Vous êtes sur la page 1sur 14

IJoDR A pharmacological view on lucid dream induction

Can we induce lucid dreams? A pharmacological


point of view
Firas Hasan Bazzari
Faculty of Pharmacy, Cairo University, Cairo, Egypt

Summary. The phenomenon of lucid dreaming, in which an individual has the ability to be conscious and in control of his
dreams, has attracted the public attention, especially in the era of internet and social media platforms. With its huge pop-
ularity, lucid dreaming triggered passionate individuals, particularly lucid dreamers, to spread their thoughts and experi-
ences in lucid dreaming, and provide a number of tips and techniques to induce lucidity in dreams. Scientific research in
the field of sleep and dreams has verified the phenomenon of lucid dreaming for decades. Nevertheless, various aspects
regarding lucid dreaming are not fully understood. Many hypotheses and claims about lucid dreaming induction are yet
to be validated, and at present lucid dreaming still lacks efficient and reliable induction methods. Understanding the
molecular basis, brain physiology, and underlying mechanisms involved in lucid dreaming can aid in developing novel
and more target-specific induction methods. This review will focus on the currently available scientific findings regarding
neurotransmitters’ behavior in sleep, drugs observed to affect dreams, and proposed supplements for lucid dreaming, in
order to discuss the possibility of inducing lucid dreams from a pharmacological point of view.

Keywords: Lucid dreaming, Dreams, REM sleep, Neurotransmitters, Supplements, Pharmacology of lucid dreaming.

1. Introduction different methods and labeled according to the method’s


success rate in inducing lucid dreams. Techniques, such as
Lucid dreaming is a unique psychological phenomenon in
mnemonic induced lucid dreams (MILD), reflection/reality
which a dreaming individual is aware that he/she is dreaming
testing, Tholey’s combined technique, light stimulus, and
(Voss, 2010). As many other distinctive unfamiliar incidences
wake back to bed (WBTB) (in combination with MILD), were
in science, lucid dreaming has been described as a prepos-
labeled to be successful methods in inducing lucid dreams
terous beyond-belief phenomenon with a lot of skepticism.
based on the findings of available scientific evidence (Stum-
However, hypotheses about lucid dreaming have been trail-
brys et al., 2012). Nowadays, lucid dreaming practitioners,
blazing towards becoming close to a fact, with a wide num-
who claim to practice and experience lucid dreaming fre-
ber of individuals from all around the globe coming together
quently, are becoming more active on various social media
to share their experience of lucidity while dreaming. It is also
platforms, in which they speak out to the public about the
suggested that lucid dreaming has been practiced for cen-
benefits of lucid dreaming, such as controlling nightmares
turies and since ancient times as some references may go
and increasing creativity. Nonetheless, what is truly becom-
back to ancient Greek (LaBerge, 1988). However, the term
ing a major concern is promoting products that insinuate
“lucid dream” was first used by the Dutch scientist Freder-
their ability to induce lucid dreaming. Even though the prod-
ik van Eeden in his 1913 study “A study of dreams” (Van
ucts’ efficacy is yet to be confirmed and validated, over-
Eeden, 1913). In a 2016 systematic meta-analysis review
excited individuals who want to experience lucid dreaming
assessing 34 published articles in the past 50 years in order
would seek such products, which could be very dangerous
to achieve approximate values of lucid dreaming prevalence
and harmful in some cases. Therefore, the major question
and frequency. The study analysis obtained a prevalence
to be answered, is the current use of drugs considered a
mean of 55% in all 34 studies and a frequency mean of 23%
feasible and practical approach to induce lucid dreaming?
in a set of 25 studies (Saunders et al., 2016). Multiple meth-
In order to find a suitable answer for this question, a broad
ods to induce lucid dreaming have been scientifically evalu-
number of aspects related to the mechanism of dreaming
ated in some empirical studies to test their effectiveness. In
should be examined and discussed. In pharmacology, the
a 2012 paper by Stumbrys et al., induction techniques were
molecular basis and physiological mechanisms of the body
mainly categorized into; cognitive, external stimulation, and
form the targets for drug therapy; in other words, the drug’s
miscellaneous techniques, each containing a number of
therapeutic activity is a result of an interaction between the
drug and a target in the body, such as a specific enzyme or
a receptor. Thus, in order to conclude the potential activity
of a drug in inducing lucid dreams, there should be a clear
Corresponding address: view about its mechanism of action or at least a suggested
Firas Hasan Bazzari, Pharmacology & Toxicology Department, hypothesis about its mechanism supported by a scientific
El-Manial, Faculty of Pharmacy, Cairo University, Cairo, Egypt. evidence. This paper will overview several aspects including
Email: firas_hasan@rocketmail.com neurotransmission systems functionality, medical agents af-
fecting dreams, supplements proposed for lucid dreaming,
Submitted for publication: November 2017 and a discussion of several factors that participate in hin-
Accepted for publication: May 2018 dering drug application in lucid dreaming.

106 International Journal of Dream Research Volume 11, No. 2 (2018)


A pharmacological view on lucid dream induction IJoDR

2. Neurotransmitters in sleep & dreams promote REM-generation (Fraigne et al., 2015). In a sug-
gested scenario of natural REM-generation, muscle paraly-
Neurotransmitters form the molecular basis of any signal- sis (atonia) during REM is mainly achieved via the inhibition
ing pathway in the brain. Changes in the behavior of neu- of skeletal motor neurons by GABAergic/glycine neurons in
rotransmitters have been linked to many physiological and the spinal cord and ventromedial medulla (Brooks & Peever,
pathophysiological processes in the human body, some of 2012). While GABA agonists are observed to induce seda-
which are well understood while the majority are surrounded tion and promote sleep, GABAergic activity in the pontine
by hypotheses that are yet to be verified. In pharmacology, reticular formation is observed to increase wakefulness
neurotransmitters are considered vital targets for drug ther- (Watson et al., 2012). In mice, blockade of GABAA recep-
apy, as various medical agents are available and known to tors in the pontine reticular formation via bicuculline, a
control the levels of neurotransmitters whether to increase GABAA antagonist, was observed to reduce wakefulness
or inhibit their activity. This control can be achieved via sev- and increase both non-rapid eye movement (NREM) and
eral pathways on the level of neurotransmitter synthesis, re- REM sleep (Flint et al., 2010). Glutamate exerts its excit-
lease, receptor activation, reuptake, and enzymatic degra- atory action via binding to α-amino-3-hydroxy-5-methyl-4-
dation. For example, antipsychotics used in the treatment of isoxazole propionic acid (AMPA), kainic acid (KARs), and
a number mental conditions; bipolar disorder, schizophrenia N-methyl-D-aspartate (NMDA) receptors. Glutamatergic
and others, are identified to exert their action via dopamine neurons constitute the majority of REM-active neurons in
inhibition (Glick et al., 2001). Nevertheless, even with se- subcoeruleus nucleus, which suggests the involvement of
lective targeting of a single neurotransmitter; for example, glutamate in REM-generation; in addition, glutamate levels
selective serotonin reuptake inhibitors (SSRIs), there are have been observed to be higher during REM compared to
several side effects reported with the use of these medical NREM sleep (Fraigne et al., 2015; Dash et al., 2009). Ace-
agents. The major point is that neurotransmission systems tylcholine is suggested to participate in multiple pathways
are widely distributed (i.e. not in a specific brain region) and in REM sleep; for instance, cholinergic projections from the
function in harmony together in a very complex manner. laterodorsal tegmental and pedunculopontine tegmental
Generally, a number of neurotransmitters; acetylcholine, his- nuclei to the pontine reticular formation is found to promote
tamine, norepinephrine, serotonin, hypocretin also known REM-generation, and endogenous acetylcholine release in
as “orexin”, gamma-aminobutyric acid (GABA), and gluta- the pontine reticular formation is found to be higher dur-
mate, are all suggested to take a part in both consciousness ing REM sleep compared to NREM or wakefulness (Watson
and sleeping (Siegel, 2004). GABA is considered the central et al., 2012). Moreover, acetylcholine is found to promote/
player in suppressing neuronal activity and promoting sleep; trigger atonia either directly via activating neurons of sub-
in contrast, all of the previously mentioned neurotransmit- laterodorsal nucleus or indirectly by increasing glutamate
ters have, to some extent, an effect on GABAergic trans- activity, which was assessed via using carbachol (a cholin-
mission; thus, participating in consciousness mechanism ergic agonist) and in vitro patch clamp recording (Weng et
(Siegel, 2004). Acetylcholine has been strongly suggested al., 2014). Despite the fact that acetylcholine was thought to
to maintain consciousness and cognitive functions (Woolf be mandatory for REM sleep initiation, newer investigations
& Butcher, 2011). In a 2016 study, the release of acetylcho- suggest that REM-generation is acetylcholine independent
line and norepinephrine from the ascending fibers is found (Grace et al., 2014). Nonetheless, blockade of cholinergic
to be highly active in the state of wakefulness (Becchetti & activity via scopolamine has reduced NREM to REM tran-
Amadeo, 2016). Moreover, neuromodulators other than the sition and lessened theta-oscillations in the hippocampus;
classic neurotransmitters system, such as nitric oxide (NO), thus, suggesting that acetylcholine is critical for maintaining
were also suggested to play a role in the wake/sleep cycle REM sleep, yet it plays a minor role in REM initiation (Grace
(Cudeiro et al., 2000; Gautier-Sauvigné et al., 2005). The et al., 2014). Melanin-concentrating hormone neurons that
proposed mechanism of NO in the wake/sleep cycle is sug- are primarily found in dorsal raphe nucleus, are other REM
gested to be via the transition between and maintenance active neurons. In a rat model, microinjections of melanin-
of awaking and rapid eye movement (REM) sleep balance; concentrating hormone into the dorsal raphe nucleus has
moreover, it is suggested to have a role in the activation of resulted in an elevated number of NRER to REM transitions,
the cortical cholinergic neurons (Mariño & Cudeiro, 2006). which suggests their importance in REM-generation (Lagos
Other studies have also pointed out the potential of NO and et al., 2009).
adenosine in sleep homeostasis (Kalinchuk et al., 2011). Generally, other neurotransmitters; serotonin, noradrena-
REM sleep cycle, also known as “paradoxical sleep”, line, dopamine, hypocretin and histamine, are believed to
is suggested to be the core of dreams; in addition, lucid promote wakefulness (Watson et al., 2010). However, their
dreaming is frequently observed to be more prevalent dur- exact effects on sleep architecture are still not unclear. Se-
ing REM sleep compared to other sleep cycles (Stumbrys rotonin has a wide range of various receptors subfamilies
& Erlacher, 2012). Moreover, alterations in REM sleep have (5-HT1 – 5-HT7), which adds up to the complexity of its
been linked to the development of some conditions, such underlying mechanisms involved in sleep (Fink & Göthert,
as REM sleep behavior disorder (RBD), cataplexy, and nar- 2007). Earlier studies have suggested that serotonin plays
colepsy (Dauvilliers et al., 2014; Peever et al., 2014). Thus, a permissive role in sleep; nevertheless, newer evidence
there has been an increased interest in determining the observed high levels of serotonin during waking state in
neuronal circuits involved in REM sleep. The pivotal REM- most of the cortical and subcortical regions (Portas et al.,
generating circuit in the brainstem is suggested to be local- 2000). In some studies, a number of serotonin receptors
ized in the mesopontine junction, and REM-active neurons that influence REM sleep have been identified; for instance,
can be found in the subcoeruleus nucleus (Fraigne et al., serotonin activation of; 5HT1A, 5HT1B, and 5HT2A/2C, is sug-
2015). Furthermore, the dorsal paragigantocellular reticu- gested to lower REM sleep (Watson et al., 2010). Moreover,
lar nucleus in medulla, which contains mainly GABAergic serotonin is also found to suppress cataplexy, as serotonin
neurons, is suggested to inhibit wake-promoting areas and

