You are on page 1of 9

Archives of Medical Research 50 (2019) 518e526

REVIEW ARTICLE
Endocannabinoids as Therapeutic Targets
Oscar Prospero-Garcıa,a Alejandra E. Ruiz Contreras,b Alette Ortega G
omez,c Andrea Herrera-Solıs,d
Monica Mendez-Dıaz,a and Grupo de Neurociencias de la Universidad Nacional Autonoma de Mexico
a
Departamento de Fisiologıa, Laboratorio de Canabinoides, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
b
Laboratorio de Neurogenomica Cognitiva, Facultad de Psicologıa, Universidad Nacional Autonoma de Mexico, Ciudad de Mexico, Mexico
c
Laboratorio de Medicina Traslacional, Instituto Nacional de Cancerologıa, Ciudad de Mexico, Mexico
d
Laboratorio Efectos Terapeuticos de los Canabinoides, Subdireccion de Investigacion Biomedica, Hospital General Dr. Manuel Gea Gonzalez, Ciudad de
Mexico, Mexico
Received for publication April 30, 2019; accepted September 30, 2019 (ARCMED_2019_385).

Most of the drugs of abuse affect the brain by interacting with naturally expressed mo-
lecular receptors. Marihuana affects a series of receptors including cannabinoid receptor
1 (CB1R) and CB2R, among others. Endogenous molecules with cannabinoid activity
interact with these receptors naturally. Receptors, ligands, synthesizing and degrading en-
zymes, as well as transporters, have been described. This endocannabinoid system mod-
ulates behaviors and physiological processes, i.e. food intake, the sleep-waking cycle,
learning and memory, motivation, and pain perception, among others. The rather broad
distribution of endocannabinoids in the brain explains the different effects marihuana in-
duces in its users. However, this very same anatomical and physiological distribution
makes this system a useful target for therapeutic endeavors. In this review, we briefly
discuss the potential of small molecules that target the endocannabinoids as therapeutic
tools to improve behaviors and treat illnesses. We believe that under medical supervision,
endocannabinoid targets offer new advantages for patients for controlling multiple med-
ical disorders. Ó 2019 IMSS. Published by Elsevier Inc.
Key Words: Cannabinoids, Endocannabinoids, Therapeutic properties, Sleep, Epilepsy, Pain,
Learning and memory.

Marihuana affects the brain by interacting with a series of how some molecules that target the eCB system under med-
naturally expressed receptors including cannabinoid recep- ical supervision are potential therapeutic tools to improve
tor 1 (CB1R) and CB2R. Endogenous molecules with behaviors and treat illnesses.
cannabinoid activity interact with these receptors naturally. Since the isolation and structural description of D9-
Similarly, receptors, ligands, synthesizing and degrading tetrahydrocannabinol (THC), the main psychoactive
enzymes, as well as transporters, have been described. This component of marihuana (Cannabis sativa) in 1964 (1),
endocannabinoid (eCB) system modulates behaviors and knowledge on how marihuana causes its effects in the user
physiological processes, that is food intake, the sleep- has expanded exponentially. Howlett’s suggestion of the ex-
waking cycle, learning and memory, motivation, and pain istence of a receptor to which THC binds (2), ignited the
perception, among others. The rather broad distribution of search for endogenous molecules. In 1992, Mechoulam’s
endocannabinoids in the brain explains the different effects group isolated anandamide (AEA) from pig brain (3). In
marihuana induces in its users. This anatomical distribution 1994, oleamide (ODA), another endocannabinoid was iso-
and physiological effects make this system a useful target lated from the cerebrospinal fluid of sleep-deprived cats
for therapeutic endeavors. In this review, we briefly discuss (4). In the same year, Mechoulam’s group isolated 2 arach-
ydonoylglycerol (2-AG) from dog intestine (5) (Table 1).
Few years after CB1R was cloned (6), CB2R were charac-
Address reprint requests to: Oscar Prospero-Garcıa, Departamento de terized (6,7). Enzymes that participate in the synthesis and
Fisiologıa, Facultad de Medicina, Universidad Nacional Autonoma de breakdown of endocannabinoids have also been identified,
Mexico, Apdo. Postal 70-250, Ciudad de Mexico, 04510 Mexico; Phone:
and the existence of an endocannabinoid transporter has
(þ52) 55 255 623 2509; FAX: (þ52) 55 2585 5623 2241.; E-mail: opg@
unam.mx been suggested (8). All this information has led to

