of
the vesicle leading to the monoamine leak into the
cytosol. Additionally, multiple high doses of AMs
(40mg/kg) lead to the redistribution of VMAT-2 to an
unknown cellular location in a rat model. On the
other hand, AMs at physiological concentrations
bind to VMAT-2 and inhibit monoamine uptake in
vesicles with the end result of increased cytosolic
and synaptic monoamine concentration. MAs also
influence DAT uptake through other proteins such
as trace amine-associated receptor-1 (TAAR1). This
is a G-protein coupled receptor for trace amines.
Activated TAAR1 decreases DAT dopamine uptake,
increases dopamine efflux, and promotes DAT
endocytosis. MA is an agonist of TAAR1. Another
mechanism of action of MAs is the inhibition of
monoamine; an enzyme on the outer membrane of
mitochondria with a role in amine catabolism in
presynaptic terminals (85, 86).
-
Orexin
Orexin has two G-protein coupled receptors, OX1R
and OX2R. Distinct G protein may contribute to
different physiological roles of orexin in certain
neurons. Both voltage-dependent calcium channels
and G-protein-gated inwardly-rectifier potassium
channels (GIRKs) can be modulated by G-protein-
coupled neurotransmitter receptors; however the
receptors might be selectively coupled to one or
more of the channels in neurons. Studies suggest
that OX1R couples exclusively to PTX-insensitive G
proteins, and OX2R couples to both PTX-sensitive
and insensitive proteins. Several lines of evidence
suggest that OX1R and OX2R signaling not only
activate neurons but also may have other roles in
the tips of developing neuritis and on presynaptic
terminals. These functions eventually lead to growth
cone collapse and increased release of
neurotransmitters. Orexin in addition can cell-
dependently increase or decrease cAMP which
might be the result of coupling the receptor with
different G-proteins. The latter might be influenced
by the receptor density. Furthermore, orexin
receptors can interact with other signaling receptors
such as cannabinoid receptors which is reported to
cause an enhancement of the orexin-A capability to
activate the mitogen-activated protein kinase
pathway. Therefore, such observations indicate that
orexin signaling events might be more complex than
initially thought (87-89).
Future directions in molecular
research
Various eukaryotic model systems can be used to
investigate the molecular mechanism of wake-
promoting drugs. For example modafinil can
promote wakefulness in fruit fly. The simplicity of
this model compared with higher eukaryotes allows
identification of the principle genes, proteins and
pathways involved in wakefulness regulation
(molecular genetics advances). Approaches such as
Large-scale random mutagenesis can be applied
to identify some of the genes involved.
Yeast is the simplest eukaryote which can exist
either haploid or diploid. This facilitates the study of
the role of genes in cellular processes. Yeast can be
grown with or without functional mitochondria and
therefore may provide an ideal model to study the
effect of modafil on mitochondria function and
oxidative stress. Yeast can also be beneficial to
study the mechanism of inhibition of monoamine
oxidase by MAs as this enzyme can be expressed in
yeast and its function can be studied in the
presence of MAs.
Moreover, several proteins involved in
waking/sleep states can be expressed in yeast.
Some of which may then create a particular
phenotype in yeast such as growth defect that can
be overcome by complementation studies with a
certain mammalian protein. This may provide the
proteins associated with each other in the
wake/sleep states and thereby provide evidence to
understand the signaling pathways. The results
obtained with the yeast model system can then be
established in higher eukaryotes such as fruit-fly
and zebrafish. Identifying the neurons responsible
for drug's effect in fruit fly can pave the way to
investigate the effect of other internal and external
regulatory systems such as food, light, motivational
states, homeostatic processes, circadian rhythms,
and memory formation.
135
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
Table 1. An overview of the key features of the mostly-studied wake-promoting agents.
Zebrafish is particularly-useful model for studying
the effect of small molecule on wake/sleep states.
This approach in combination with the gene
expression profiling can identify the drug-activated
neurons. Subsequently, gene ablation methods can
determine whether the gene is essential for drug-
induced effect.
Additionally, viral-mediated focal replacement of
certain genes in this model can provide valuable
information for deciphering the wake/sleep
circuitry. Since there is significant homology
between zebrafish and mammalian brain structure,
observations made in this model can have
correspondence in mammals.
Conditional knockout technology in which viruses
injected in the brain of adult mice provides both
temporal and spatial control of wake/sleep gene
function. Moreover siRNA-mediated mRNA
knockdown has facilitated investigating the function
of certain proteins and receptors.
136
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
Another approach is comparing the
transcriptome/proteom profile of the wakefulness
with the sleep state. This has already resulted in the
identification of several genes associated with
either wakefulness or sleep state. Genes such as
those responsible for expressing cytokines, TNFR1,
TNFR2, TNF-α, and lymophotaxin which regulate the
wake/sleep state can also be studied using
mutagenesis as well as polymorphism and
association with known wake/sleep genes.
Optogenetic inhibition or activation of the desired
genes permits identification of drug targets without
altering the function of a certain gene for good.
However studying wake/sleep states through "locus
by locus" approach might be hindered by the
redundancy of the circuitry involved (87-89).
Conclusion
The present review was an attempt to provide an
overview on wake-promoting therapeutics within
the basic-clinical spectrum. The use of wake-
promoting agents in the event of EDS need to be
carefully assessed since it may masquerade the
underlying sleep-related disorder. Novel
therapeutics in this vein are being developed
through targeting new molecular targets. Given the
health-related and societal consequences of EDS,
basic, translational and clinical research to unravel
its various dimensions should gain impetus.
References
1. Shimizu T. [Excessive daytime sleepiness (EDS)]. Nihon rinsho
Japanese journal of clinical medicine. 2015;73(6):937-41.
2. Kara B, Kilic O. Predictors of poor sleep quality and excessive
daytime sleepiness in Turkish adults with type 2 diabetes. Journal of
clinical nursing. 2015;24(9-10):1436-9.
3. Begley KJ, Castillo S, Steinshouer CR, Malesker MA. Role of
pharmacists in the treatment of excessive daytime sleepiness. The
Consultant pharmacist : the journal of the American Society of Consultant
Pharmacists. 2014;29(11):741-52.
4. Fong SY, Ho CK, Wing YK. Comparing MSLT and ESS in the
measurement of excessive daytime sleepiness in obstructive sleep
apnoea syndrome. Journal of psychosomatic research. 2005;58(1):55-60.
5. Schreier DR, Roth C, Mathis J. Subjective perception of
sleepiness in a driving simulator is different from that in the Maintenance
of Wakefulness Test. Sleep medicine. 2015;16(8):994-8.
6. Tanaka H. [Multiple sleep latency test, maintenance of
wakefulness test and suggestive immobilization test]. Nihon rinsho
Japanese journal of clinical medicine. 2015;73(6):971-9.