International Journal of Dream Research Volume 11, No. 2 (2018) 107


IJoDR A pharmacological view on lucid dream induction

neurons in the dorsal raphe nucleus are considered the only observed to affect dreams but also recommended as
downstream target of hypocretin (Hasegawa et al., 2017). substances to enhance dream recall and lucidity with low
On the other hand, serotonin microinjection into amygdala adverse events (LaBerge, 2004). Their mode of action is to
during NREM reduced the transition latency to REM sleep boost cholinergic activity via inhibiting acetylcholinesterase,
in rats (Sanford et al., 1995). Dopamine is strongly asso- an enzyme responsible for acetylcholine degradation in the
ciated with wakefulness and reduction of both NREM and synapses (Colovic et al., 2013). Examples include medical
REM sleep (Isaac & Berridge, 2003; Andersen et al., 2009). agents used mainly for the treatment of Alzheimer’s disease,
Dopamine also participates in RBD pathophysiology via in- such as donepezil, rivastigmine, and galantamine (Stumbrys
hibiting dorsal subcoeruleus nucleus neurons, which is sug- & Erlacher, 2014). Further studies have established more
gested to be through its activity on α2-adrenergic receptors evidence regarding the use of AChEIs; for instance, galan-
instead of its endogenous receptors (Yang et al., 2014). In tamine administration increased the dreams’ length and
a study on patients with Parkinson disease to investigate vividness in lucid dreamers compared to non-galantamine
the role of the dopaminergic system in dream recall, has users (Sparrow et al., 2015). The use of serotonergic anti-
found that dreams visual vividness positively correlates to depressants; selective serotonin reuptake inhibitors (SSRIs)
the volume of amygdala and the thickness of subcortical‐ (e.g. fluoxetine, paroxetine, sertraline, citalopram) and sero-
medial prefrontal cortex. The study concluded that these tonin-norepinephrine reuptake inhibitors (SNRIs) (e.g. ven-
findings may suggest the involvement of the dopaminer- lafaxine), has resulted in a reduction of REM sleep duration
gic system in dream generation and recall (De Gennaro et and an increase REM latency (Winkelman & James, 2004).
al., 2016). The noradrenergic system of locus coeruleus is On the other hand, the withdrawal of; SSRIs, SNRIs, tricy-
found to promote wakefulness and decrease REM sleep clic antidepressants (TCAs), and monoamine oxidase inhibi-
(Watson et al., 2010). Noradrenaline pathway in inhibiting tors (MAOIs), led to more vivid dreams incidences (Keks et
REM sleep is suggested to be by acting on the cholinergic al., 2016). Antipsychotics effects on sleep are still not clear,
REM-active neurons; however, noradrenaline release is in- while some studies have shown promising findings in reduc-
hibited by GABA for normal REM regulation (Pal & Mallick, ing nightmares associated with post-traumatic stress disor-
2006). While noradrenaline major action is to inhibit REM der, others observed an increase in dreams frequency and
sleep, bilateral microinjections of prazosin (α1-antagonist), nightmares (Adetunji et al., 2005; Stanniland & Taylor, 2000).
clonidine (α2-agonist), and propranolol (ß-antagonist) into Loxapine, chlorpromazine, trifluoperazine, and haloperidol,
the pedunculopontine tegmentum, a group of cholinergic are all observed to increase sleep quantity, decrease sleep
neurons posterior to substantia nigra and next to the supe- latency, prolong REM latency, and lower REM duration (Foral
rior cerebellar peduncle, have increased the total time spent et al., 2011). Dopamine agonists used in the treatment of
in REM sleep (Pal & Mallick, 2006). At last, increased his- Parkinson disease are observed to have a general increase
tamine activity has a prominent role in wakefulness regula- in bad dreams and excessive daytime sleepiness (Happe
tion (Ramesh et al., 2004). Histamine deficiency in histidine- et al., 2001). Chronic treatment with levodopa, a dopamine
decarboxylase knockout mice has shown high-amplitude precursor used in Parkinson disease, has participated in the
theta wave bursts during REM (Bastianini et al., 2016). Suc- development of vivid dreams, night terrors, and nightmares
cessful dream recall is found to be associated to a higher (Sharf et al., 1978). Vivid dreams as adverse events were
theta activity in the frontal brain region with values of 5 – 7 also reported with use of other dopamine agonists, such
Hz, especially after REM morning awakening (Marzano et as ropinirole, bromocriptine, and pramipexole (Im et al.,
al., 2011). However, it has been observed that monoamine 2003; Pinter et al., 1999). In a study on controlled-release
containing neurons, such as adrenergic, serotonergic, and anticonvulsants, carbamazepine, lamotrigine, and gaba-
histaminergic neurons, are highly active during wakefulness pentin were used to assess their effects on nocturnal sleep
and their activity lessen during NREM and further reduced in epileptic patients. The study findings showed that acute
in REM sleep. Therefore, it is thought that they are not es- administration of carbamazepine has reduced REM sleep
sential for REM-generation, but more likely modulate REM and increased REM fragmentation. In contrast, lamotrigine
expression (Siegel, 2005). increased REM sleep and decreased percentage of slow-
wave sleep (SWS), and gabapentin increased REM sleep
3. Drugs affecting sleep & dreams duration with a lower number of awakenings (Placidi et al.,
2000). Older anticonvulsants (e.g. barbiturates, phenytoin,
In this section, a number of medical agents observed to carbamazepine) tend to lower sleep latency, reduce REM,
have an effect on sleep and dreams will be discussed. and increase daytime drowsiness. On the other hand, newer
Medical agents will be categorized according to their major ones (e.g. levetiracetam, lamotrigine, gabapentin) are ob-
pharmacological classes. The rationale is attempting to de- served to increase REM sleep (Bazil, 2003). In a case report
termine potential drug candidates for lucid dreaming induc- of a 42-year old woman with bipolar disorder, lamotrigine
tion and/or exclude less relevant ones, based on their ef- dose of 100mg/day was observed to lower sleep continuity
fects observed in clinical investigations. In addition, clinical and increase vivid dream-like experiences; however, lower-
findings regarding drugs that target neurotransmitters may ing lamotrigine dose has ameliorated these events (Uher &
enhance the current understanding of the neurotransmis- Jones, 2006). Such findings may highlight the importance of
sion systems’ behavior in relation to sleep and dreams. considering dose-dependent events in order to evaluate the
actual effects of drugs. Therefore, it is well recommended
3.1. Central nervous system drugs to include a range of different drug dose in any futuristic
Medical agents in this group have a superior ability to pen- studies intended to test drug-induced lucid dreams. Lastly,
etrate into the central nervous system (CNS); thus, hav- benzodiazepines are mainly found to shorten sleep latency,
ing more tendency to alter sleep and dreams. Acetylcho- reduce REM sleep, reduce SWS, increase sleep continuity,
linesterase inhibitors (AChEIs) top the list, as they are not and daytime somnolence (Bazil, 2003).

108 International Journal of Dream Research Volume 11, No. 2 (2018)


A pharmacological view on lucid dream induction IJoDR

3.2. Drugs for cardiovascular conditions niques was observed to guide the content of dream recalls
with a 90% probability, and increase recallable dream ratio
Vivid dreams and nightmares have been reported with the independently from anesthetic method used (Gyulaházi et
use of beta-blockers (Chrysant et al., 2008). The relative li- al., 2015). Many studies have reported on the elevated vivid
pophilicity of some beta-blockers; for instance, propranolol, and recallable dreams incidence during or after anesthesia
atenolol, metoprolol, and carvedilol, may explain their CNS with propofol and ketamine use (Eer et al., 2009; Blagrove
related adverse events (Bazzari et al., 2018). Their exact ef- et al., 2009). Generally, ketamine was linked to a prolonged
fect on sleep cycles and dreams are yet to be determined; REM sleep; however, propofol possess a REM suppressive
nonetheless, they are suggested to have a REM sleep sup- activity (Lazic et al., 2017; Kondili et al., 2012). Ultimately,
pressive activity (Thompson et al., 1999). Angiotensin-con- even there is a number of distinct physiological differences
verting enzyme inhibitors, calcium channel blockers, and between sleep and anesthesia; however, developing evi-
diuretics are also found to induce sleep disturbances (Mor- dence suggests that they are more similar than previously
gan et al., 2001). Different doses of clonidine, a centrally thought, and anesthesia may exert its action via the activa-
acting antihypertensive agent (α2-agonist), were observed tion of neural circuits commonly related to sleep (Tung &
to vary in their effects on sleep. Single administration of a Mendelson, 2004).
small 25µg clonidine dose was observed to significantly in-
crease REM and decrease NREM; in contrast, medium dose
of 150µg had the opposite effects. A possible explanation
3.5. Substances of abuse
is that the low dose acts pre-synaptically on α2-adrenergic Common substances of abuse; alcohol, cocaine, heroin,
receptor of noradrenergic neurons in locus coeruleus, which amphetamines, and opiates, are found to influence individu-
in turn reduces the release of noradrenaline. On the other als’ dreams in a phenomenon usually referred to as “Drug
hand, medium dose acts primarily on post-synaptic neurons Dreams” (Johnson, 2001). This phenomenon is more com-
(Miyazaki et al., 2004). Other centrally acting agents, such mon in addictive users undergoing detoxification, in which
as methyldopa and ketanserin, were observed to induce they frequently report dreaming about the drug use and/or
sleep disturbances and reduce REM sleep (Novak & Shap- drug craving, frequently accompanied with lack of sleep. The
iro, 1997; Bazzari, 2017). upregulation of neurotransmitters receptors due to chronic
drug exposure may explain the acute urge of such events;
3.3. Lipid-lowering Drugs however, these side effects usually ease with time after
drugs discontinuation (Christo & Franey, 1999). Alcohol use
Statins have been investigated for their neuroprotective was observed to suppress REM sleep and increase REM la-
properties in neurodegenerative diseases (Van der Most et tency; however, acute withdrawal was observed to increase
al., 2009). The use of; lovastatin, pravastatin, simvastatin, REM sleep and lower overall sleep duration (Brower, 2001).
atorvastatin, and fluvastatin, has been associated with the In a rat model, cocaine effects on REM were observed to
development of peripheral neuropathy, insomnia, unusual differ according to the dose; REM decreased with cocaine
dreams, and sleep or concentration problems (Clark, 2003). injections of 5 – 10mg/kg, and increased at lower doses of
Nevertheless, limited data available to explain their exact 2.5 – 5mg/kg (Knapp et al., 2007). Opiates (e.g. morphine)
mode in precipitating these events. use is generally found to suppress both NREM and REM
sleep (de Andrés & Corpas, 1991). In acute heroin withdraw-
3.4. Anesthetics al, EEG data showed a decrease in SWS and REM sleep
The use of anesthetics has been linked to influence the (Howe et al., 1980). Methylenedioxymethamphetamine also
dreams of patients undergoing medical procedures. Ear- known as “MDMA” or “Ecstasy”, was observed to induce a
lier studies have reported that between 1 – 57% of patients permanent alteration in natural sleep, which is characterized
were able to recall dreaming during anesthesia (Samuels- by a shortening in NREM and overall sleep duration (Allen
son et al., 2008). In a postoperative report of patients un- et al., 1993). Lysergic acid diethylamide (LSD) was found to
derwent cholecystectomy and gastroplasty, anesthesia was increase REM sleep first and second periods by 30 – 240%
induced using thiopental, nitrous oxide (N2O), isoflurane, and shortening in the following periods with an overall pro-
and fentanyl. Sleep disruption and fragmentation was noted longation of REM sleep, when low doses were administered
during the first two nights after operations with absence of 1 hour before sleep (Passie et al., 2008). N,N-Dimethyltrypt-
SWS and REM; however, in the successive 2 – 4 nights REM amine (DMT) was found to inhibit REM and SWS in healthy
cycle reappeared in a greater amount than preoperative val- individuals (Barbanoj et al., 2008). However, is has been
ues with patients reports of having distressing dreams or suggested that LSD and DMT-induced psychedelic expe-
vivid nightmares (Knill et al., 1990). The incidence of dreams riences may share some neuropharmacological similarities
reported was also found to differ significantly regarding an- with lucid dreaming, as it is argued to be an induced state
esthetics used, as patients who received total intravenous of consciousness which enhance self-knowledge and psy-
anesthesia reported to have more dreams (Brandner et al., chological insight (Kraehenmann, 2017; Kraehenmann et
1997). In a 2015 randomized-controlled trial, evoked images al., 2017). Phencyclidine also known as “angel dust” was
and suggestions directly prior anesthesia were used in or- firstly proposed for anesthesia; however, it was abandoned
der to assess any influence on dreams content and recall. due to its unpleasant side effects (Siegel, 1978). Similar to
The study findings showed a significant increase in dream ketamine in its mode of action on NMDA receptors; vivid
frequency and dream recall in patients who had preopera- dreams, nightmares, frightening hallucinations, and delu-
tive suggestions applied before and during anesthesia in- sions, were reported with the use of phencyclidine (Miller,
duction; in addition, formation of dreams and dream recalls 1991).
were observed to be dependent on the anesthetic tech-
nique used. Moreover, the use of these psychological tech-