0188-4409/$ - see front matter. Copyright Ó 2019 IMSS. Published by Elsevier Inc.
https://doi.org/10.1016/j.arcmed.2019.09.005
Cannabinoids and Medicine 519

Table 1. Endogenous cannabinoids (endocannabinoids) and related compounds

Endocannabinoids and related compounds Common name Functional properties at cannabinoid receptors

N-arachidonylethanolamine Anandamide Partial CB1R agonist, Weak CB2R agonist. Sleep and food intake inducer
2-arachidonoyl-glycerol 2-AG Full CB1R and CB2R agonist. Sleep and food intake inducer
Cis-9,10 octadecenamide Oleamide Weak CB1R, CB2R agonist. Sleep and food intake inducer
N-palmitoyl ethanolamine PEA PPAR-a agonist It inhibits the expression of FAAH
N-oleoyl ethanolamine OEA TRPV1 agonist PPARa agonist. Waking and anorexia inducer
2-Arachidonoylglyceryl ether Noladin ether CB1R agonist
O-arachidonoylethanolamine Virodhamine Partial CB2R agonist and CB1R antagonist
N-Arachidonoyl-Dopamine NADA CB1R agonist and potent TRPV1 agonist

expectations of cannabinoid-based therapies for treatment Marihuana is classified as a schedule I substance by the
of several illnesses and drug-dependence (Table 2). U.S. Drug Enforcement Administration (DEA), indicating
In the following section, we discuss some evidence sup- that marihuana is a drug with high potential for abuse.
porting the potential use of the facilitation or blockade of However, THC and a synthetic analog of THC (Dronabinol)
endocannabinoids to improve behaviors and illnesses. have been approved by the Food and Drug Administration
(FDA) for patients suffering from serious, chronic, or debil-
itating medical conditions. THC itself or THC analogs, as
Dronabinol, are approved for treatment of the severe nausea
Therapeutic Properties
and vomiting associated with cancer chemotherapy (16);
Ritual use of marihuana have been suggested by remains of weight loss associated with debilitating illnesses, such as
seeds and leaves found in China next to a human skeleton HIV infection or cancer (17); spasticity associated with
which was estimated to be 10 thousand years old (9). It neurological diseases, such as multiple sclerosis and dis-
has been speculated that marihuana has been used to cure eases accompanied by intense pain and glaucoma (18).
or control several diseases such as inflammation and pain, Otherwise, any physician prescribing marihuana for dis-
anxiety, depression and psychosis, fatigue and insomnia, eases different from the above mentioned is committing a
loss of appetite and digestive disorders, such as: nausea, crime in the USA and in other countries. In Mexico, the
diarrhea, and constipation, among others (10). Both mari- law is so permissive that those physicians that are prescrib-
huana and THC have been used to lower elevated intraoc- ing raw marijuana extracts to treat patients do not have to
ular pressure (glaucoma) (11). Similarly, the manipulation face legal charges. In the vast majority of countries, it is
of endogenous cannabinoids may be useful for the control definitely illegal to even suggest marihuana use. On the
of drug addiction (12). THC has been useful in asthma other hand, the information indicating that California,
and as an analgesic (13). Many of these alleged effects USA, has an estimated 100,000 users of medical marihuana
are still under investigation. Comparably, Cannabidiol is absolutely fascinating. Some of them claim to use mari-
(CBD) seems to have properties to control epilepsy (14), huana for medical conditions, but they may be faking the
and in combination with THC to control pain (15). disease to obtain marihuana for recreational purposes.