7. van der Heide A, van Schie MK, Lammers GJ, Dauvilliers Y,
Arnulf I, Mayer G, et al. Comparing Treatment Effect Measurements in
Narcolepsy: The Sustained Attention to Response Task, Epworth
Sleepiness Scale and Maintenance of Wakefulness Test. Sleep.
2015;38(7):1051-8.
8. Bioulac S, Chaufton C, Taillard J, Claret A, Sagaspe P,
Fabrigoule C, et al. Excessive daytime sleepiness in adult patients with
ADHD as measured by the Maintenance of Wakefulness Test, an
electrophysiologic measure. The Journal of clinical psychiatry.
2015;76(7):943-8.
9. Chen Z, Wu C, Zhong M, Lyu N, Huang Z. Identification of
common features of vehicle motion under drowsy/distracted driving: A
case study in Wuhan, China. Accident; analysis and prevention.
2015;81:251-9.
10. Sadeghniiat-Haghighi K, Moller HJ, Saraei M, Aminian O,
Khajeh-Mehrizi A. The Epworth Sleepiness Scale for screening of the
drowsy driving: comparison with the maintenance of wakefulness test in
an Iranian sample of commercial drivers. Acta medica Iranica.
2014;52(2):125-9.
11. Anderson C, Horne JA. Driving drowsy also worsens driver
distraction. Sleep medicine. 2013;14(5):466-8.
12. Dawson J. Putting the brakes on drowsy driving. The American
nurse. 2012;44(2):9.
13. Rosenberg RP. Clinical assessment of excessive daytime
sleepiness in the diagnosis of sleep disorders. The Journal of clinical
psychiatry. 2015;76(12):e1602.
14. Rose D, Gelaye B, Sanchez S, Castaneda B, Sanchez E, Yanez
ND, et al. Morningness/eveningness chronotype, poor sleep quality, and
daytime sleepiness in relation to common mental disorders among
Peruvian college students. Psychology, health & medicine.
2015;20(3):345-52.
15. Haregu A, Gelaye B, Pensuksan WC, Lohsoonthorn V,
Lertmaharit S, Rattananupong T, et al. Circadian rhythm characteristics,
poor sleep quality, daytime sleepiness and common psychiatric disorders
among Thai college students. Asia-Pacific psychiatry : official journal of the
Pacific Rim College of Psychiatrists. 2015;7(2):182-9.
16. Suzuki K, Miyamoto M, Miyamoto T, Inoue Y, Matsui K,
Nishida S, et al. The Prevalence and Characteristics of Primary Headache
and Dream-Enacting Behaviour in Japanese Patients with Narcolepsy or
Idiopathic Hypersomnia: A Multi-Centre Cross-Sectional Study. PloS one.
2015;10(9):e0139229.
17. Pereira D, Lopes E, da Silva Behrens NS, de Almeida Fonseca
H, Sguillar DA, de Araujo Lima TF, et al. Prevalence of periodical leg
movements in patients with narcolepsy in an outpatient facility in Sao
Paulo. Sleep Science. 2014;7(1):69-71.
18. Heier MS, Evsiukova T, Wilson J, Abdelnoor M, Hublin C, Ervik
S. Prevalence of narcolepsy with cataplexy in Norway. Acta neurologica
Scandinavica. 2009;120(4):276-80.
19. Bjorvatn B, Lehmann S, Gulati S, Aurlien H, Pallesen S, Saxvig
IW. Prevalence of excessive sleepiness is higher whereas insomnia is
lower with greater severity of obstructive sleep apnea. Sleep & breathing
= Schlaf & Atmung. 2015;19(4):1387-93.
20. Signal TL, Paine SJ, Sweeney B, Priston M, Muller D, Smith A,
et al. Prevalence of abnormal sleep duration and excessive daytime
sleepiness in pregnancy and the role of socio-demographic factors:
comparing pregnant women with women in the general population. Sleep
medicine. 2014;15(12):1477-83.
21. Liviya Ng W, Freak-Poli R, Peeters A. The prevalence and
characteristics associated with excessive daytime sleepiness among
Australian workers. Journal of occupational and environmental medicine /
American College of Occupational and Environmental Medicine.
2014;56(9):935-45.
22. Bjorvatn B, Pallesen S, Gronli J, Sivertsen B, Lehmann S.
Prevalence and correlates of insomnia and excessive sleepiness in adults
with obstructive sleep apnea symptoms. Perceptual and motor skills.
2014;118(2):571-86.
23. Hayley AC, Williams LJ, Kennedy GA, Berk M, Brennan SL,
Pasco JA. Prevalence of excessive daytime sleepiness in a sample of the
Australian adult population. Sleep medicine. 2014;15(3):348-54.
24. Verbruggen AE, Dieltjens M, Wouters K, De Volder I, Van de
Heyning PH, Braem MJ, et al. Prevalence of residual excessive sleepiness
during effective oral appliance therapy for sleep-disordered breathing.
Sleep medicine. 2014;15(2):269-72.
25. Vashum KP, McEvoy MA, Hancock SJ, Islam MR, Peel R, Attia
JR, et al. Prevalence of and associations with excessive daytime
sleepiness in an Australian older population. Asia-Pacific journal of public
health / Asia-Pacific Academic Consortium for Public Health.
2015;27(2):NP2275-84.
26. Risco J, Ruiz P, Marinos A, Juarez A, Ramos M, Salmavides F,
et al. Excessive sleepiness prevalence in public transportation drivers of a
developing country. Traffic injury prevention. 2013;14(2):145-9.
27. Gunes Z, Sahbaz M, Tugrul E, Gunes H. Prevalence and risk
factors for excessive daytime of sleepiness in rural western Anotolia
(Turkey): the role of obesity and metabolic syndrome. The Southeast Asian
journal of tropical medicine and public health. 2012;43(3):747-55.
28. Dunn D, Hostetler G, Iqbal M, Messina-McLaughlin P, Reiboldt
A, Lin YG, et al. Wake-promoting agents: search for next generation
modafinil: part I. Bioorganic & medicinal chemistry letters.
2012;22(6):2312-4.
29. Lesur B, Lin YG, Marcy VR, Aimone LD, Gruner J, Bacon ER, et
al. Aryl-heteroaryl derivatives as novel wake-promoting agents. Chemical
biology & drug design. 2013;81(3):429-32.
30. Louvet P, Schweizer D, Gourdel ME, Riguet E, Yue C, Marcy
VR, et al. Wake-promoting agents: search for next generation modafinil:
part IV. European journal of medicinal chemistry. 2012;54:949-51.
137
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
31. Dunn D, Hostetler G, Iqbal M, Marcy VR, Lin YG, Jones B, et al.
Wake promoting agents: search for next generation modafinil, lessons
learned: part III. Bioorganic & medicinal chemistry letters.
2012;22(11):3751-3.
32. Dunn D, Hostetler G, Iqbal M, Messina-McLaughlin P, Reiboldt
A, Lin YG, et al. Wake-promoting agents: search for next generation
modafinil: part II. Bioorganic & medicinal chemistry letters.