International Journal of Dream Research Volume 11, No. 2 (2018) 109


IJoDR A pharmacological view on lucid dream induction

3.6. Medicinal plants who received dexmedetomidine (Nirmala et al., 2016). Dex-
tromethorphan, an over-the-counter (OTC) cough suppres-
Hypericum perforatum (St. John’s wort) was found to in- sants, is sometimes abused by individuals for its over-dose
crease REM sleep latency with no effects on the other sleep adverse events referred to as “high-state”, which is de-
stages (Sharpley et al., 1998). Valeriana officinalis (Valerian) scribed as a separation from the surrounding environment
use was observed to reduce sleep latency (Leathwood & accompanied with euphoria (Hilmas, 2001). Vivid dreams
Chauffard, 1985). Valerian did not show any significant ef- and hallucinations are common side effects of dextrometho-
fect on sleep stages; however, elevated delta activity was rphan (Hilmas, 2001). Dextromethorphan mode of action as
observed during NREM (Shinomiya et al., 2005). Ginkgo a noncompetitive agonist of NMDA receptors may explain
biloba (Ginkgo) has been widely used for its cognitive and the occurrence of such events (Martinak et al., 2017).
memory enhancing abilities (Bazzari et al., 2018). Ginkgo
was found to promote sleep continuity, reduce number of
awakenings, enhancing SWS, and reduce REM sleep (Hem-
4. Supplements for lucid dreaming
meter et al., 2001). Panax ginseng is suggested to enhance A number of synthetic and natural products have been
sleep quality with minor effects on SWS and REM sleep speculated for their potential in inducing or enhancing lucid
(Rhee et al., 1990). Myristica fragrans (Nutmeg), commonly dreaming. Some of which have been previously mentioned
used to treat gastrointestinal conditions, such as diarrhea and suggested by Thomas Yuschak (2006) in his book “Ad-
and stomach spasm, is speculated to increase dream re- vanced Lucid Dreaming: The Power of Supplements”. The
call and dream vividness; however, these claims still lack author provided a number of pharmacokinetic/dynamic
proper supportive evidence (Nagano, 2008). Olfactory ex- properties of these agents in order to find a reasonable
posure of lavender oil was observed to promote sleep and link between them and the suggested hypotheses of lucid
increase SWS; however, effects on REM sleep varied be- dreaming. Other supplements may only have poor or anec-
tween genders; decreased in women and increased in men, dotal evidence. However, using the term “supplements” and
for unexplained reasons (Goel et al., 2005). Jasminum offici- their OTC availability are perhaps the major reasons behind
nale flowers were observed to have a CNS suppressive and the huge popularity of these products among the commu-
sleep promoting activity (Elisha et al., 1988). nity, as they frequently provide the sense of using safe op-
tions with no potential harm. Agents may contribute to lucid
3.7. Hormonal supplementation dreaming induction in various ways; for instance, reducing
sleep latency (i.e. sedative activity), altering neurotransmit-
Hormonal supplementation was found to influence individu- ters, or via other pathways. This section will provide an
als sleep. Decreased overall sleep quality and REM sleep overview of the available clinical findings on supplements
are usually observed in old men with low levels of circulat- suggested to induce/enhance lucid dreaming, in order to
ing testosterone (Andersen & Tufik, 2008). However, testos- evaluate their actual potential.
terone supplementation was also observed to impair sleep,
increase number of awakenings, and nightmares, in male
and female subjects, which suggests that both low and high
4.1. Huperzine A
levels of testosterone may influence sleep (Andersen et al., Huperzine A is a potent AChEIs, which is naturally derived
2011). Low doses of estradiol administered in women with from Huperzia serrata, and used in the management of Al-
hot flashes were observed to enhance sleep quality and zheimer’s disease. In animal studies, huperzine A is found
ameliorate insomnia (Ensrud et al., 2015). Estrogens ad- to reduce neuronal inflammation, oxidative stress, and en-
ministration in hypogonadal women was associated with hance behavioral recovery after repetitive traumatic brain
increased REM sleep and lower sleep latency (Schiff et al., injury in mice (Mei et al., 2017). Furthermore, huperzine A
1979). Progesterone was also found to increase sleep qual- is also observed to have an anticonvulsant activity, antide-
ity and duration, which is suggested to be via interacting pressant-like properties, enhanced recent verbal memory,
with GABA; nonetheless, no significant difference was ob- and showed to be effective in ameliorating cognitive impair-
served in REM sleep (Caufriez & Copinschi, 2016). ments associated with neurodegenerative diseases (Fer-
reira et al., 2016; Du et al., 2017; Solomon et al., 2016). In
3.8. Miscellaneous drugs clinical trials, intense dreams were reported with the use of
huperzine A dose of 400 µg/b.i.d (Little et al., 2008). Similar
First generation antihistamines use is commonly associated to galantamine in its main mode of action as AChEIs ex-
with increased nightmares (Pagel, 2010). However, classi- plains the rationale behind its potential in lucid dreaming
cal antihistamines (e.g. chlorpheniramine, a histamine H1- induction (LaBerge, 2004). However, both agents are ob-
blocker) were observed to increase REM sleep latency and served to differ in their activity on NMDA receptors. Galan-
reduce REM duration (Ozdemir et al., 2014). Nightmares as tamine was observed to potentiate NMDA receptors activ-
a side effect were reported with the use of some antimi- ity, which is suggested to partially contribute to cognitive
crobial agents, such as amantadine, ciprofloxacin, eryth- and learning enhancement in Alzheimer’s disease patients
romycin, fleroxacin, ganciclovir, and gusperimus (Pagel & (Moriguchi et al., 2004). On the other hand, huperzine A was
Helfter, 2003). Dexmedetomidine, a potent α2-adrenergic found to antagonize NMDA receptors and provide protec-
agonist used to induce mild/moderate sedation in critically- tion against NMDA-induced seizures (Coleman et al., 2008).
ill patients, was observed to promote NREM sleep, enhance Unfortunately, even huperzine A is a powerful candidate for
overall sleep quality, and lessen sleep fragmentation with lucid dreaming induction/enhancement, yet it still lacks a
no significant difference in SWS or REM sleep (Alexopou- strong conclusive scientific evidence to support its use in
lou et al., 2014). A study has investigated dexmedetomi- lucid dreaming induction. However, a comparative study of
dine and dreaming during sedation, the study findings have huperzine A and galantamine would be interesting, not only
shown a significant reduction in dreams among individuals to test huperzine A actual potential, but also to shed some

110 International Journal of Dream Research Volume 11, No. 2 (2018)


A pharmacological view on lucid dream induction IJoDR

light on NMDA receptors role in lucid dreaming. activity, galantamine was also observed to enhance α7-
receptors activity (Texidó et al., 2005). In clinical trials, var-
4.2. Alpha-GPC enicline sleep-related side effects, include; insomnia, sleep
fragmentation, NREM parasomnias, REM sleep aggression
Alpha-glycerylphosphorylcholine (α-GPC) is a prodrug (similar to RBD), and vivid/lucid dreams (Savage et al., 2015;
of choline, an acetylcholine precursor, and considered an Patterson et al., 2017). Theoretically, such findings may cor-
essential nutrient to boost acetylcholine levels. In epilep- relate to varenicline potential in lucid dreaming; nonethe-
sy, α-GPC use was observed to reduce disruptions of the less, it is still a hypothetical assumption that requires further
blood-brain barrier (BBB), lower neuronal death, promote clinical assessment.
neurogenesis, and enhance cognitive functions (Lee et al.,
2017). In theory, elevated acetylcholine activity is linked to
4.4. 5-hydroxytryptophan
lucid dreaming enhancement, such as the case of AChEIs;
thus, α-GPC has been suggested in a number of anecdot- 5-hydroxytryptophan (5-HTP) is an intermediate metabolite
al findings as a proposed supplement for lucid dreaming. in the biosynthesis of the neurotransmitter serotonin from
However, α-GPC was tested in a double-blind placebo-con- its amino acid precursor L-tryptophan (Kato et al., 1974).
trolled trial of 33 individuals with a little to advanced experi- 5-HTP is found to be clinically-effective in increasing levels
ence in lucid dreaming. The study results showed neither a of serotonin in the CNS and suggested to have a number of
significant difference in lucid dreaming incidence nor altera- therapeutic applications, such as depression, chronic head-
tions in dream content (Kern et al., 2016). These findings ache, and insomnia (Birdsall, 1998). Furthermore, the use of
may limit the actual potential of α-GPC as a supplement for GABA/5-HTP mixture, in a caffeine-treated fruit flies model,
lucid dreaming induction. was found to be effective in promoting sleep, which is ob-
served through its ability in modulating; sleep episodes,
4.3. Nicotine night-time activity, and sleep duration (Hong et al., 2016).
In 2012, a dietary supplement product, containing; 5-HTP,
Following the same principle of increasing the cholinergic Calea ternifolia, Vinpocetine, melatonin (as a secondary in-
activity for lucid dreaming induction/enhancement, nicotine gredient), and a number of vitamins, received a patent as
serves as an exogenous agonist for acetylcholine nicotinic a method to promote sleep and lucid dreaming (Luciano,
receptors (Gotti et al., 2006). In 2006, a study was conduct- 2012). While the product description presents a summary
ed on 15 smokers to test the effects of wearing 24-hour of the role of its ingredients, limited clinical data provided to
transdermal nicotine patches on sleep and dreams. The support claims about 5-HTP role in lucid dreaming induc-
study results showed an increase in sleep fragmentation, tion. Therefore, 5-HTP is still in need of a robust clinical trial
no effects on NREM sleep, reduced REM duration, and an to support its use in lucid dreaming.
increase in REM-dreams quality with a higher frequency of “REM sleep rebound” is a common term among lucid
recallable visual imagery reports (Page et al., 2006). In addi- dreamers, which is defined as an occurrence of prolonged
tion, previous reports have shown that sleep disturbances, REM sleep as a result of previous periods of REM suppres-
such as increased sleep latency, daytime sleepiness, sleep sion (Yuschak, 2006). Elevated levels of serotonin are linked
fragmentation, are linked to nicotine stimulation in both male to REM suppression, which can also be observed with the
and female smokers (Wetter & Young, 1994). The use of nic- use of serotonergic antidepressants; SSRIs and SNRIs
otine is controversial due to potential risks associated with (Ursin, 2002; Winkelman & James, 2004). Moreover, acute
its use, such as nicotine dependence observed in smokers. withdrawal of serotonergic antidepressants was observed
Therefore, risks of using tobacco smoking alternatives in to increase vivid dreams incidence (Keks et al., 2016). Hy-
non-smokers are still unclear yet. However, reduced confu- pothetically, these findings seem consistent with the REM
sion, increased reaction time (assessed via vigilance task sleep rebound effect; therefore, the use of 5-HTP was pro-
performance), and improved total mood, were observed posed as an indirect method to increase REM sleep dura-
with the use of nicotine transdermal patches 7mg/24hr, with tion, and in turn, increase the chance of having a lucid dream
a total time of 6 hours in healthy non-smokers participants (Yuschak, 2006). Nonetheless, REM sleep deprivation to
(McComas et al., 2015). Nonetheless, further investigations achieve REM sleep rebound is a questionable approach. In
should be undertaken in order to obtain more conclusive a number of human and animal studies, REM sleep depriva-
findings regarding nicotine use for lucid dreaming. tion has resulted in a number of neurobiological and behav-
Varenicline, a drug used for smoking cessation, can pos- ioral alterations, such as increased hyperactivity, aggres-
sibly be an alternative to nicotine. The nicotinic acetylcho- siveness, cellular stress, glial dysfunction, impaired spatial
line receptor α4ß2 subtype is abundant in the human brain, memory, and a reduction in BrdU+ hippocampal cells (Soto-
and it is suggested to be responsible for mediating nico- Rodriguez et al., 2016). REM sleep suppression was also
tine dependence (Benowitz, 2009). While nicotine exhibits found to enhance emotional reactivity (Rosales-Lagarde et
full agonism on α4ß2, varenicline exerts partial agonistic and al., 2012). Regardless of REM deprivation consequences,
competitive inhibition activity with nicotine on α4ß2 recep- there is an apparent inconsistency in 5-HTP effects on sleep
tors. Varenicline activity on α4ß2 receptors is found to reduce architecture observed in clinical data. In early investigations,
mesolimbic dopamine release, which results in lowering 5-HTP was observed to slightly reduce NREM, increase
craving symptoms (Elrashidi & Ebbert, 2014). Despite va- REM sleep duration, reduce REM latency, increase dream-
renicline partial agonistic activity on heteromeric neuronal ing frequency and dream recall in human subjects (Mandell
nicotinic receptors, it is found to have a potent full-agonistic et al. 1965; Oswald et al. 1966; Wyatt et al., 1971). However,
activity on α7-nicotinic receptor (Mihalak et al., 2006). De- when 5-HTP was tested in cats, there was a complete ab-
creased activity of α7-nicotinic receptors in the human brain sence of REM for 6 hours after 5-HTP administration (Ursin,
was found in a number of neurological and psychiatric 1976). While results in animal experiments may not always
disorders. In addition to its acetylcholinesterase inhibition correlate to humans, 5-HTP was further assessed by Na-