Table 2. Synthetic cannabinoids

Compound Characteristics

CP-55940 CB1R agonist. Highly psychoactive. 40 times more potent than THC
HU-210 CB1R full agonist. 100 times more potent than THC
WIN55,212-2 CB1R agonist
JWH-133 CB2R agonist
URB597 (KDS-4103) FAAH selective inhibitor
AM251 CB1R antagonist. Potential anti-hypersomnia aid
AM404 Endogenous cannabinoid reuptake inhibitor, Vanilloid receptor agonist.
SR144528 CB2R antagonist

Therapeutic Compound Use


Dronabinol (Marinol) THC analog. Prescribed as an appetite stimulant for patients with AIDS, chemotherapy and gastric bypass patients.
Antiemetic. Potential sleep inducer and pain killer
Nabilone (Cesament) THC analog. Antiemetic and analgesic. Potential sleep inducer and pain killer
Rimonabant (SR141716A, Complain, CB1R Inverse agonist. Anorectic anti-obesity. Prescribed for patients with body mass index greater than 30 kg/m2
Riobant, Slimona) or 27 kg/m2 associated to risk factor. Potential anti-hypersomnia aid
Sativex THC and CBD analog, used for neuropathic pain of multiple sclerosis
520 Prospero-Garcıa et al./ Archives of Medical Research 50 (2019) 518e526