2012;22(6):2315-7.
33. Lynch J, Jr., Regan C, Stump G, Tannenbaum P, Stevens J,
Bone A, et al. Hemodynamic and cardiac neurotransmitter-releasing
effects in conscious dogs of attention- and wake-promoting agents: a
comparison of d-amphetamine, atomoxetine, modafinil, and a novel
quinazolinone H3 inverse agonist. Journal of cardiovascular
pharmacology. 2009;53(1):52-9.
34. Stocking EM, Letavic MA. Histamine H3 antagonists as wake-
promoting and pro-cognitive agents. Current topics in medicinal chemistry.
2008;8(11):988-1002.
35. Makris AP, Rush CR, Frederich RC, Kelly TH. Wake-promoting
agents with different mechanisms of action: comparison of effects of
modafinil and amphetamine on food intake and cardiovascular activity.
Appetite. 2004;42(2):185-95.
36. Oken BS, Salinsky MC, Elsas SM. Vigilance, alertness, or
sustained attention: physiological basis and measurement. Clinical
neurophysiology : official journal of the International Federation of Clinical
Neurophysiology. 2006;117(9):1885-901.
37. Ebrahim IO, Howard RS, Kopelman MD, Sharief MK, Williams
AJ. The hypocretin/orexin system. Journal of the Royal Society of Medicine.
2002;95(5):227-30.
38. McWhirter D, Bae C, Budur K. The Assessment, Diagnosis,
and Treatment of Excessive Sleepiness: Practical Considerations for the
Psychiatrist. Psychiatry (Edgmont). 2007;4(9):26-35.
39. Wisor JP, Nishino S, Sora I, Uhl GH, Mignot E, Edgar DM.
Dopaminergic role in stimulant-induced wakefulness. The Journal of
neuroscience : the official journal of the Society for Neuroscience.
2001;21(5):1787-94.
40. Ferraro L, Tanganelli S, O'Connor WT, Antonelli T, Rambert F,
Fuxe K. The vigilance promoting drug modafinil increases dopamine
release in the rat nucleus accumbens via the involvement of a local
GABAergic mechanism. European journal of pharmacology. 1996;306(1-
3):33-9.
41. Gerrard P, Malcolm R. Mechanisms of modafinil: A review of
current research. Neuropsychiatric disease and treatment.
2007;3(3):349-64.
42. Saper CB, Scammell TE. Modafinil: a drug in search of a
mechanism. Sleep. 2004;27(1):11-2.
43. Ishizuka T, Murotani T, Yamatodani A. Modanifil activates the
histaminergic system through the orexinergic neurons. Neuroscience
letters. 2010;483(3):193-6.
44. Broughton RJ, Fleming JA, George CF, Hill JD, Kryger MH,
Moldofsky H, et al. Randomized, double-blind, placebo-controlled
crossover trial of modafinil in the treatment of excessive daytime
sleepiness in narcolepsy. Neurology. 1997;49(2):444-51.
45. Kumar R. Approved and investigational uses of modafinil : an
evidence-based review. Drugs. 2008;68(13):1803-39.
46. Dauvilliers Y, Neidhart E, Billiard M, Tafti M. Sexual
dimorphism of the catechol-O-methyltransferase gene in narcolepsy is
associated with response to modafinil. The pharmacogenomics journal.
2002;2(1):65-8.
47. Saper CB, Scammell TE. Emerging therapeutics in sleep.
Annals of neurology. 2013;74(3):435-40.
48. Zaharna M, Dimitriu A, Guilleminault C. Expert opinion on
pharmacotherapy of narcolepsy. Expert opinion on pharmacotherapy.
2010;11(10):1633-45.
49. Golicki D, Bala MM, Niewada M, Wierzbicka A. Modafinil for
narcolepsy: systematic review and meta-analysis. Medical science monitor
: international medical journal of experimental and clinical research.
2010;16(8):RA177-86.
50. Schwartz JR, Roth T. Shift work sleep disorder: burden of
illness and approaches to management. Drugs. 2006;66(18):2357 -70.
51. Schwartz JRL. Modafinil in the treatment of excessive
sleepiness. Drug design, development and therapy. 2008;2:71-85.
52. Nishino S, Okuro M. Armodafinil for excessive daytime
sleepiness. Drugs of today. 2008;44(6):395-414.
53. Randomized trial of modafinil for the treatment of
pathological somnolence in narcolepsy. US Modafinil in Narcolepsy
Multicenter Study Group. Annals of neurology. 1998;43(1):88-97.
54. Edgar CJ, Pace-Schott EF, Wesnes KA. Approaches to
measuring the effects of wake-promoting drugs: a focus on cognitive
function. Human psychopharmacology. 2009;24(5):371-89.
1. Shimizu T. [Excessive daytime sleepiness (EDS)]. Nihon rinsho
Japanese journal of clinical medicine. 2015;73(6):937-41.
2. Kara B, Kilic O. Predictors of poor sleep quality and excessive
daytime sleepiness in Turkish adults with type 2 diabetes. Journal of
clinical nursing. 2015;24(9-10):1436-9.
3. Begley KJ, Castillo S, Steinshouer CR, Malesker MA. Role of
pharmacists in the treatment of excessive daytime sleepiness. The
Consultant pharmacist : the journal of the American Society of Consultant
Pharmacists. 2014;29(11):741-52.
4. Fong SY, Ho CK, Wing YK. Comparing MSLT and ESS in the
measurement of excessive daytime sleepiness in obstructive sleep
apnoea syndrome. Journal of psychosomatic research. 2005;58(1):55-60.
5. Schreier DR, Roth C, Mathis J. Subjective perception of
sleepiness in a driving simulator is different from that in the Maintenance
of Wakefulness Test. Sleep medicine. 2015;16(8):994-8.
6. Tanaka H. [Multiple sleep latency test, maintenance of
wakefulness test and suggestive immobilization test]. Nihon rinsho
Japanese journal of clinical medicine. 2015;73(6):971-9.
7. van der Heide A, van Schie MK, Lammers GJ, Dauvilliers Y,
Arnulf I, Mayer G, et al. Comparing Treatment Effect Measurements in
Narcolepsy: The Sustained Attention to Response Task, Epworth
Sleepiness Scale and Maintenance of Wakefulness Test. Sleep.
2015;38(7):1051-8.
8. Bioulac S, Chaufton C, Taillard J, Claret A, Sagaspe P,
Fabrigoule C, et al. Excessive daytime sleepiness in adult patients with
ADHD as measured by the Maintenance of Wakefulness Test, an
electrophysiologic measure. The Journal of clinical psychiatry.
2015;76(7):943-8.
9. Chen Z, Wu C, Zhong M, Lyu N, Huang Z. Identification of
common features of vehicle motion under drowsy/distracted driving: A
case study in Wuhan, China. Accident; analysis and prevention.