International Journal of Dream Research Volume 11, No. 2 (2018) 111


IJoDR A pharmacological view on lucid dream induction

kazawa et al. (1980), in which no effects were observed on sleep latency, reduce sleep fragmentation, and increase
sleep stages, except for stage 1, with the use of 200mg/ overall sleep quality (Peuhkuri et al., 2012).
p.o. 5-HTP. However, when individuals were deprived from
sleep for one night, followed by administration of 5-HTP on 4.6. Melatonin
the first recovery night, 5-HTP inhibited SWS rebound for
the next two consecutive nights. Moreover, an increase in N-acetyl-5-methoxy tryptamine, also known as “melato-
REM sleep was observed on the second night after both; nin”, is a hormone produced by the penial gland, which
5-HTP and sleep deprivation, recovery phase (Nakazawa regulates sleep/wake cycle, circadian rhythm, and has an
et al., 1980). Para-chlorophenylalanine (pCPA), a selective immune-stimulatory and cytoprotective activity (Hardeland
irreversible inhibitor of tryptophan hydroxylase that results et al., 2006). In sleep and circadian rhythm regulation, mela-
in a significant depletion of serotonin in the CNS, is com- tonin exerts its action via binding to melatonin; MT1 and MT2
monly used to induce insomnia in animals characterized receptors, found in the suprachiasmatic nucleus (SCN) of
by a marked inhibition of SWS and REM (Borbély et al., the hippocampus (Srinivasan et al., 2009). MT1 action was
1981). In numerous studies, 5-HTP administration following observed to inhibit neuronal firing in SCN, while MT2 was
pCPA treatment was successful in restoring normal sleep found to inhibit phase shift circadian firing rhythms in the
patterns, which gave a rise to the “serotonin sleep theory” SCN (Dubocovich, 2007). In both human and animal mod-
(Smith & Kennedy, 2003). An introduced possible explana- els, melatonin use was found to promote sleep and improve
tion is that the depletion of serotonin levels due to sleep overall sleep quality (Zhdanova et al., 1997; Garfinkel et al.,
deprivation increases the metabolism of 5-HTP and bypass 1995). Melatonin use is also found to affect sleep architec-
into nerve terminals; in contrast, during normal sleep sero- ture. In early investigations, the use of 5mg melatonin prior
tonin levels in the CNS are self-sufficient, which lessen the bedtime has increased REM latency, with no alterations in
uptake of exogenous 5-HTP (Nakazawa et al., 1980). None- overall REM duration and percentage (James et al., 1987).
theless, due to the fact that there is no hypnogenic target In addition, using 250mg/q.i.d melatonin for 6 consecutive
identified in order to explain 5-HTP activity in restoring sleep days has shown a significant increase in stage 2 sleep, and
following pCAP, the “serotonin sleep theory” was dismissed a lesser duration of stage 4 sleep (Anton-Tay, 1974). In a
(Jouvet, 1999). Recent investigations have provided some 2004 randomized placebo-controlled trial on individuals
insight about serotonin role in sleep, as serotonin was found with reduced REM sleep duration, the use of 3mg melatonin
to differentially modulate neurons; GABAergic and gluta- for 4 weeks significantly improved REM sleep percentage
matergic, of the ventrolateral preoptic nucleus, which is the and continuity (Kunz et al., 2004). These findings have led to
main structure responsible for triggering NREM (Sangare et the suggestion that elevated melatonin levels enhance sleep
al., 2016). Up to date, the role of serotonin in sleep/wake stages (i.e. stage 2 and REM), in which dreams commonly
regulation is still not fully understood and considered a point occur (Kahan et al., 2000). In patients with RBD, melatonin
of debate. Combined all together, there is neither a clear was observed to suppress REM motor activity, yet it did not
view about the exact 5-HTP molecular mode of action nor a completely eliminate it; in addition, high doses of melato-
conclusive evidence to support the anecdotal claims about nin were associated with an increase in nightmares (Howell
its potential in lucid dreaming. et al., 2011). In a placebo-controlled study on 22 college
students to assess melatonin effects on dreams, the study
results have shown an increase in dream recall, vividness,
4.5. Magnesium L-threonate
and bizarreness. In addition, there was no effect observed
Magnesium L-threonate (MgT) is a novel compound with in REM sleep duration; nonetheless, EEG data showed a
a high ability to cross the BBB. Numerous studies have marked increase in REM density (i.e. high amplitude waves
investigated its beneficial role in memory enhancement, during REM) (Kahan et al., 2000). While the available data
neurological conditions with related memory-loss, and syn- may support the anecdotal claims that melatonin promotes
aptoprotective properties (Li et al., 2014; Lou et al., 2017). REM sleep, and in turn, increase dream frequency and viv-
Generally, magnesium (Mg) has shown efficacy and toler- idness, further investigations should be undertaken in order
ability in the management of sleep disturbances and anxiety to confirm such claims.
in patients receiving chemotherapy, and it is suggested to Melatonin interaction mechanisms with neurotransmitters
be via its relative muscle relaxing activity (Carson, 2017). is still not fully understood. However, the increased inter-
Furthermore, Mg is also found to be vital in modulating the est in melatonin activity in regulating sleep and circadian
brain biochemistry, and low dietary intake of Mg has been rhythm has led to the development of a number of medi-
attached to a number of medical conditions; for instance, cal agents that are more selective and have a higher affinity
depression, apathy, agitation, delirium, anxiety, and confu- to melatonin receptors. For instance, tasimelteon, a selec-
sion (Serefko et al., 2016). Moreover, extracellular Mg+2 ions tive melatonin MT1 and MT2 receptors agonist, is the first
in the brain were observed to modulate the activity of the drug to be approved by the food and drug administration
NMDA receptor, as reduced Mg+2 levels greatly potentiate (FDA) in the United States for treating non-24-hour sleep-
NMDA receptor activity and vice versa (Nowak et al., 1984). wake disorder (Lavedan et al., 2015). Tasimelteon possess a
Reduced NMDA receptor excitatory activity was observed unique pharmacological profile, as it is not only a full agonist
to provide neuroprotection, sedation, induce reversible con- of melatonin receptors (with more affinity for MT2) but also
sciousness loss and loss of righting reflux (Franks, 2008). did not show any affinity to other pharmacologically related
Mg deficiency was also observed to affect sleep architec- receptors, such as GABAergic, adrenergic, dopaminergic,
ture, which is characterized by an increase in light SWS and nicotinic, histamine, serotonin, and many other receptors
a decrease in REM sleep duration, in patients with restless (Lavedan et al., 2015). Tasimelteon was observed to reduce
leg syndrome caused by Mg deficiency (Popoviciu et al., sleep latency, increase sleep efficiency, and reduce REM
1993). At last, Mg major role in enhancing lucid dreaming sleep latency (i.e. REM occurred more rapidly) in individu-
may only be related to its ability to promote sleep, lower als receiving 20, 50, and 100mg tasimelteon compared to