Addiction and pathology associated with marijuana abuse but of the commercial availability of rimonabant. The main
psychiatric symptoms rimonabant induced, according to the
Some studies indicate that increasing endocannabinoids by
Food and Drug Administration report (2007), were anxiety
inhibiting their breakdown enzymes prevent alcohol, nico-
(more than 3% with 5 mg and more than 6% with 20 mg),
tine or even heroin reinstatement (19e21). In contrast, some
insomnia (almost 3% with 5 mg and more than 5% with
other studies relate endocannabinoids and motivation and
20 mg), depressed mood (almost 3% with 5 mg, almost
drug dependence (12,22), that is it has been observed that
4% with 20 mg), depression (more than 2% with 5 mg
CB1R blockade may prevent development of drug-seeking
and more than 3% with 20 mg), and several other minor
behavior and reinstatement in rats and mice. However,
problems were also observed.
CB1R blockade in humans, may induce negative emotional
CB2R exhibits a role opposite to CB1R role. Some data
effects. Regarding CB2R, its activation inhibits cocaine
support obesity leads to increased expression of the CB2R,
self-administration (23); however, CB2R KO mice do not
in both adipose tissue and liver, in high-fat fed and ob/ob
develop nicotine-induced conditioned place preference
mice (39). While CB2R deficiency prevents body weight
(CPP) (24). These dissimilar effects seem to be parts of the
gain during the feeding of a high-fat diet and obesity-
same puzzle that are, at present time, difficult to conciliate.
associated inflammation, insulin resistance and fatty liver
Further studies will shed light to this contradiction.
(40). It should be noted that unlike CB1R the activation
Regarding the effects of marijuana chronic use, delete-
of CB2R reduces VTA DA-neurons firing, suggesting that
rious effects have been consistently reported. Therefore,
these receptors may be a therapeutic target in obesity
the Diagnostic and Statistical Manual of Mental Disorders
without affecting hedonism.
V (DSM, American Psychiatric Association, APA, 2013)
includes a description of marijuana dependence syndrome
(25). Marijuana tolerance and dependence (26) includes
anxiety, irritability, insomnia, hyporexia, restlessness, and eCB System also Regulate Food Intake
an intense desire for consuming marijuana (craving), after
ODA, AEA and 2-AG increase food intake in rats (41e43),
24e74 h of abstinence. In addition, marijuana users have
By acting on brain structures like the hypothalamus and the
several brain structural changes, that is reduction in the hip-
nucleus accumbens (44e46). Rats exhibit CPP induced by
pocampus, amygdala, nucleus accumbens and prefrontal
regular Lab Chow food, but CPP is stronger when they are
cortex, and a reduction of their connections, when they
under the effect of OLE or AEA. Such CPP is prevented by
consume it before they were 20 years of age (27e29). Simi-
CB1 inverse agonist AM251, suggesting that OLE and
larly, some studies have suggested a reduction in the Intel-
AEA increase the food palatability (47). Hence, CB1R
ligence Quotient (IQ) (30,31), while other studies revealed
seems to mediate in part the liking phase of reward. More-
that marijuana induces the first psychotic or bipolar
over, endocannabinoids increase its levels in a close rela-
outbreak in susceptible users (32). Due to these consider-
tionship with feeding (48). AEA, AA5HT (FAAH
ations, although marijuana derivatives may have some me-
inhibitor) and OMDM-1 (AEA reuptake inhibitor) increase
dicinal properties, it is advisable that their use must be
AEA and 2-AG levels in the NAc, while inducing hypotha-
subjected to strict medical supervision.
lamic nuclei activation and food intake (46). All these ef-
fects were prevented by AM251 (46). This study opens a
potential avenue to target endocannabinoid-breakdown
molecules for treatment of eating disorders.
Food Intake
Two or three marihuana joints increase average daily
caloric intake, mostly as in between-meal snacks that cause
Sleep
an increase in meal size per se. An increase in body weight
was also observed (33,34). THC increases food intake in Users report, anecdotally, that marihuana induces drowsi-
satiated rats (35); however, they consume sweet more than ness or sleepiness. Babor TF, et al. (49) observed that mod-
less palatable foods (36). Free-feeding rats receiving THC erate to heavy marihuana use induced better sleep during
increase their high-fat and high-fat sweetened food intake the days following the consumption. Regrettably, heavy
(37). Complementary, SR141716A (rimonabant, a CB1R use of marihuana may trigger anxiety crises, and psychotic
antagonist) induces anorectic effects (38). Hence, cannabi- outbreaks, complicating its use to induce sleep (50). Both
noids effects on food intake occur through the activation of ODA and AEA increase sleep, particularly REM sleep, in
the CB1R. This finding opens the possibility to use these rats. One single dose of either systemic ODA or intra lateral
kinds of drugs as pharmacological tools in the treatment hypothalamus 2-AG increases REM sleep. Intracerebroven-
of obesity, see rimonabant in obesity (RIO) study by Van tricular infusion of either ODA or 2-AG increases REM
Gaal (38). Volunteers in this study developed psychiatric sleep after one single dose or after 15 d of daily administra-
symptoms that led to the suspension, not only of the study, tion, see review (51). Moreover, both ODA and 2-AG
Cannabinoids and Medicine 521