2015;81:251-9.
10. Sadeghniiat-Haghighi K, Moller HJ, Saraei M, Aminian O,
Khajeh-Mehrizi A. The Epworth Sleepiness Scale for screening of the
drowsy driving: comparison with the maintenance of wakefulness test in
an Iranian sample of commercial drivers. Acta medica Iranica.
2014;52(2):125-9.
11. Anderson C, Horne JA. Driving drowsy also worsens driver
distraction. Sleep medicine. 2013;14(5):466-8.
12. Dawson J. Putting the brakes on drowsy driving. The American
nurse. 2012;44(2):9.
13. Rosenberg RP. Clinical assessment of excessive daytime
sleepiness in the diagnosis of sleep disorders. The Journal of clinical
psychiatry. 2015;76(12):e1602.
14. Rose D, Gelaye B, Sanchez S, Castaneda B, Sanchez E, Yanez
ND, et al. Morningness/eveningness chronotype, poor sleep quality, and
daytime sleepiness in relation to common mental disorders among
Peruvian college students. Psychology, health & medicine.
2015;20(3):345-52.
15. Haregu A, Gelaye B, Pensuksan WC, Lohsoonthorn V,
Lertmaharit S, Rattananupong T, et al. Circadian rhythm characteristics,
poor sleep quality, daytime sleepiness and common psychiatric disorders
among Thai college students. Asia-Pacific psychiatry : official journal of the
Pacific Rim College of Psychiatrists. 2015;7(2):182-9.
16. Suzuki K, Miyamoto M, Miyamoto T, Inoue Y, Matsui K,
Nishida S, et al. The Prevalence and Characteristics of Primary Headache
and Dream-Enacting Behaviour in Japanese Patients with Narcolepsy or
Idiopathic Hypersomnia: A Multi-Centre Cross-Sectional Study. PloS one.
2015;10(9):e0139229.
17. Pereira D, Lopes E, da Silva Behrens NS, de Almeida Fonseca
H, Sguillar DA, de Araujo Lima TF, et al. Prevalence of periodical leg
movements in patients with narcolepsy in an outpatient facility in Sao
Paulo. Sleep Science. 2014;7(1):69-71.
18. Heier MS, Evsiukova T, Wilson J, Abdelnoor M, Hublin C, Ervik
S. Prevalence of narcolepsy with cataplexy in Norway. Acta neurologica
Scandinavica. 2009;120(4):276-80.
19. Bjorvatn B, Lehmann S, Gulati S, Aurlien H, Pallesen S, Saxvig
IW. Prevalence of excessive sleepiness is higher whereas insomnia is
lower with greater severity of obstructive sleep apnea. Sleep & breathing
= Schlaf & Atmung. 2015;19(4):1387-93.
20. Signal TL, Paine SJ, Sweeney B, Priston M, Muller D, Smith A,
et al. Prevalence of abnormal sleep duration and excessive daytime
sleepiness in pregnancy and the role of socio-demographic factors:
comparing pregnant women with women in the general population. Sleep
medicine. 2014;15(12):1477-83.
21. Liviya Ng W, Freak-Poli R, Peeters A. The prevalence and
characteristics associated with excessive daytime sleepiness among
Australian workers. Journal of occupational and environmental medicine /
American College of Occupational and Environmental Medicine.
2014;56(9):935-45.
22. Bjorvatn B, Pallesen S, Gronli J, Sivertsen B, Lehmann S.
Prevalence and correlates of insomnia and excessive sleepiness in adults
138
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
with obstructive sleep apnea symptoms. Perceptual and motor skills.
2014;118(2):571-86.
23. Hayley AC, Williams LJ, Kennedy GA, Berk M, Brennan SL,
Pasco JA. Prevalence of excessive daytime sleepiness in a sample of the
Australian adult population. Sleep medicine. 2014;15(3):348-54.
24. Verbruggen AE, Dieltjens M, Wouters K, De Volder I, Van de
Heyning PH, Braem MJ, et al. Prevalence of residual excessive sleepiness
during effective oral appliance therapy for sleep-disordered breathing.
Sleep medicine. 2014;15(2):269-72.
25. Vashum KP, McEvoy MA, Hancock SJ, Islam MR, Peel R, Attia
JR, et al. Prevalence of and associations with excessive daytime
sleepiness in an Australian older population. Asia-Pacific journal of public
health / Asia-Pacific Academic Consortium for Public Health.
2015;27(2):NP2275-84.
26. Risco J, Ruiz P, Marinos A, Juarez A, Ramos M, Salmavides F,
et al. Excessive sleepiness prevalence in public transportation drivers of a
developing country. Traffic injury prevention. 2013;14(2):145-9.
27. Gunes Z, Sahbaz M, Tugrul E, Gunes H. Prevalence and risk
factors for excessive daytime of sleepiness in rural western Anotolia
(Turkey): the role of obesity and metabolic syndrome. The Southeast Asian
journal of tropical medicine and public health. 2012;43(3):747-55.
28. Dunn D, Hostetler G, Iqbal M, Messina-McLaughlin P, Reiboldt
A, Lin YG, et al. Wake-promoting agents: search for next generation
modafinil: part I. Bioorganic & medicinal chemistry letters.
2012;22(6):2312-4.
29. Lesur B, Lin YG, Marcy VR, Aimone LD, Gruner J, Bacon ER, et
al. Aryl-heteroaryl derivatives as novel wake-promoting agents. Chemical
biology & drug design. 2013;81(3):429-32.
30. Louvet P, Schweizer D, Gourdel ME, Riguet E, Yue C, Marcy
VR, et al. Wake-promoting agents: search for next generation modafinil:
part IV. European journal of medicinal chemistry. 2012;54:949-51.
31. Dunn D, Hostetler G, Iqbal M, Marcy VR, Lin YG, Jones B, et al.
Wake promoting agents: search for next generation modafinil, lessons
learned: part III. Bioorganic & medicinal chemistry letters.
2012;22(11):3751-3.
32. Dunn D, Hostetler G, Iqbal M, Messina-McLaughlin P, Reiboldt
A, Lin YG, et al. Wake-promoting agents: search for next generation
modafinil: part II. Bioorganic & medicinal chemistry letters.
2012;22(6):2315-7.
33. Lynch J, Jr., Regan C, Stump G, Tannenbaum P, Stevens J,
Bone A, et al. Hemodynamic and cardiac neurotransmitter-releasing
effects in conscious dogs of attention- and wake-promoting agents: a
comparison of d-amphetamine, atomoxetine, modafinil, and a novel
quinazolinone H3 inverse agonist. Journal of cardiovascular
pharmacology. 2009;53(1):52-9.
34. Stocking EM, Letavic MA. Histamine H3 antagonists as wake-
promoting and pro-cognitive agents. Current topics in medicinal chemistry.
2008;8(11):988-1002.