112 International Journal of Dream Research Volume 11, No. 2 (2018)


A pharmacological view on lucid dream induction IJoDR

placebo or with a lower dose of 10mg (Bonacci et al., 2015). products, it may not exceed the “placebo effect”.
Furthermore, nightmares and abnormal dreams were also
reported with the use of tasimelteon (Leger et al., 2015).
These findings may highlight the potential of melatonin re- 5. Factors hindering drug application in lucid
ceptors selective agonists as target-specific candidates for dreaming
lucid dreaming. However, further investigations are needed
in order to confirm this assumption and to explore their ex- While there are many potential candidates for lucid dream-
act underlying mechanisms. ing, yet, for now, limited evidence available to support their
use. This might be due to a number of factors that hinder
4.7. Herbal supplements drugs’ use for lucid dreaming, which can either be; individu-
als, drugs, or research related factors. These factors are
Herbal products are widely used in traditional Chinese and summarized in the sections below:
Ayurvedic medicine for the management of various medical
conditions. A number of which are identified to have noo- 5.1. Individuals related factors
tropic properties and CNS stimulating/suppressing activ-
ity that are commonly observed among users. While some Individuals’ variations are critical when it comes to the pro-
have been observed to affect sleep and sleep architecture cess of drug discovery and development. Even with a large
in scientific research (see section 3.6.), there was no conclu- sample size, there is always a need for pharmacovigilance
sive evidence to validate their use for dream recall or lucid and post-marketing surveillance for further assessment.
dreaming. However, isolation and standardization of chemi- Similarly, a number of studies have identified a number of in-
cally active components with a clear image of their mode of dividuals related factors that may influence lucid dreaming.
action, such as huperzine A derived from Huperzia serrata, For instance, in a study on students from different nationali-
may aid in introducing novel products for lucid dreaming. ties in a Japanese university, there was a significant differ-
Up to date, the use of a number of herbal products with rela- ence in lucid dreaming incidence between individuals from
tive sedative and sleep-promoting activity (e.g. St. John’s different countries; in addition to, a difference between indi-
wort, valerian, lavender) may explain the anecdotal claims viduals unaware about the concept of lucid dreaming com-
regarding their use for enhancing lucid dreaming. pared to individuals who claim they had experienced a lucid
dream (Erlacher et al., 2008). Moreover, individuals’ person-
4.8. B vitamins ality may also correlate to the frequency of lucid dreaming.
Several studies have indicated that a number of the Big Five
A number of B vitamins; B3, B6, B12, are observed to influ- personality factors, such as neuroticism, openness, and
ence sleep and dreams via multiple suggested roles. B3 agreeableness, had a small, yet, a significant correlation to
administration was observed to increase REM sleep and lucid dreaming frequency (Schredl & Erlacher, 2004; Hess
improve sleep quality in patients with moderate/severe in- et al., 2017). Furthermore, age and gender are also found
somnia (Robinson et al., 1977). B3 is biosynthesized from to influence lucid dreaming, as lucid dreaming is found to
tryptophan via kynurenine pathway; thus, it is speculated be negatively correlated to age, and women had a higher
that B3 supplementation would increase the amount of tryp- lucid dreams recall (Schredl & Erlacher, 2011). In addition
tophan available for melatonin and serotonin synthesis (i.e. to the previously mentioned involuntary factors, a number
negative feedback on kynurenine pathway) (Peuhkuri et al., of individuals’ daily-life habits are also found to alter sleep
2012). In a double-blind placebo-controlled study on 12 and dreams in general. For example, tobacco smoking and
college students, B6 is found to increase dream vividness alcohol consumption are both observed to affect sleep and
and bizarreness, which is speculated to be via increased dream (see sections 3.5. & 4.3.). Diet is also found to influ-
cortical arousal during REM (Ebben et al., 2002). Generally, ence sleep; for example, a diet with high-carbohydrate low-
B6 mainly contribute to the synthesis of 5-HTP, and further fat content was observed to reduce SWS and increase REM
serotonin, from tryptophan, which may explain their influ- sleep; in contrast, a fat-rich diet with low-carbohydrate per-
ence on dreams. At last, B12 is also found to participate in centage had the opposite effects (Peuhkuri et al., 2012). At
melatonin synthesis/secretion and regulation of sleep-wake last, individuals’ health status (i.e. healthy vs. non-healthy)
cycle (Peuhkuri et al., 2012). Hypothetically, the use of B vi- is a very important factor to be considered, which may also
tamins for lucid dreaming seems promising, yet still require influence other major factors (i.e. Drugs & Research related
further investigations to test their efficacy. factors). As the risk of drugs’ toxicity rises in patients with
liver and/or renal impairments. In addition, patients with
4.9. Homeopathy chronic health conditions, who are maintained on certain
drugs, may elevate the risk of drug-drug interactions, and,
Homeopathy is generally referred to as pseudoscience (i.e.
most importantly, patients with CNS disorders who already
misrepresented as true science), which has many prac-
have a disturbed neurotransmitters’ behavior.
tice flaws and lacks any scientific plausibility and evidence
(Smith, 2012). In 2015, the Australian National Health and
Medical Research Council (NHMRC) published an informa- 5.2. Drugs related factors
tion paper regarding the effectiveness of homeopathy in Multiple concerns surround the use of medical agents that
health conditions. The paper concluded that homeopathy may influence their efficacy, especially that, there is no clear
is not effective in the management of any medical condi- image about their actual potential for lucid dreaming with a
tion and lacks any high-quality placebo-controlled studies lot of speculations. In a general context, most of the medi-
to support its use (NHMRC, 2015). Therefore, homeopathy cal agents have a well-established evidence regarding their
is not a reliable approach for lucid dreaming and, if any, major mechanism of action, and in some cases, EEG re-
positive outcomes observed with the use of homeopathic cordings have helped in identifying their effects on sleep

International Journal of Dream Research Volume 11, No. 2 (2018) 113


IJoDR A pharmacological view on lucid dream induction

architecture. Nonetheless, the complex nature of the CNS neurotransmitters’ behavior, especially during REM sleep,
limits the full understanding of all molecular interactions of as their activity is more profound compared to NREM. How-
the drugs and how they correlate to the physiology of sleep ever, further studies should be undertaken to focus more
and dreams. Moreover, the drug dose is a vital factor that in- on neural mechanisms during NREM and sleep as a whole,
fluences its behavior, such as the case of clonidine (see sec- which may aid in explaining the increase in dreams frequen-
tion 3.2.) or medical agents with narrow therapeutic index; cy with the use of medical agents that are identified to have
therefore, in practice, using different drug doses is well-rec- a REM suppressive activity, such as propofol (Kim et al.,
ommended. Furthermore, drug-delivery into the CNS may 2011; Kondili et al., 2012).
not be a major issue, since drug candidates are relatively In the field of lucid dreaming, it is observed that there
lipophilic and have the ability to cross BBB. However, it can has been a greater focus on cognitive induction techniques
be an issue if the aim is to target a specific sleep stage (e.g. compared to other methods, which is, perhaps, due to over
REM). Pharmaceutical formulations, such as modified or exaggeration by lucid dreamers about the efficacy of such
sustained-release formulation, may be a good and simple techniques. However, many of which failed to show any sig-
option compared to other techniques; for instance, waking- nificance or conclusive evidence in inducing lucid dreams
up individuals to administer the drug or using intravenous (Stumbrys et al., 2012; Dyck et al., 2017). While the use
infusions/injections, which may interfere with individuals’ of supplements and a number of medical agents for lucid
sleep and reduce their compliance. Moreover, selective dreaming has recently started to gain some momentum, yet
targeting of a particular brain region, such as REM sleep- it seems that it is following the same pattern as the previ-
active neurons in the subcoeruleus nucleus (Fraigne et al., ously suggested cognitive techniques. With limited evidence
2015), is not a practical approach in humans. Nonetheless, to support their use, the scientific research favorably tends
further investigations using animal models can, to some ex- to test these agents based on poor or anecdotal claims sur-
tent, enhance the understanding of the drugs’ local effects rounding their use. For instance, even with boosting acetyl-
on various brain regions and neuronal circuits. Considering choline activity hypothesis to support α-GPC use for lucid
a combination of multiple agents, with minimal drug-drug dreaming, it did not show any promising outcomes (Kern
interactions, in order to target multiple neurotransmitters et al., 2016). Testing every single agent claimed to increase
pathways or a combination with sleep enhancers, may aid lucid dreaming, especially with no theory to back its use,
in promoting lucid dreaming. For example, the lucid dream- is a time and effort consuming approach. It can be argued
ing patented supplement product, which contains multiple that it is better than testing random agents; nonetheless,
natural and synthetic agents that are intended to play mul- it may distract researchers from investigating other major
tiple roles in lucid dreaming induction (see section 4.4.) (Lu- players in sleep that may participate in lucid dreaming, such
ciano, 2012). However, further studies are required in order as GABA or glutamate (see section 2.). Since the public has
to introduce medical agents for lucid dreaming with more a restricted access to a vast number of drugs and only lim-
target-specificity, clear mode of action, and a standardized ited to medical purposes, no claims can be observed about
dose. their potential for lucid dreaming. Nevertheless, previous
clinical findings serve as a valuable source of data about
5.3. Research related factors the drugs’ effects on sleep and dreams nature, which may
correlate to lucid dreaming. For example, drugs observed to
These factors address the gaps in research between differ- increase dreams’ vividness may possibly be candidates to
ent fields; neurophysiology, neuropharmacology, and sleep enhance lucid dreaming, and since there is a relatively clear
research, in regards to lucid dreaming. Despite the huge view about their mode of action; hence, they may aid in un-
efforts invested in CNS research for decades, yet no con- derstanding neural mechanisms mediating lucid dreaming.
clusive findings on drug-induced lucid dreaming. Multiple
factors have participated in this, one of which is that drugs’
6. Conclusion
effects on sleep and dreams were not within the scope (i.e.
endpoints) or a major concern of the majority of previously With the continuous increase in understanding neuronal cir-
undertaken clinical research. While in some cases, when cuits and neurotransmitters’ behavior involved in sleep; in
medical agents are observed to affect sleep and/or dreams addition to, a wide range of medical agents full of possibili-
nature, they are usually being reported as side effects of the ties, there is no doubt that drug application will become a
drug use, such as increased nightmares or vivid dreams, major and more reliable method for lucid dreaming induc-
with no further assessment (see sections 3.1. – 3.8.). Even tion in the future. Identifying potential drug candidates for
in studies that have used EEG, to assess medical agents lucid dreaming puts us a one step closer towards achieving
reported to alter sleep architecture and dreams, did not successful outcomes. However, extensive investigations
address lucid dreaming in their investigations. Moreover, should be undertaken, not only in testing various medical
there is a debate in the field of dream research of whether agents but also to obtain more knowledge about the under-
dreams are REM sleep phenomenon or not, and was re- lying mechanisms and the validity of the current hypotheses
cently reviewed by Montangero (2018), in which the author surrounding lucid dreaming.
presented both sides of the argument supported by evi-
dence. In addition, Montangero has highlighted the conse- Disclaimer
quences of the scientific denial in hindering the progress
of dream research (Montangero, 2018). Some studies have Unless for medical/scientific purposes and under the super-
reported on lucid dreaming occurrence during NREM sleep; vision of medical experts, the author does not promote or
nonetheless, lucid dreaming is observed to be more com- support the use of any of the medical agents or supple-
mon during REM and its incidence in NREM is found to be ments mentioned earlier in this paper for lucid dreaming,
rare and hard to achieve (Stumbrys & Erlacher, 2012). Ear- dream recall, or any other indication. The main and only aim
lier investigations have provided a decent knowledge about of this paper is to investigate their potential in lucid dream-