increase NREM and REM sleep in a rodent model of deficits on working and short-term memory in rats and mice
insomnia (52,53). Congruently, Santucci V, et al. (54) re- (74). Terranova JP, et al. (75) described that SR141716A fa-
ported that SR141716A increases wakefulness. Similarly, cilitates memory formation (76), and delays the extinction
fatty-acid amide hydrolase (FAAH)-knockout (KO) mice of fear conditioning (77).
(FAAH hydrolyzes AEA and ODA) exhibit more NREM Cannabis users have shown an impairment in memory
sleep compared to their wild type (WT) littermates (55). tasks performance (78,79). THC induces WM deficiency
While CB1R-KO mice spent more time in W than their while reducing the activity of the dorsolateral prefrontal
WT littermates (56,57). In humans, the FDA meta- cortex, posterior parietal cortex, anterior cingulate and tem-
analysis (2007) of the RIO Europe study (38) emphasized poral cortices (80). Moreover, a reduction of BOLD signal
that more than 5% of the volunteers receiving rimonabant at the posterior parietal cortex was detected in adults with
(20 mg) developed insomnia. These findings complement early onset of cannabis use when performing a working
those obtained in animal models, and further support the memory task, even if subjects smoked only a few times,
notion that the facilitation of the eCB system may improve as long as it was at early age (81).
sleep in insomniac patients. Contrasting studies have failed to document memory
Regarding Cannabidiol (CBD), it is one of the main deficiency in cannabis users (82,83), promoting controversy
components of Cannabis sativa, that antagonizes the bind- regarding cannabis effects on memory (84,85). It should be
ing of CB1 and CB2 receptor agonists (58). At first, it considered that THC negative effect on memory can be
was reported that CBD decreased the latency to sleep onset, attenuated by CBD (86). Studies in rats have suggested that
while increasing duration of non-rapid-eye-movement cannabis effect in memory in humans could depend on
(NREM) sleep (59). Chagas MH, et al. 2013 (60), reported doses (87), that is THC low doses (3 mg/kg) disrupts
an increase in the total amount of sleep in rats during the WM performance, whereas high doses (10 mg/kg) affects
light phase. Conversely, some other studies reported that spatial reference memory in mice (88). In rats, a
CBD increased waking and decreased REM sleep, leaving semichronic (7d) and chronic (21d) THC administration
NREM sleep unchanged (61,62). Some studies have re- induced hippocampal neurogenesis and upregulation of
ported that CBD (600 mg) induced sedative effects in brain-derived neurotrophic factor, while inducing a better
healthy volunteers (63); while some others have shown performance in short-term and long-term memory novel-
that low doses of CBD (15 mg/d) combined with THC recognition tasks (89). Subjects genetic variation on cogni-
(15 mg/d) increased wakefulness while reducing NREM tive performance should be considered, too (90e92), that is
sleep stage 3 (64). An absence of effects have also been re- polymorphisms of the CNR1, gene which codes for CB1R,
ported in healthy subjects with 300 mg of 99% pure CBD have been associated with differential efficiency in WM
(65). Unfortunately, the experimental evidence do not sup- and attentional control in healthy subjects (90,91). It is
port CBD sleep inducer. possible that some genotypes are less vulnerable to the
As an additional note, Nabilone, a synthetic CB1R agonist, deleterious effect on memory when cannabis is consumed.
has exhibited sleep-promoting effects in patients suffering Endocannabinoids also participate in memory processes.
from fibromyalgia, very possible by reducing pain (66). In 1998, our group reported the memory-modulating prop-
erties of AEA (93). AEA impairs memory consolidation in
rats in an inhibitory avoidance task, further supporting that
cannabinoids regulate memory acquisition or retrieve (94).
Learning and Memory
However, it is also possible that endocannabinoids modu-
The effect of marihuana and endocannabinoids on learning late learning and memory by regulating the use of different
and memory has been studied intensively (67e69). Hence, strategies to solve a memory task (95).
it is now accepted that endocannabinoids play a crucial role However, one eCB system important role in memory
in the modulation of cognitive processes (70). regulation is in memory extinction. For example, CB1R
The CB1R mediates the impairing effect induced by knockout mice exhibit a significant deficit in solving a
cannabinoids on memory. CB1R is widely expressed in spatial task, that is the water maze (69). The principal
memory systems, such as in the hippocampus (71). Rats un- deficit observed in these mice is in memory extinction, that
der THC effects performed worse than vehicle rats in a has been interpreted as an inability to substitute old strate-
working memory (WM) task (72). CB1R agonists reduce gies with newer and more adaptive ones. CB1R activation
hippocampal neurons firing rate during encoding and delay in the amygdala promotes aversive memories extinction
period in WM in rats (73). In contrast, CB1R knock-out (96) and prevents the development of post-traumatic stress
mice were able to learn the Morris water maze, albeit disorder (PTSD)-like in rats, such as enhancement of corti-
increasing perseverance errors after the platform position costerone levels and the startle response generated by a
was changed (69), suggesting that CB1R participates in ex- single-prolonged stress (97). Furthermore, stressed mice in-
tinguishing memories and updating new information in crease the endocannabinoids levels in the basolateral amyg-
WM. Congruently, SR141716A prevents THC-induced dala during the extinction process, and if they are treated
522 Prospero-Garcıa et al./ Archives of Medical Research 50 (2019) 518e526