35. Makris AP, Rush CR, Frederich RC, Kelly TH. Wake-promoting
agents with different mechanisms of action: comparison of effects of
modafinil and amphetamine on food intake and cardiovascular activity.
Appetite. 2004;42(2):185-95.
36. Oken BS, Salinsky MC, Elsas SM. Vigilance, alertness, or
sustained attention: physiological basis and measurement. Clinical
neurophysiology : official journal of the International Federation of Clinical
Neurophysiology. 2006;117(9):1885-901.
37. Ebrahim IO, Howard RS, Kopelman MD, Sharief MK, Williams
AJ. The hypocretin/orexin system. Journal of the Royal Society of Medicine.
2002;95(5):227-30.
38. McWhirter D, Bae C, Budur K. The Assessment, Diagnosis,
and Treatment of Excessive Sleepiness: Practical Considerations for the
Psychiatrist. Psychiatry (Edgmont). 2007;4(9):26-35.
39. Wisor JP, Nishino S, Sora I, Uhl GH, Mignot E, Edgar DM.
Dopaminergic role in stimulant-induced wakefulness. The Journal of
neuroscience : the official journal of the Society for Neuroscience.
2001;21(5):1787-94.
40. Ferraro L, Tanganelli S, O'Connor WT, Antonelli T, Rambert F,
Fuxe K. The vigilance promoting drug modafinil increases dopamine
release in the rat nucleus accumbens via the involvement of a local
GABAergic mechanism. European journal of pharmacology. 1996;306(1-
3):33-9.
41. Gerrard P, Malcolm R. Mechanisms of modafinil: A review of
current research. Neuropsychiatric disease and treatment.
2007;3(3):349-64.
42. Saper CB, Scammell TE. Modafinil: a drug in search of a
mechanism. Sleep. 2004;27(1):11-2.
43. Ishizuka T, Murotani T, Yamatodani A. Modanifil activates the
histaminergic system through the orexinergic neurons. Neuroscience
letters. 2010;483(3):193-6.
44. Broughton RJ, Fleming JA, George CF, Hill JD, Kryger MH,
Moldofsky H, et al. Randomized, double-blind, placebo-controlled
crossover trial of modafinil in the treatment of excessive daytime
sleepiness in narcolepsy. Neurology. 1997;49(2):444-51.
45. Kumar R. Approved and investigational uses of modafinil : an
evidence-based review. Drugs. 2008;68(13):1803-39.
46. Dauvilliers Y, Neidhart E, Billiard M, Tafti M. Sexual
dimorphism of the catechol-O-methyltransferase gene in narcolepsy is
associated with response to modafinil. The pharmacogenomics journal.
2002;2(1):65-8.
47. Saper CB, Scammell TE. Emerging therapeutics in sleep.
Annals of neurology. 2013;74(3):435-40.
48. Zaharna M, Dimitriu A, Guilleminault C. Expert opinion on
pharmacotherapy of narcolepsy. Expert opinion on pharmacotherapy.
2010;11(10):1633-45.
49. Golicki D, Bala MM, Niewada M, Wierzbicka A. Modafinil for
narcolepsy: systematic review and meta-analysis. Medical science monitor
: international medical journal of experimental and clinical research.
2010;16(8):RA177-86.
50. Schwartz JR, Roth T. Shift work sleep disorder: burden of
illness and approaches to management. Drugs. 2006;66(18):2357-70.
51. Schwartz JRL. Modafinil in the treatment of excessive
sleepiness. Drug design, development and therapy. 2008;2:71-85.
52. Nishino S, Okuro M. Armodafinil for excessive daytime
sleepiness. Drugs of today. 2008;44(6):395-414.
53. Randomized trial of modafinil for the treatment of
pathological somnolence in narcolepsy. US Modafinil in Narcolepsy
Multicenter Study Group. Annals of neurology. 1998;43(1):88-97.
54. Edgar CJ, Pace-Schott EF, Wesnes KA. Approaches to
measuring the effects of wake-promoting drugs: a focus on cognitive
function. Human psychopharmacology. 2009;24(5):371-89.
55. Berman SM, Kuczenski R, McCracken JT, London ED.
Potential adverse effects of amphetamine treatment on brain and
behavior: a review. Molecular psychiatry. 2009;14(2):123-42.
56. Banerjee D, Vitiello MV, Grunstein RR. Pharmacotherapy for
excessive daytime sleepiness. Sleep medicine reviews. 2004;8(5):339-
54.
57. Billiard M. Narcolepsy: current treatment options and future
approaches. Neuropsychiatric disease and treatment. 2008;4(3):557-66.
58. Akintomide GS, Rickards H. Narcolepsy: a review.
Neuropsychiatric disease and treatment. 2011;7:507 -18.
59. Cochen De Cock V, Dauvilliers Y. Current and future
therapeutic approaches in narcolepsy. Future Neurology. 2011;6(6):771-
82.
60. Guilleminault C, Carskadon M, Dement WC. On the treatment
of rapid eye movement narcolepsy. Archives of neurology. 1974;30(1):90-
3.
61. Tunnicliff G. Sites of Action of Gamma-Hydroxybutyrate
(GHB)A Neuroactive Drug with Abuse Potential. Journal of Toxicology:
Clinical Toxicology. 1997;35(6):581-90.
62. Tsujino N, Sakurai T. Orexin/hypocretin: a neuropeptide at the
interface of sleep, energy homeostasis, and reward system.
Pharmacological reviews. 2009;61(2):162-76.
63. Cao M, Guilleminault C. Hypocretin and its emerging role as a
target for treatment of sleep disorders. Current neurology and
neuroscience reports. 2011;11(2):227-34.
64. Equihua AC, De La Herran-Arita AK, Drucker-Colin R. Orexin
receptor antagonists as therapeutic agents for insomnia. Frontiers in
pharmacology. 2013;4:163.
65. Huang Z-L, Qu W-M, Eguchi N, Chen J-F, Schwarzschild MA,
Fredholm BB, et al. Adenosine A2A, but not A1, receptors mediate the
arousal effect of caffeine. Nature neuroscience. 2005;8(7):858-9.
66. Rihel J, Schier AF. Sites of action of sleep and wake drugs:
insights from model organisms. Current opinion in neurobiology.
2013;23(5):831-40.
67. Dauvilliers Y, Bassetti C, Lammers GJ, Arnulf I, Mayer G,
Rodenbeck A, et al. Pitolisant versus placebo or modafinil in patients with
narcolepsy: a double-blind, randomised trial. The Lancet Neurology.
2013;12(11):1068-75.
68. Hasan S, Pradervand S, Ahnaou A, Drinkenburg W, Tafti M,
Franken P. How to keep the brain awake? The complex molecular
pharmacogenetics of wake promotion. Neuropsychopharmacology :
official publication of the American College of Neuropsychopharmacology.
2009;34(7):1625-40.
69. Abbott A. Neuroscience: the molecular wake-up call. Nature.
2007;447(7143):368-70.