114 International Journal of Dream Research Volume 11, No. 2 (2018)


A pharmacological view on lucid dream induction IJoDR

ing by exploring the related scientific evidence. The author Brandner, B., Blagrove, M., McCallum, G., & Bromley, L. M.
declares no conflict of interest or received any funding for (1997). Dreams, images and emotions associated with
this work. propofol anaesthesia. Anaesthesia, 52(8), 750-755.
Brooks, P. L., & Peever, J. H. (2012). Identification of the trans-
mitter and receptor mechanisms responsible for REM
References sleep paralysis. Journal of Neuroscience, 32(29), 9785-
Adetunji, B., Mathews, M., Williams, A., Budur, K., Mathews, 9795.
M., Mahmud, J., & Osinowo, T. (2005). Use of antipsy- Brower, K. J. (2001). Alcohol’s effects on sleep in alcoholics. Al-
chotics in the treatment of post-traumatic stress disor- cohol research & health: the journal of the National Insti-
der. Psychiatry (Edgmont), 2(4), 43-47. tute on Alcohol Abuse and Alcoholism, 25(2), 110-125.
Alexopoulou, C., Kondili, E., Diamantaki, E., Psarologakis, C., Carson, C. (2017). Magnesium oxide supplements for the treat-
Kokkini, S., Bolaki, M., & Georgopoulos, D. (2014). Ef- ment of post-chemotherapy sleep disturbance. Journal
fects of Dexmedetomidine on Sleep Quality in Critically of Clinical Oncology, 35(Suppl 5 ), 175.
Ill PatientsA Pilot Study. Anesthesiology: The Journal of Caufriez, A., & Copinschi, G. (2016). U.S. Patent No. 9,504,697.
the American Society of Anesthesiologists, 121(4), 801- Washington, DC: U.S. Patent and Trademark Office.
807. Christo, G., & Franey, C. (1996). Addicts drug-related dreams:
Allen, R. P., McCann, U. D., & Ricaurte, G. A. (1993). Persistent their frequency and relationship to six-month outcomes.
effects of (±) 3, 4-methylenedioxymethamphetamine Substance Use & Misuse, 31(1), 1-15.
(MDMA,“ecstasy”) on human sleep. Sleep, 16(6), 560- Chrysant, S. G., Chrysant, G. S., & Dimas, B. (2008). Current
564. and Future Status of Beta‐blockers in the Treatment of
Andersen, M. L., Alvarenga, T. F., Mazaro-Costa, R., Hachul, H. Hypertension. Clinical cardiology, 31(6), 249-252.
C., & Tufik, S. (2011). The association of testosterone, Clark, L. T. (2003). Treating dyslipidemia with statins: the risk-
sleep, and sexual function in men and women. Brain benefit profile. American heart journal, 145(3), 387-396.
research, 1416, 80-104. Coleman, B. R., Ratcliffe, R. H., Oguntayo, S. A., Shi, X.,
Andersen, M. L., Margis, R., Frey, B. N., Giglio, L. M., Kapczin- Doctor, B. P., Gordon, R. K., & Nambiar, M. P. (2008).
ski, F., & Tufik, S. (2009). Electrophysiological correlates [+]-Huperzine A treatment protects against N-methyl-
of sleep disturbance induced by acute and chronic ad- D-aspartate-induced seizure/status epilepticus in rats.
ministration of D-amphetamine. Brain research, 1249, Chemico-biological interactions, 175(1-3), 387-395.
162-172. Cudeiro, J., Rivadulla, C., & Grieve, K. (2000). A possible role
Andersen, M. L., & Tufik, S. (2008). The effects of testoster- for nitric oxide at the sleep/wake interface. Sleep, 32(6),
one on sleep and sleep-disordered breathing in men: 829-835.
its bidirectional interaction with erectile function. Sleep Dash, M. B., Douglas, C. L., Vyazovskiy, V. V., Cirelli, C., & Tono-
Medicine Reviews, 12(5), 365-379. ni, G. (2009). Long-term homeostasis of extracellular
Anton-Tay, F. (1974). Melatonin: effects on brain function. Ad- glutamate in the rat cerebral cortex across sleep and
vances in biochemical psychopharmacology, 11, 315- waking states. Journal of Neuroscience, 29(3), 620-629.
324. Dauvilliers, Y., Siegel, J.M., Lopez, R., Torontali, Z.A., & Peever,
Barbanoj, M. J., Riba, J., Clos, S., Giménez, S., Grasa, E., & J.H. (2014) Cataplexy – clinical aspects, pathophysiol-
Romero, S. (2008). Daytime Ayahuasca administration ogy and management strategy. Nature Reviews Neurol-
modulates REM and slow-wave sleep in healthy volun- ogy, 10(7), 386–395.
teers. Psychopharmacology, 196(2), 315-326. de Andrés, I., & Corpas, I. (1991). Morphine effects in brain-
Bazil, C. W. (2003). Effects of antiepileptic drugs on sleep struc- stem-transected cats: II. Behavior and sleep of the de-
ture. CNS drugs, 17(10), 719-728. cerebrate cat. Behavioural brain research, 44(1), 21-26.
Bazzari, A. H., & Bazzari, F. H. (2018). Medicinal plants for Al- De Gennaro, L., Lanteri, O., Piras, F., Scarpelli, S., Assogna,
zheimer’s disease: An updated review. Journal of Me- F., Ferrara, M., Caltagirone, C. & Spalletta, G. (2016).
dicinal Plants Studies, 6(2), 81-85. Dopaminergic system and dream recall: an MRI study
Bazzari, F. H., & Bazzari, A. H. (2018). Drug-Induced Delirium: A in Parkinson’s disease patients. Human brain mapping,
Mini Review. BMH Medical Journal, 5(2), 51-56. 37(3), 1136-1147.
Bazzari, F. H. (2017). Centrally acting antihypertensive agent Du, Y., Liang, H., Zhang, L., & Fu, F. (2017). Administration of
Ketanserin potential role in delirium management: A re- Huperzine A exerts antidepressant-like activity in a rat
view. Open Science Journal, 2(3). model of post-stroke depression. Pharmacology Bio-
Benowitz, N. L. (2009). Pharmacology of nicotine: addiction, chemistry and Behavior, 158, 32-38.
smoking-induced disease, and therapeutics. Annual re- Dubocovich, M. L. (2007). Melatonin receptors: role on sleep
view of pharmacology and toxicology, 49, 57-71. and circadian rhythm regulation. Sleep medicine, 8, 34-
Birdsall, T. (1998). 5-Hydroxytryptophan: a clinically-effective 42.
serotonin precursor. Alternative medicine review: a jour- Dyck, S., Schredl, M., & Kühnel, A. (2017). Lucid dream induc-
nal of clinical therapeutic, 3(4), 271-280. tion using three different cognitive methods. Interna-
Blagrove, M., Morgan, C. J., Curran, H. V., Bromley, L., & Brand- tional Journal of Dream Research, 10(2), 151-156.
ner, B. (2009). The incidence of unpleasant dreams af- Ebben, M., Lequerica, A., & Spielman, A. (2002). Effects of
ter sub-anaesthetic ketamine. Psychopharmacology, pyridoxine on dreaming: a preliminary study. Perceptual
203(1), 109-120. and motor skills, 94(1), 135-140.
Bonacci, J. M., Venci, J. V., & Gandhi, M. A. (2015). Tasimelteon Eer, A. S., Padmanabhan, U., & Leslie, K. (2009). Propofol dose
(Hetlioz™) A New Melatonin Receptor Agonist for the and incidence of dreaming during sedation. European
Treatment of Non-24-Hour Sleep-Wake Disorder. Jour- Journal of Anaesthesiology, 26(10), 833-836.
nal of pharmacy practice, 28(5), 473-478. Elisha, E. E., Al-Maliki, S. J., & Ibrahem, D. K. (1988). Effects
Borbély, A. A., Neuhaus, H. U., & Tobler, I. (1981). Effect of p- ofJasminum officinale Flowers on the Central Nervous
chlorophenylalanine and tryptophan on sleep, EEG and System of the Mouse. International Journal of Crude
motor activity in the rat. Behavioural brain research, Drug Research, 26(4), 221-227.
2(1), 1-22.

International Journal of Dream Research Volume 11, No. 2 (2018) 115


IJoDR A pharmacological view on lucid dream induction

Elrashidi, M. Y., & Ebbert, J. O. (2014). Emerging drugs for the Hardeland, R., Pandi-Perumal, S. R., & Cardinali, D. P. (2006).
treatment of tobacco dependence: 2014 update. Expert Melatonin. The international journal of biochemistry &
opinion on emerging drugs, 19(2), 243-260. cell biology, 38(3), 313-316.
Ensrud, K. E., Guthrie, K. A., Hohensee, C., Caan, B., Carpen- Hasegawa, E., Maejima, T., Yoshida, T., Masseck, O. A., Her-
ter, J. S., Freeman, E. W., LaCroix, A.Z., Landis, C.A., litze, S., Yoshioka, M., Sakurai, T. & Mieda, M. (2017).
Manson, J., Newton, K.M. & Otte, J. (2015). Effects of Serotonin neurons in the dorsal raphe mediate the anti-
estradiol and venlafaxine on insomnia symptoms and cataplectic action of orexin neurons by reducing amyg-
sleep quality in women with hot flashes. Sleep, 38(1), dala activity. Proceedings of the National Academy of
97-108. Sciences, 114(17), E3526-E3535.
Erlacher, D., Schredl, M., Watanabe, T., Yamana, J., & Gantzert, Hemmeter, U., Annen, B., Bischof, R., Brüderlin, U., Hatzinger,
F. (2008). The incidence of lucid dreaming within a Japa- M., Rose, U., & Holsboer-Trachsler, E. (2001). Polysom-
nese university student sample. International Journal of nographic effects of adjuvant ginkgo biloba therapy in
Dream Research, 1(2), 39-43. patients with major depression medicated with trimipra-
Ferreira, A., Rodrigues, M., Fortuna, A., Falcão, A., & Alves, G. mine. Pharmacopsychiatry, 34(02), 50-59.
(2016). Huperzine A from Huperzia serrata: a review of Hess, G., Schredl, M., & Goritz, A. (2017). Lucid Dreaming Fre-
its sources, chemistry, pharmacology and toxicology. quency and the Big Five Personality Factors. Imagina-
Phytochemistry reviews, 15(1), 51-85. tion, Cognition and Personality, 36(3), 240-253.
Fink, K. B., & Göthert, M. (2007). 5-HT receptor regulation of Hilmas, E. (2001). Adolescent dextromethorphan abuse. Tox-
neurotransmitter release. Pharmacological reviews, alert. Maryland Poison Center: University of Maryland
59(4), 360-417. School of Pharmacy, 18, 1-3.
Flint, R. R., Chang, T., Lydic, R., & Baghdoyan, H. A. (2010). Hong, K., Park, Y., & Suh, H. (2016). Sleep-promoting effects of
GABAA receptors in the pontine reticular formation of a GABA/5-HTP mixture: Behavioral changes and neuro-
C57BL/6J mouse modulate neurochemical, electro- modulation in an invertebrate model. Life sciences, 150,
graphic, and behavioral phenotypes of wakefulness. 42-49.
Journal of Neuroscience, 30(37), 12301-12309. Howe, R. C., Hegge, F. W., & Phillips, J. L. (1980). Acute heroin
Foral, P., Knezevich, J., Dewan, N., & Malesker, M. (2011). Med- abstinence in man: I. Changes in behavior and sleep.
ication-induced sleep disturbances. The Consultant Drug & Alcohol Dependence, 5(5), 341-356.
Pharmacist®, 26(6), 414-425. Howell, M. J., Arneson, P. A., & Schenck, C. H. (2011). A novel
Fraigne, J. J., Torontali, Z. A., Snow, M. B., & Peever, J. H. therapy for REM sleep behavior disorder (RBD). Journal
(2015). REM sleep at its core–circuits, neurotransmit- of clinical sleep medicine, 7(6), 639-644A.
ters, and pathophysiology. Frontiers in neurology, 6, Im, J. H., Ha, J. H., Cho, I. S., & Lee, M. C. (2003). Ropinirole as
123. an adjunct to levodopa in the treatment of Parkinson’s
Franks, N. P. (2008). General anaesthesia: from molecular tar- disease. Journal of neurology, 250(1), 90-96.
gets to neuronal pathways of sleep and arousal. Nature Isaac, S. O., & Berridge, C. W. (2003). Wake-promoting actions
Reviews Neuroscience, 9(5), 370-386. of dopamine D1 and D2 receptor stimulation. Journal of
Garfinkel, D., Laudon, M., Nof, D., & Zisapel, N. (1995). Improve- Pharmacology and Experimental Therapeutics, 307(1),
ment of sleep quality in elderly people by controlled- 386-394.
release melatonin. The Lancet, 346(8974), 541-544. James, S. P., Mendelson, W. B., Sack, D. A., Rosenthal, N. E.,
Gautier-Sauvigné, S., Colas, D., Parmantier, P., Clement, P., & Wehr, T. A. (1987). The effect of melatonin on normal
Gharib, A., Sarda, N., & Cespuglio, R. (2005). Nitric ox- sleep. Neuropsychopharmacology, 1(1), 41-44.
ide and sleep. Sleep medicine reviews, 9(2), 101-113. Johnson, B. (2001). Drug dreams: a neuropsychoanalytic hy-
Glick, I., Murray, S., Vasudevan, P., Marder, S., & Hu, R. (2001). pothesis. Journal of the American Psychoanalytic As-
Treatment with atypical antipsychotics: new indications sociation, 49(1), 75-96.
and new populations. Journal of psychiatric research, Jouvet, M. (1999). Sleep and serotonin: an unfinished story.
35(3), 187-191. Neuropsychopharmacology, 21(S1), 24S.
Goel, N., Kim, H., & Lao, R. P. (2005). An olfactory stimulus Kahan, T., Hays, J., Hirashima, B., & Johnston, K. (2000). Ef-
modifies nighttime sleep in young men and women. fects of melatonin on dream bizarreness among male
Chronobiology International, 22(5), 889-904. and female college students. Sleep and Hypnosis, 2(2),
Gotti, C., Zoli, M., & Clementi, F. (2006). Brain nicotinic acetyl- 74-83.
choline receptors: native subtypes and their relevance. Kalinchuk, A., McCarley, R., Porkka‐Heiskanen, T., & Basheer,
Trends in pharmacological sciences, 27(9), 482-491. R. (2011). The time course of adenosine, nitric oxide
Grace, K. P., Vanstone, L. E., & Horner, R. L. (2014). Endog- (NO) and inducible NO synthase changes in the brain
enous cholinergic input to the pontine REM sleep gen- with sleep loss and their role in the non‐rapid eye move-
erator is not required for REM sleep to occur. Journal of ment sleep homeostatic cascade. Journal of neuro-
Neuroscience, 34(43), 14198-14209. chemistry, 116(2), 260-272.
Gyulaházi, J., Varga, K., Iglói, E., Redl, P., Kormos, J., & Fül- Kato, Y., Nakai, Y., Imura, H., Chihara, K., & Ohgo, S. (1974). Ef-
esdi, B. (2015). The effect of preoperative suggestions fect of 5-hydroxytryptophan (5-HTP) on plasma prolac-
on perioperative dreams and dream recalls after admin- tin levels in man. The Journal of Clinical Endocrinology
istration of different general anesthetic combinations: a & Metabolism, 38(4), 695-697.
randomized trial in maxillofacial surgery. BMC anesthe- Keks, N., Hope, J., & Keogh, S. (2016). Switching and stopping
siology, 15(1), 11. antidepressants. Australian prescriber, 39(3), 76-83.
Happe, S., Berger, K., & FAQT Study Investigators. (2001). The Kern, S., Appel, K., Schredl, M., & Pipa, G. (2017). No effect of
association of dopamine agonists with daytime sleepi- α GPC on lucid dream induction or dream contentKeine
ness, sleep problems and quality of life in patients with Wirkung von α GPC auf die Induktion von Klarträumen
Parkinson’s disease. Journal of neurology, 248(12), oder Trauminhalte. Somnologie, 21(3), 180-186.
1062-1067.