with SR141716A the aversive memory extinction is de- proliferative effects and cell death comprise: inhibition of
layed (74). In addition, it has been shown that CBD pre- mitogenic proteins, sustained stimulation of ceramide and
vented the consolidation of aversive memories in a fear inhibition of RAS-MAPK pathway, through pro-apoptotic
conditioning task when it is given immediately after the proteins such as BCL-2 (112,113). It is already known that
acquisition (98). somatic or hereditary mutations on cancer, like KRAS or
Another line of therapeutic target that is an open window PI3K act like active mitogenic proteins (114,115). This ge-
of research opportunity is cognitive function in ageing. netic heterogeneity among tumoral tissues is associated to a
Some studies have revealed that the systemic administra- wide range of effects through cannabinoids actions. In this
tion of low doses of THC (3 mg/kg) during 28 d improved context, we speculate that cannabinoids interact directly
learning and memory in mature (12 m) and old mice with active mutations exerting cytotoxic effects, inhibiting
(18 m), behaving like young vehicle animals. In these aged proliferation, migration or activating apoptotic mechanisms
animals, also, several synaptic, spine density and synaptic by increasing caspase-3 and PARP activation (116).
formation proteins increased (99). Cannabinoids promote different cell death mechanisms
With all, proper doses of THC or CBD must be precisely as their anti-proliferative cancer actions. AEA induces late
determined to prevent adverse effects in memory, and in apoptosis/necrosis in different tumors; while synthetic ana-
other cognitive functions, such as attention, perception, ex- logs such as Meth-Anandamide induce early apoptosis and
ecutive functions, and decision making (78,100). CP,55-940 induces necrosis (117). Hence, cannabinoids
activate selective mechanisms that induce different patterns
Pain of cell death in different tumor cells. Further studies will
clarify the utility of these findings.
Activation of the eCB system appears to reduce neuropathic
Phytocannabinoids, synthetic cannabinoids and endo-
pain (101e103). Several studies have documented a reduc-
cannabinoids exert their anti-tumoral effects by a negative
tion in the perception of nociceptive stimuli in animal
cross-talk between GPR55 and CB2 receptors, and a bidi-
models (104) and pain in humans. Although results are very
rectional cross-antagonism between both receptors (118).
consistent, some controversies have arisen since the advan-
These molecular mechanisms highlight the importance of
tages of using CB1R agonists over already commercially
inhibiting endocannabinoids synthesizing and degrading
available drugs are dubious. Since some cannabinoids, that
enzymes as anti-proliferative actions demand (118e120).
is nabilone, marinol, have efficacy as pain relievers as some
data in the clinic have shown, and may target neuropathic
pain, as with opioids, the challenge remains to find pain
relieving endocannabinoid modulating molecules that do Epilepsy
not have abuse potential (66).
Although a wide variety of medication is available to treat epi-
lepsy, at least one-third of patients are drug-resistant (121), that
Cancer
is, the current medication cannot control their seizures. Several
The involvement of metabolic changes in the eCB system cultures have used cannabis to treat seizures, including the Su-
associated with cancer has been documented. Regrettably, merians around 1800 BCE and the Arabians in the 12th century
poor prognosis through CB1R receptor up-regulation in (122,123). In the 18th century, O’Shaughnessy and Gowers re-
malignant tissues has been describe until now (105,106). ported the use cannabis to treat medication-resistant epileptics
CB1R was considered a potential clinical target using direct (124,125). The effects of marijuana compounds have been stud-
or inverse agonists; however, as we have abovementioned ied more consistently in recent times.
these treatments interfere with normal cognitive function Mounting evidence supports the anti-seizure effects of
(107). Therefore, new indirect agonists have been devel- THC, CBD, cannabidavarin (CBDV), endocannabinoids,
oped for specific actions mediated by the activity of cata- and synthetic cannabinoid receptor agonists in animal
lytic enzymes and/or allosteric positive modulation of models (126). Although THC has anti-seizure effects, it
endocannabinoids activity (108e110). can also promote them. Again, as marijuana psychotropic
Munson AE, et al. reported anti-proliferative actions of effects that can result in tolerance and addiction, we have
phytocannabinoids in animal models, documenting THC to consider its therapeutic limitations.
and CBD inhibition of tumor growth, long before the dis- In this context, the case of Charlotte has captured the
covery of cannabinoid receptors and endocannabinoids attention of the general population. Charlotte, who has Dra-
(111). Nowadays, it is known that phytocannabinoids, en- vet syndrome, had nearly 50 seizures per day. After she
docannabinoids and synthetic cannabinoids exert inhibitory began to use a CBD: THC extract, along with her pre-
effects on cancer growth and spreading, in diverse cancer scribed antiepileptic drugs, her seizures decreased to 2 or
cell lines and animal models, albeit findings from pre- 3 nocturnal convulsions per month (127).
clinic studies remain under debate. The inhibition of CBD in combination with prescribed antiepileptic drugs
tumoral pathways in which cannabinoids exert anti- has exhibited anticonvulsive effects. One double-blind
Cannabinoids and Medicine 523