70. In Y, Tomoo K, Ishida T, Sakamoto Y. Crystal and molecular
structure of an (S)-(+)-enantiomer of modafinil, a novel wake-promoting
agent. Chemical & pharmaceutical bulletin. 2004;52(10):1186 -9.
71. Choi S, Kim MY, Joo KY, Park S, Kim JA, Jung JC, et al.
Modafinil inhibits K(Ca)3.1 currents and muscle contraction via a cAMP-
dependent mechanism. Pharmacological research. 2012;66(1):51-9.
139
Journal of Advanced Medical Sciences and Applied Technologies (JAMSAT). 2016; 2(1)
72. Tahsili-Fahadan P, Carr GV, Harris GC, Aston-Jones G.
Modafinil blocks reinstatement of extinguished opiate-seeking in rats:
mediation by a glutamate mechanism. Neuropsychopharmacology :
official publication of the American College of Neuropsychopharmacology.
2010;35(11):2203-10.
73. Mitchell HA, Bogenpohl JW, Liles LC, Epstein MP, Bozyczko-
Coyne D, Williams M, et al. Behavioral responses of dopamine beta-
hydroxylase knockout mice to modafinil suggest a dual noradrenergic-
dopaminergic mechanism of action. Pharmacology, biochemistry, and
behavior. 2008;91(2):217-22.
74. Wisor JP, Eriksson KS. Dopaminergic-adrenergic interactions
in the wake promoting mechanism of modafinil. Neuroscience.
2005;132(4):1027-34.
75. Xiao YL, Fu JM, Dong Z, Yang JQ, Zeng FX, Zhu LX, et al.
Neuroprotective mechanism of modafinil on Parkinson disease induced by
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Acta pharmacologica
Sinica. 2004;25(3):301-5.
76. Lee H, Kim KC, Cho MS, Suh SH, Hong YM. Modafinil improves
monocrotaline-induced pulmonary hypertension rat model. Pediatric
research. 2016.
77. Kuan YC, Wu D, Huang KW, Chi NF, Hu CJ, Chung CC, et al.
Effects of Modafinil and Armodafinil in Patients With Obstructive Sleep
Apnea: A Meta-Analysis of Randomized Controlled Trials. Clinical
therapeutics. 2016.
78. Ramachandra B. A Critical Review of Properties of Modafinil
and Analytical, Bio Analytical Methods for Its Determination. Critical
reviews in analytical chemistry / CRC. 2016:0.
79. Karila L, Leroy C, Dubol M, Trichard C, Mabondo A, Marill C, et
al. Dopamine Transporter Correlates and Occupancy by Modafinil in
Cocaine-Dependent Patients: A Controlled Study with High-Resolution PET
and [C]-PE2I. Neuropsychopharmacology : official publication of the
American College of Neuropsychopharmacology. 2016.
80. De Ron P, Dremier S, Winlow P, Jenkins A, Hanon E, Nogueira
da Costa A. Correlating behaviour and gene expression endpoints in the
dopaminergic system after modafinil administration in mouse. European
neuropsychopharmacology : the journal of the European College of
Neuropsychopharmacology. 2016.
81. Chapman JL, Vakulin A, Hedner J, Yee BJ, Marshall NS.
Modafinil/armodafinil in obstructive sleep apnoea: a systematic review
and meta-analysis. The European respiratory journal. 2016.
82. Andrade C. A method for deciding about the possible safety of
modafinil and armodafinil in patients with seizure disorder. The Journal of
clinical psychiatry. 2016;77(1):e25-8.
83. Battleday RM, Brem AK. Modafinil for cognitive
neuroenhancement in healthy non-sleep-deprived subjects: A systematic
review. European neuropsychopharmacology : the journal of the European
College of Neuropsychopharmacology. 2016;26(2):391.
84. Hosak L, Horacek J, Beranek M, Cermakova E. Molecular
heterosis in metamphetamine abusers. International journal of psychiatry
in clinical practice. 2007;11(3):250-2.
85. Cadet JL, Krasnova IN, Jayanthi S, Lyles J. Neurotoxicity of
substituted amphetamines: molecular and cellular mechanisms.
Neurotoxicity research. 2007;11(3-4):183-202.
86. Abdou MM. A molecular approach to the study of structure-
activity correlation for some amphetamines. Journal of psychoactive
drugs. 2001;33(3):295-300.
87. Ganjavi H, Shapiro CM. Hypocretin/Orexin: a molecular link
between sleep, energy regulation, and pleasure. The Journal of
neuropsychiatry and clinical neurosciences. 2007;19(4):413 -9.
88. Willie JT, Chemelli RM, Sinton CM, Tokita S, Williams SC,
Kisanuki YY, et al. Distinct narcolepsy syndromes in Orexin receptor-2 and
Orexin null mice: molecular genetic dissection of Non-REM and REM sleep
regulatory processes. Neuron. 2003;38(5):715-30.
89. Chemelli RM, Willie JT, Sinton CM, Elmquist JK, Scammell T,
Lee C, et al. Narcolepsy in orexin knockout mice: molecular genetics of
sleep regulation. Cell. 1999;98(4):437-51.
140
... The general treatment of fatigue in disorders other than insomnia is to use wake-promoting agents such as psychostimulants, or off-label bupropion, methylphenidate, modafinil, or amantadine [47,48]. Such drugs may have the potential for adverse reactions or carry the risk of abuse and dependence [47,49,50]. Involvement of the orexin system has been proposed in both multiple sclerosis pathology [51] and fatigue in patients with multiple sclerosis, but the data to date are conflicting [52]. ...
Article
Full-text available
Objective: Fatigue is a common symptom in patients with insomnia. This analysis evaluated whether treatment of night-time symptoms of insomnia with a dual orexin receptor antagonist, lemborexant, might also reduce fatigue. Methods: Analyses were conducted of two phase 3 studies of subjects with insomnia disorder. Subjects received placebo, lemborexant 5 mg, or lemborexant 10 mg in the 12-month (6 months placebo-controlled) Study E2006-G000-303 (Study 303: SUNRISE-2) of adults (N = 949; full analysis set [FAS]), and the 1-month, placebo- and active-controlled Study E2006-G000-304 (Study 304; SUNRISE-1) of older adults (females ≥55 years, males ≥65 years) (N = 1006; FAS). Fatigue was assessed using the Fatigue Severity Scale (FSS). Patient-reported sleep onset and maintenance endpoints were analyzed using data from electronic sleep diaries. Results: Lemborexant significantly reduced subject-reported fatigue versus placebo over a 6-month treatment period (FSS total score least squares mean treatment difference of -2.50 for 5 mg and -2.56 for 10 mg of lemborexant; p < 0.05 for both). This reduction was sustained over 12 months of lemborexant in both the overall population and in subjects with clinically meaningful fatigue (FSS total score ≥36) at baseline. Improvements in fatigue over time positively correlated with improvements in sleep onset and maintenance parameters. Improvements in sleep quality were evident as early as 1 week after lemborexant treatment, whereas longer-term treatment (>1 month) may be needed for improvements in insomnia-related fatigue. Conclusions: In addition to improving sleep onset and sleep maintenance in subjects with insomnia disorder, lemborexant provides further benefit by reducing daytime fatigue. Clinical trial registration: https://clinicaltrials.gov/ct2/show/NCT02952820 and https://clinicaltrials.gov/ct2/show/NCT02783729.