116 International Journal of Dream Research Volume 11, No. 2 (2018)


A pharmacological view on lucid dream induction IJoDR

Kim, D. K., Joo, Y., Sung, T. Y., Kim, S. Y., & Shin, H. Y. (2011). Lou, Z., Chen, W., Xue, W., Ding, J., Yang, Q., & Wang, H.
Dreaming in sedation during spinal anesthesia: a com- (2017). Dietary intake of magnesium-L-threonate allevi-
parison of propofol and midazolam infusion. Anesthesia ates memory deficits induced by developmental lead
& Analgesia, 112(5), 1076-1081. exposure in rats. RSC Advances, 7(14), 8241-8249.
Knapp, C. M., Datta, S., Ciraulo, D. A., & Kornetsky, C. (2007). Luciano, J. (2012). U.S. Patent No. 8,092,840. Washington, DC:
Effects of low dose cocaine on REM sleep in the freely U.S. Patent and Trademark Office.
moving rat. Sleep and biological rhythms, 5(1), 55-62. Mandell, M. P., Mandell, A. J., & Jacobson, A. (1965). Biochemi-
Knill, R. L., Moote, C. A., Skinner, M. I., & Rose, E. A. (1990). An- cal and neurophysiological studies of paradoxical sleep.
esthesia with abdominal surgery leads to intense REM Recent advances in biological psychiatry, 7, 115-122.
sleep during the first postoperative week. Anesthesiol- Mariño, J., & Cudeiro, J. (2006). How does the brain wake up?
ogy, 73(1), 52-61. The nitric oxide blow. Revista de neurologia, 42(9), 535-
Kondili, E., Alexopoulou, C., Xirouchaki, N., & Georgopoulos, 541.
D. (2012). Effects of propofol on sleep quality in me- Martinak, B., Bolis, R. A., Black, J. R., Fargason, R. E., & Birur,
chanically ventilated critically ill patients: a physiological B. (2017). Dextromethorphan in Cough Syrup: The Poor
study. Intensive care medicine, 38(10), 1640-1646. Man’s Psychosis. Psychopharmacology bulletin, 47(4),
Kraehenmann, R. (2017). Dreams and psychedelics: neurophe- 59-63.
nomenological comparison and therapeutic implica- Marzano, C., Ferrara, M., Mauro, F., Moroni, F., Gorgoni, M.,
tions. Current neuropharmacology, 15(7), 1032-1042. Tempesta, D., Cipolli, C. & De Gennaro, L. (2011). Re-
Kraehenmann, R., Pokorny, D., Aicher, H., Preller, K. H., Po- calling and forgetting dreams: theta and alpha oscil-
korny, T., Bosch, O. G., Seifritz, E. & Vollenweider, F. X. lations during sleep predict subsequent dream recall.
(2017). LSD Increases Primary Process Thinking via Se- Journal of Neuroscience, 31(18), 6674-6683.
rotonin 2A Receptor Activation. Frontiers in pharmacol- McComas, M., Harvey, A., & Hahn, B. (2015, December). Rela-
ogy, 8, 814. tionship Between the Effects of Nicotine on Subjective
Kunz, D., Mahlberg, R., Müller, C., Tilmann, A., & Bes, F. (2004). State and Vigilance Task Performance in Healthy Non-
Melatonin in patients with reduced REM sleep duration: smokers. In NEUROPSYCHOPHARMACOLOGY (Vol.
two randomized controlled trials. The Journal of Clinical 40, pp. S354-S355). Macmillan Building, 4 Crinan St,
Endocrinology & Metabolism, 89(1), 128-134. London N1 9xw, England: Nature Publishing Group.
LaBerge S. Lucid dreaming in western literature. InConscious Mei, Z., Zheng, P., Tan, X., Wang, Y., & Situ, B. (2017). Huperzine
mind, sleeping brain 1988 (pp. 11-26). Springer, Boston, A alleviates neuroinflammation, oxidative stress and im-
MA. proves cognitive function after repetitive traumatic brain
LaBerge, S., 2004. Substances that enhance recall and lucidity injury. Metabolic Brain Disease, 1-9.
during dreaming. U.S. Patent Application 10/604,138. Mihalak, K. B., Carroll, F. I., & Luetje, C. W. (2006). Varenicline
Lagos, P., Torterolo, P., Jantos, H., Chase, M. H., & Monti, J. is a partial agonist at α4β2 and a full agonist at α7 neuro-
M. (2009). Effects on sleep of melanin-concentrating nal nicotinic receptors. Molecular pharmacology, 70(3),
hormone (MCH) microinjections into the dorsal raphe 801-805.
nucleus. Brain research, 1265, 103-110. Miller, N. S. (1991). The Pharmacology of Phencyclidine. In The
Lavedan, C., Forsberg, M., & Gentile, A. J. (2015). Tasimelteon: Pharmacology of Alcohol and Drugs of Abuse and Ad-
a selective and unique receptor binding profile. Neuro- diction (pp. 259-264). Springer, New York, NY.
pharmacology, 91, 142-147. Miyazaki, S., Uchida, S., Mukai, J., & Nishihara, K. (2004).
Lazic, K., Petrovic, J., Ciric, J., Kalauzi, A., & Saponjic, J. (2017). Clonidine effects on all‐night human sleep: Opposite
REM sleep disorder following general anesthesia in rats. action of low‐and medium‐dose clonidine on human
Physiology & behavior, 168, 41-54. NREM–REM sleep proportion. Psychiatry and clinical
Leathwood, P. D., & Chauffard, F. (1985). Aqueous extract of neurosciences, 58(2), 138-144.
valerian reduces latency to fall asleep in man. Planta Montangero, J. (2018). Dreaming and REM-sleep: History of a
medica, 51(02), 144-148. scientific denial whose disappearance entailed a recon-
Lee, S. H., Choi, B.Y., Kim, J.H., Kho, A.R., Sohn, M., Song, ciliation of the neuroscience and the cognitive psycho-
H.K., Choi, H.C., & Suh, S.W. (2017). Late treatment logical approaches to dreaming. International Journal of
with choline alfoscerate (l-alpha glycerylphosphorylcho- Dream Research, 11(1), 30-45.
line, α-GPC) increases hippocampal neurogenesis and Morgan, T. O., Anderson, A. I., & MacInnis, R. J. (2001). ACE
provides protection against seizure-induced neuronal inhibitors, beta-blockers, calcium blockers, and diuret-
death and cognitive impairment. Brain research, 1654, ics for the control of systolic hypertension. American
66-76. journal of hypertension, 14(3), 241-247.
Leger, D., Quera-Salva, M. A., Vecchierini, M. F., Ogrizek, P., Moriguchi, S., Marszalec, W., Zhao, X., Yeh, J. Z., & Narahashi,
Perry, C. A., & Dressman, M. A. (2015). Safety profile of T. (2004). Mechanism of action of galantamine on N-
tasimelteon, a melatonin MT1 and MT2 receptor ago- methyl-D-aspartate receptors in rat cortical neurons.
nist: pooled safety analyses from six clinical studies. Journal of Pharmacology and Experimental Therapeu-
Expert opinion on drug safety, 14(11), 1673-1685. tics, 310(3), 933-942.
Li, W., Yu, J., Liu, Y., Huang, X., Abumaria, N., Zhu, Y., Huang, Nagano, I. (2008). Myristica fragrans: an exploration of the nar-
X., Xiong, W., Ren, C., Liu, X. G., & Chui, D. (2014). El- cotic spice. The Entheogen Review. Vernal Equinox,
evation of brain magnesium prevents synaptic loss and 16(1):15-24.
reverses cognitive deficits in Alzheimer’s disease mouse NHMRC (2015, March). NHMRC Statement on Homeopathy
model. Molecular brain, 7(1), 65. and NHMRC Information Paper - Evidence on the effec-
Little, J. T., Walsh, S., & Aisen, P. S. (2008). An update on hu- tiveness of homeopathy for treating health conditions.
perzine A as a treatment for Alzheimer’s disease. Expert Retrieved from https://www.nhmrc.gov.au/guidelines-
opinion on investigational drugs, 17(2), 209-215. publications/cam02