Article
Full-text available
Modafinil or armodafinil (ar/mod) may be considered for patients with approved or unapproved indications, including excessive daytime drowsiness, fatigue, attention-deficit/hyperactivity disorder (ADHD), or addictions. Ar/mod is classified as a psychostimulant, and psychostimulants have been associated with a small risk of seizures. There is no guidance about the use of ar/mod in patients who are at risk of seizures. This article suggests how a physician may explore the safety of ar/mod if indicated in a patient at such risk. In summary, reading the prescribing information, writing to the drug manufacturer, and searching research databases suggest the following: Ar/mod and its metabolites and derivatives have dose-dependent anticonvulsant action in animal models; ar/mod is not associated with seizures as an adverse event in populations at risk, such as those with ADHD, head injury, and brain tumors; it is not associated with worsening of seizure disorder in patients with current seizure disorder; and it is not associated with seizures in overdose. These findings are reassuring. However, not all the data are of high quality, and potential ar/mod interactions with antiepileptic drugs (and other concurrent medications that affect the seizure threshold) need to be considered because ar/mod can induce the metabolism of some drugs and inhibit the metabolism of others. Decisions should be individualized, and decision-making should be a shared effort between patient and physician.
Article
Full-text available
Modafinil is an FDA-approved eugeroic that directly increases cortical catecholamine levels, indirectly upregulates cerebral serotonin, glutamate, orexin, and histamine levels, and indirectly decreases cerebral gamma-amino-butrytic acid levels. In addition to its approved use treating excessive somnolence, modafinil is thought to be used widely off-prescription for cognitive enhancement. However, despite this popularity, there has been little consensus on the extent and nature of the cognitive effects of modafinil in healthy, non-sleep-deprived humans. This problem is compounded by methodological discrepancies within the literature, and reliance on psychometric tests designed to detect cognitive effects in ill rather than healthy populations. In order to provide an up-to-date systematic evaluation that addresses these concerns, we searched MEDLINE with the terms "modafinil" and "cognitive", and reviewed all resultant primary studies in English from January 1990 until December 2014 investigating the cognitive actions of modafinil in healthy non-sleep-deprived humans. We found that whilst most studies employing basic testing paradigms show that modafinil intake enhances executive function, only half show improvements in attention and learning and memory, and a few even report impairments in divergent creative thinking. In contrast, when more complex assessments are used, modafinil appears to consistently engender enhancement of attention, executive functions, and learning. Importantly, we did not observe any preponderances for side effects or mood changes. Finally, in light of the methodological discrepancies encountered within this literature, we conclude with a series of recommendations on how to optimally detect valid, robust, and consistent effects in healthy populations that should aid future assessment of neuroenhancement.
Article
Modafinil is a synthetic molecule used for the treatment of narcolepsy. Narcolepsy is a neurological disorder, due to which people experience frequent excessive daytime sleepiness. Nevertheless, there are some concerns about modafnil quality control. The modafinil enantiomers are both biologically active. However, it has been reported that the pharmacological properties of the both enantiomers are different and that the S-enantiomer is eliminated three times faster than the R-enantiomer. Therefore, most of the pharmaceutical companies have shifted to produce of armodafinil (R- enantiomer) instead of the racemate. This article discusses about the critical review of the literature, the impact of the use of modafinil in the treatment of narcolepsy patients and other diseases, its physicochemical properties, toxicological properties, synthetic methods, analytical and bio analytical methods, and challenges that remain in order to ensure the quality. This article mainly focused on review of process related impurities, enantiomeric separation, metabolites of modafinil in various biofluids and pharmaceutical formulations using HPLC, LC-MS, GC-MS, CE, HPTLC and spectrophotometric methods.
Article
Background: Pulmonary arterial hypertension (PAH) progressively leads to increases in pulmonary vasoconstriction. Modafinil plays a role in vasorelaxation and blocking KCa3.1 channel with a result of elevating intracellular cAMP levels. The purpose of this study is to evaluate the effects on modafinil in monocrotaline (MCT)-induced PAH rat. Methods: The rats were separated into 3 groups: the control group; the monocrotaline (M) group (MCT 60 mg/kg); the modafinil (MD) group (MCT 60 mg/kg + modafinil. Results: Reduced right ventricular pressure (RVP) was observed in the MD group. Right ventricular hypertrophy was improved in the MD group. Reduced number of intra-acinar pulmonary arteries and medial wall thickness were noted in the MD group. After the administration of modafinil, protein expressions of endothelin-1 (ET-1), endothelin receptor A (ERA) and KCa3.1 channel were significantly reduced. Modafinil suppressed pulmonary artery smooth muscle cell (PASMC) proliferation via cAMP and KCa3.1 channel. Additionally, we confirmed protein expressions such as Bcl-2-associated X, vascular endothelial growth factor, tumor necrosis factor-a and interleukin-6 were reduced in the MD group. Conclusion: Modafinil improved PAH by vasorelaxation and a decrease in medial thickening via ET-1, ERA and KCa3.1 down regulation. This is a meaningful study of a modafinil in PAH model.Pediatric Research (2016); doi:10.1038/pr.2016.38.
Article
Purpose: Obstructive sleep apnea (OSA) is associated with nocturnal hypoxemia, excessive daytime sleepiness (EDS), and sympathetic hyperactivation. Continuous positive airway pressure is the first-line treatment for OSA. However, some patients may have residual EDS. Modafinil and its R-enantiomer, armodafinil, are wakefulness-promoting agents known to be effective in alleviating sleepiness. Methods: We performed a systematic review and meta-analysis of data from published randomized controlled trials (RCTs) that evaluated the efficacy of modafinil and armodafinil in treating EDS in patients with OSA. Electronic databases, including PubMed, EMBASE, and the Cochrane Central Register of Controlled Trials, were searched for articles on OSA published before October 2015. Findings: We identified 11 RCTs of modafinil involving 723 patients and 5 RCTs of armodafinil involving 1009 patients. A pooled estimate of the mean differences in sleepiness parameters versus placebo were calculated using the random-effects model. Epworth Sleepiness Scale scores improved significantly in the modafinil group (weighted mean difference [WMD], -2.96 [95% confidence interval (CI), -3.73 to -2.19]) and in the armodafinil group (WMD, -2.63; 95% CI, -3.4 to -1.85) compared with those in the placebo group. Sleep latency, as measured on the Maintenance of Wakefulness Test, was significantly prolonged in the modafinil group (WMD, 2.51 [95% CI, 1.5-3.52]) and in the armodafinil group (WMD, 2.71 [95% CI, 0.04-5.37]). Patients tolerated the adverse events with both medications well. Implications: The findings from our study suggest that both modafinil and armodafinil significantly improved subjective and objective daytime sleepiness. Thus, modafinil and armodafinil may be recommended to patients with OSA, particularly those with EDS.
Article
Modafinil is a candidate compound for the treatment of cocaine addiction that binds to the dopamine transporter (DAT) in healthy humans, as observed by positron emission tomography (PET). This mechanism, analogous to that of cocaine, might mediate a putative therapeutic effect of modafinil on cocaine dependence. Though, the binding of modafinil to DAT has never been assessed in cocaine-dependent patients. We aimed at quantifying the DAT availability during a controlled treatment by modafinil, and its clinical and psychometric correlates in cocaine-dependent patients at the onset of abstinence initiation. Twenty-nine cocaine-dependent male patients were enrolled in a 3-month trial for cocaine abstinence. Modafinil was used in a randomized double-blind placebo-controlled design and was administered as follows: 400 mg/day for 26 days, then 300 mg/day for 30 days and 200 mg/day for 31 days. Participants were examined twice during a seventeen-day hospitalization for their DAT availability using PET and [(11)C]-PE2I, and for assessments of craving, depressive symptoms, working memory, and decision-making. Cocaine abstinence was further assessed during a ten-week outpatient follow-up period. Baseline [(11)C]-PE2I binding potential covaried with risk-taking and craving index in striatal and extrastriatal regions. A 65.6% decrease of binding potential was detected in patients receiving modafinil for two weeks, whereas placebo induced no significant change. During hospitalization, an equivalent improvement in clinical outcomes was observed in both treatment groups, and during the outpatient follow-up there were more therapeutic failures in the modafinil-treated group. Therefore, these results do not support the usefulness of modafinil to treat cocaine addiction.Neuropsychopharmacology accepted article preview online, 19 February 2016. doi:10.1038/npp.2016.28.
Article
The mechanisms of action of modafinil continue to be poorly characterised and its potential for abuse in preclinical models remains controverted. The aim of this study was to further elucidate the mechanism of action of modafinil, through a potential behavioural and molecular association in the mouse. A conditioned place preference (CPP) paradigm was implemented to investigate the rewarding properties of modafinil. Whole genome expression and qRT-PCR analysis were performed on the ventral tegmental area (VTA), nucleus accumbens (NAC) and prefrontal cortex (PFC) of modafinil-treated and control animals. Modafinil administration (65 mg/kg) induced an increase in locomotor activity, an increase in the change of preference for the drug-paired side after a conditioning period as well as changes to gene expression profiles in the VTA (120 genes), NAC (23 genes) and PFC (19 genes). A molecular signature consisting of twelve up-regulated genes was identified as common to the three brain regions. Multiple linear correlation analysis showed a strong correlation (R2>0.70) between the behavioural and molecular endpoints in the three brain regions. We show that modafinil had a concomitant effect on CPP, locomotor activity, and up-regulation of interferon-γ (IFN-γ) regulated genes (Gbp2, Gbp3, Gbp10, Cd274, Igtp), while correlating the latter set of genes with behaviour changes evaluated through the CPP. A potential association can be proposed based on the dysregulation of p47 family genes and Gbp family of IFN-γ induced GTPases. In conclusion, these findings suggest a link between the behavioural and molecular events in the context of modafinil administration.
Article
To test whether sleep-deprived, healthy subjects who do not always signal spontaneously perceived sleepiness (SPS) before falling asleep during the Maintenance of Wakefulness Test (MWT) would do so in a driving simulator. Twenty-four healthy subjects (20-26 years old) underwent a MWT for 40 min and a driving simulator test for 1 h, before and after one night of sleep deprivation. Standard electroencephalography, electrooculography, submental electromyography, and face videography were recorded simultaneously to score wakefulness and sleep. Subjects were instructed to signal SPS as soon as they subjectively felt sleepy and to try to stay awake for as long as possible in every test. They were rewarded for both "appropriate" perception of SPS and staying awake for as long as possible. After sleep deprivation, seven subjects (29%) did not signal SPS before falling asleep in the MWT, but all subjects signalled SPS before falling asleep in the driving simulator (p <0.004). The previous results of an "inaccurate" SPS in the MWT were confirmed, and a perfect SPS was shown in the driving simulator. It was hypothesised that SPS is more accurate for tasks involving continuous feedback of performance, such as driving, compared to the less active situation of the MWT. Spontaneously perceived sleepiness in the MWT cannot be used to judge sleepiness perception while driving. Further studies are needed to define the accuracy of SPS in working tasks or occupations with minimal or no performance feedback. Copyright © 2015 Elsevier B.V. All rights reserved.
Article
In search of a next generation molecule to the novel wake promoting agent modafinil, a series of diphenyl ether derived wakefulness enhancing agents (in rat) was developed. From this work, racemic compound 16 was separated into its chiral enantiomers to profile them individually.
Article
To quantify the objective level of sleepiness in adult attention-deficit/hyperactivity disorder (ADHD) patients and to determine the relationship between excessive daytime sleepiness and simulated driving performance. Forty adult ADHD patients (DSM-IV criteria) and 19 matched healthy control subjects were included between June 30, 2010, and June 19, 2013. All participants completed the Epworth Sleepiness Scale and the Manchester Driving Behavior Questionnaire. After nocturnal polysomnography, they performed 2 neuropsychological tests, a 4 × 40-minute Maintenance of Wakefulness Test, and a 1-hour driving session. The primary outcome measure was the mean sleep latency on the Maintenance of Wakefulness Test. ADHD patients were divided into 3 groups defined by their Maintenance of Wakefulness Test scores. Participants (patients and control subjects) were allocated as follows: sleepy ADHD (0-19 min), intermediate ADHD (20-33 min), alert ADHD (34-40 min), and control group (34-40 min). The driving performance outcome was the mean standard deviation of lateral position of the vehicle during the simulated session. The group mean (SD) Epworth Sleepiness Scale score was higher in ADHD patients (12.1 [4.4]) than in controls (6.0 [2.7]) (P < .001). On the basis of the Maintenance of Wakefulness Test scores, 14 patients (35%) were in the sleepy group, 20 (50%) were in the intermediate group, and only 6 (15%) were in the alert group. Sleepy ADHD patients exhibited significantly deteriorated driving performance compared to the other 3 groups (P < .01). Our study shows that a significant proportion of adult ADHD patients exhibit an objective excessive daytime sleepiness, which, in addition, has an impact on simulated driving performance. Excessive daytime sleepiness, therefore, may be a key element needed to better evaluate these ADHD patients. ClinicalTrials.gov identifier: NCT01160874. © Copyright 2015 Physicians Postgraduate Press, Inc.