International Journal of Dream Research Volume 11, No. 2 (2018) 117


IJoDR A pharmacological view on lucid dream induction

Nirmala, B. C., Sowmya, G., Supriya, D. P., & Vinita, V. T. (2016). Ramesh, V., Thakkar, M. M., Strecker, R. E., Basheer, R., & Mc-
A comparative study of sedation with dexmedetomidine Carley, R. W. (2004). Wakefulness-inducing effects of
or midazolam during spinal anesthesia. AAnaesth Pain histamine in the basal forebrain of freely moving rats.
& Intensive Care, 20(3), 328-333. Behavioural brain research, 152(2), 271-278.
Nakazawa, Y., Hasuzawa, H., Kotorii, T., Ohkawa, T., Sakurada, Rhee, Y. H., Lee, S. P., Honda, K., & Inoué, S. (1990). Panax
H., Nonaka, K., & Dainoson, K. (1980). Study on the Ef- ginseng extract modulates sleep in unrestrained rats.
fects of L‐5HTP on the Stages of Sleep in Man as Evalu- Psychopharmacology, 101(4), 486-488.
ated by Using Sleep Deprivation. Psychiatry and Clini- Robinson, C. R., Pegram, G. V., Hyde, P. R., Beaton, J. M., &
cal Neurosciences, 34(2), 83-87. Smythies, J. R. (1977). The effects of nicotinamide upon
Novak, M., & Shapiro, C. M. (1997). Drug-induced sleep distur- sleep in humans. Biological psychiatry, 12(1), 139-143.
bances. Drug safety, 16(2), 133-149. Rosales-Lagarde, A., Armony, J. L., del Río-Portilla, Y., Trejo-
Nowak, L., Bregestovski, P., Ascher, P., Herbet, A., & Prochi- Martínez, D., Conde, R., & Corsi-Cabrera, M. (2012). En-
antz, A. (1984). Magnesium gates glutamate-activated hanced emotional reactivity after selective REM sleep
channels in mouse central neurones. Nature, 307(5950), deprivation in humans: an fMRI study. Frontiers in be-
462-465. havioral neuroscience, 6, 25.
Oswald, I., Ashcroft, G. W., Berger, R. J., Eccleston, D., Evans, Samuelsson, P., Brudin, L., & Sandin, R. H. (2008). BIS does not
J. I., & Thacore, V. R. (1966). Some experiments in the predict dreams reported after anaesthesia. Acta Anaes-
chemistry of normal sleep. The British Journal of Psy- thesiologica Scandinavica, 52(6), 810-814.
chiatry, 112(485), 391-399. Sanford, L. D., Tejani-Butt, S. M., Ross, R. J., & Morrison, A.
Ozdemir, P. G., Karadag, A. S., Selvi, Y., Boysan, M., Bilgili, S. R. (1995). Amygdaloid control of alerting and behavioral
G., Aydin, A., & Onder, S. (2014). Assessment of the arousal in rats: involvement of serotonergic mecha-
effects of antihistamine drugs on mood, sleep quality, nisms. Archives italiennes de biologie, 134(1), 81-99.
sleepiness, and dream anxiety. International journal of Sangare, A., Dubourget, R., Geoffroy, H., Gallopin, T., & Ran-
psychiatry in clinical practice, 18(3), 161-168. cillac, A. (2016). Serotonin differentially modulates
Page, F., Coleman, G., & Conduit, R. (2006). The effect of trans- excitatory and inhibitory synaptic inputs to putative
dermal nicotine patches on sleep and dreams. Physiol- sleep-promoting neurons of the ventrolateral preoptic
ogy & behavior, 88(4-5), 425-432. nucleus. Neuropharmacology, 109, 29-40.
Pagel, J. F. (2010). Drugs, dreams, and nightmares. Sleep Medi- Saunders, D. T., Roe, C. A., Smith, G., & Clegg, H. (2016). Lucid
cine Clinics, 5(2), 277-287. dreaming incidence: A quality effects meta-analysis of
Pagel, J. F., & Helfter, P. (2003). Drug induced nightmares—an 50 years of research. Consciousness and cognition, 43,
etiology based review. Human Psychopharmacology: 197-215.
Clinical and Experimental, 18(1), 59-67. Savage, R. L., Zekarias, A., & Caduff-Janosa, P. (2015). Vareni-
Pal, D., & Mallick, B. N. (2006). Role of noradrenergic and GA- cline and abnormal sleep related events. Sleep, 38(5),
BA-ergic inputs in pedunculopontine tegmentum for 833-837.
regulation of rapid eye movement sleep in rats. Neuro- Schiff, I., Regestein, Q., Tulchinsky, D., & Ryan, K. J. (1979). Ef-
pharmacology, 51(1), 1-11. fects of estrogens on sleep and psychological state of
Passie, T., Halpern, J. H., Stichtenoth, D. O., Emrich, H. M., & hypogonadal women. Jama, 242(22), 2405-2407.
Hintzen, A. (2008). The pharmacology of lysergic acid Schredl, M., & Erlacher, D. (2004). Lucid dreaming frequency
diethylamide: a review. CNS Neuroscience & Therapeu- and personality. Personality and Individual Differences,
tics, 14(4), 295-314. 37(7), 1463-1473.
Patterson, F., Grandner, M. A., Malone, S. K., Rizzo, A., Davey, Schredl, M., & Erlacher, D. (2011). Frequency of lucid dreaming
A. & Edwards, D. G. (2017). Sleep as a Target for Op- in a representative German sample. Perceptual and mo-
timized Response to Smoking Cessation Treatment. tor skills, 112(1), 104-108.
Nicotine & Tobacco Research, ntx236. Serefko, A., Szopa, A., & Poleszak, E. (2016). Magnesium and
Peever, J., Luppi, P. H., & Montplaisir, J. (2014). Breakdown in depression. Magnesium research, 29(3), 112-119.
REM sleep circuitry underlies REM sleep behavior dis- Sharpley, A. L., McGavin, C. L., Whale, R., & Cowen, P. J. (1998).
order. Trends in neurosciences, 37(5), 279-288. Antidepressant-like effect of Hypericum perforatum (St
Peuhkuri, K., Sihvola, N., & Korpela, R. (2012). Diet promotes John’s wort) on the sleep polysomnogram. Psychophar-
sleep duration and quality. Nutrition research, 32(5), macology, 139(3), 286-287.
309-319. Shinomiya, K., Fujimura, K., Kim, Y., & Kamei, C. (2005). Effects
Pinter, M. M., Pogarell, O., & Oertel, W. H. (1999). Efficacy, of valerian extract on the sleep-wake cycle in sleep-dis-
safety, and tolerance of the non-ergoline dopamine turbed rats. Acta Med Okayama, 59(3), 89-92.
agonist pramipexole in the treatment of advanced Par- Siegel, J. (2004). The neurotransmitters of sleep. Journal of
kinson’s disease: a double blind, placebo controlled, Clinical Psychiatry, 65(Suppl 16), 4-7.
randomised, multicentre study. Journal of Neurology, Siegel, J. M. (2005). REM sleep. Principles and practice of sleep
Neurosurgery & Psychiatry, 66(4), 436-441. medicine, 4, 120-135.
Placidi, F., Diomedi, M., Scalise, A., Marciani, M. G., Romigi, A., Siegel, R. K. (1978). Phencyclidine and ketamine intoxication:
& Gigli, G. L. (2000). Effect of anticonvulsants on noctur- a study of four populations of recreational users. NIDA
nal sleep in epilepsy. Neurology, 54(5 Suppl 1), S25-32. Res Monogr, 21(21), 119-147.
Popoviciu, L., Aşgian, B., Delast-Popoviciu, D., Alexandrescu, Smith, K. (2012). Homeopathy is unscientific and unethical.
A., Petruţiu, S., & Bagathal, I. (1993). Clinical, EEG, elec- Bioethics, 26(9), 508-512.
tromyographic and polysomnographic studies in rest- Smith, R. L., & Kennedy, C. H. (2003). Increases in avoid-
less legs syndrome caused by magnesium deficiency. ance responding produced by REM sleep deprivation
Revue Roumaine de Neurologie et Psychiatrie. or serotonin depletion are reversed by administration
Portas, C. M., Bjorvatn, B., & Ursin, R. (2000). Serotonin and of 5-hydroxytryptophan. Behavioural brain research,
the sleep/wake cycle: special emphasis on microdialy- 140(1-2), 81-86.
sis studies. Progress in neurobiology, 60(1), 13-35.

118 International Journal of Dream Research Volume 11, No. 2 (2018)


A pharmacological view on lucid dream induction IJoDR

Solomon, T., Leech, J., deBros, G., Murphy, C., Budson, A., Weng, F. J., Williams, R. H., Hawryluk, J. M., Lu, J., Scammell, T.
Vassey, E., & Solomon, P. (2016). A randomized, double‐ E., Saper, C. B., & Arrigoni, E. (2014). Carbachol excites
blind, placebo controlled, parallel group, efficacy study sublaterodorsal nucleus neurons projecting to the spinal
of alpha BRAIN® administered orally. Human Psycho- cord. The Journal of physiology, 592(7), 1601-1617.
pharmacology: Clinical and Experimental, 31(2), 135- Wetter, D. W., & Young, T. B. (1994). The relation between ciga-
143. rette smoking and sleep disturbance. Preventive medi-
Soto-Rodriguez, S., Lopez-Armas, G., Luquin, S., Ramos- cine, 23(3), 328-334.
Zuñiga, R., Jauregui-Huerta, F., Gonzalez-Perez, O., & Winkelman, J. W., & James, L. (2004). Serotonergic antidepres-
Gonzalez-Castañeda, R. E. (2016). Rapid eye move- sants are associated with REM sleep without atonia.
ment sleep deprivation produces long-term detrimental Sleep, 27(2), 317-321.
effects in spatial memory and modifies the cellular com- Woolf, N., & Butcher, L. (2011). Cholinergic systems mediate
position of the subgranular zone. Frontiers in cellular action from movement tohigher consciousness. Behav-
neuroscience, 10, 132. ioural brain research, 221(2), 488-498.
Sparrow, G. S., Hurd, R., & Carlson, R. (2016). Assessing the Wyatt, R. J., Zarcone, V., Engelman, K., Dement, W. C., Snyder,
perceived differences in post-Galantamine lucid dreams F., & Sjoerdsma, A. (1971). Effects of 5-hydroxytrypto-
vs. non-Galantamine lucid dreams. International Jour- phan on the sleep of normal human subjects. Clinical
nal of Dream Research, 9(1), 71-74. Neurophysiology, 30(6), 505-509.
Srinivasan, V., Pandi-Perumal, S. R., Trahkt, I., Spence, D. W., Yang, N., Zhang, K. Y., Wang, F. F., Hu, Z. A., & Zhang, J. (2014).
Poeggeler, B., Hardeland, R., & Cardinali, D. P. (2009). Dopamine inhibits neurons from the rat dorsal subcoe-
Melatonin and melatonergic drugs on sleep: possible ruleus nucleus through the activation of α2-adrenergic
mechanisms of action. International Journal of Neuro- receptors. Neuroscience letters, 559, 61-66.
science, 119(6), 821-846. Yuschak, T. (2006). Advanced Lucid Dreaming, The Power of
Stanniland, C., & Taylor, D. (2000). Tolerability of atypical anti- Supplements. Lulu Enterprises, Raleigh.
psychotics. Drug safety, 22(3), 195-214. Zhdanova, I. V., & Wurtman, R. J. (1997). Efficacy of melato-
Stumbrys, T., & Erlacher, D. (2012). Lucid dreaming during nin as a sleep-promoting agent. Journal of Biological
NREM sleep: Two case reports. International Journal of Rhythms, 12(6), 644-650.
dream research, 5(2), 151-155.
Stumbrys, T., & Erlacher, D. (2014). The science of lucid dream
induction. Lucid Dreaming: New Perspectives on Con-
sciousness in Sleep, 1, 77-102.
Stumbrys T, Erlacher D, Schädlich M, Schredl M (2012). Induc-
tion of lucid dreams: A systematic review of evidence.
Consciousness and Cognition, 21(3),1456-1475.
Texidó, L., Ros, E., Martín‐Satué, M., López, S., Aleu, J., Marsal,
J., & Solsona, C. (2005). Effect of galantamine on the
human α7 neuronal nicotinic acetylcholine receptor, the
Torpedo nicotinic acetylcholine receptor and spontane-
ous cholinergic synaptic activity. British journal of phar-
macology, 145(5), 672-678.
Thompson, D. F., & Pierce, D. R. (1999). Drug-induced night-
mares. Annals of Pharmacotherapy, 33(1), 93-98.
Tung, A., & Mendelson, W. B. (2004). Anesthesia and sleep.
Sleep medicine reviews, 8(3), 213-225.
Uher, R., & Jones, H. M. (2006). Hallucinations during lamotrig-
ine treatment of bipolar disorder. American Journal of
Psychiatry, 163(4), 749-750.
Ursin, R. (1976). The effects of 5-hydroxytryptophan andl-tryp-
tophan on wakefulness and sleep patterns in the cat.
Brain research, 106(1), 105-115.
Ursin, R. (2002). Serotonin and sleep. Sleep medicine reviews,
6(1), 55-67.
Van der Most, P. J., Dolga, A. M., Nijholt, I. M., Luiten, P. G., &
Eisel, U. L. (2009). Statins: mechanisms of neuroprotec-
tion. Progress in neurobiology, 88(1), 64-75.
Van Eeden, F. (1913). A study of dreams. Proceedings of the
Society for Psychical Research, 26(47), 431-461.
Voss, U. (2010). Lucid dreaming: Reflections on the role of in-
trospection. International Journal of Dream Research,
3(1), 52-53.
Watson, C. J., Baghdoyan, H. A., & Lydic, R. (2010). Neurophar-
macology of sleep and wakefulness. Sleep medicine
clinics, 5(4), 513-528.
Watson, C. J., Baghdoyan, H. A., & Lydic, R. (2012). Neuro-
pharmacology of sleep and wakefulness: 2012 update.
Sleep medicine clinics, 7(3), 469-486.

International Journal of Dream Research Volume 11, No. 2 (2018) 119

Vous aimerez peut-être aussi