clinical trial compared CBD (200e300 mg) daily versus pla- 2. Howlett AC, Bidaut-Russell M, Devane WA, et al. The cannabinoid
cebo, for 4 months, in patients suffering from generalized receptor: biochemical, anatomical and behavioral characterization.
Trends Neurosci 1990;13:420e423.
epilepsy secondary to a temporal focus, reported that 4 out 3. Devane WA, Hanus L, Breuer A, et al. Isolation and structure of a
of 8 patients had virtually no convulsions during CBD treat- brain constituent that binds to the cannabinoid receptor. Science
ment, while 3 experienced partial improvements. One pa- 1992;258:1946e1949.
tient did not improve (128). The first open-label 4. Lerner RA, Siuzdak G, Prospero-Garcia O, et al. Cerebrodiene: a
interventional trial was conducted with 162 patients (chil- brain lipid isolated from sleep-deprived cats. Proc Natl Acad Sci U
S A 1994;91:9505e9508.
dren and adults) suffering from Dravet and Lennox- 5. Mechoulam R, Ben-Shabat S, Hanus L, et al. Identification of
Gastaut syndrome. CBD (Epidiolex) 2e5 mg/kg/d for an endogenous 2-monoglyceride, present in canine gut, that
12 weeks reduced seizures by 36% (129). Another study binds to cannabinoid receptors. Biochem Pharmacol 1995;50:
used CBD (Epidiolex) 20 mg/kg/d to treat 120 children 83e90.
and young adults with Dravet syndrome and drug-resistant 6. Matsuda LA, Lolait SJ, Brownstein MJ, et al. Structure of a canna-
binoid receptor and functional expression of the cloned cDNA. Na-
seizures, decreasing the frequency of convulsive seizures ture 1990;346:561e564.
per month from 12.4e5.9, and 5% of patients became 7. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a
seizure-free during the treatment (130). One study more, peripheral receptor for cannabinoids. Nature 1993;365:61e65.
included 171 children and middle-aged patients suffering 8. Yates ML, Barker EL. Organized trafficking of anandamide and
from Lennox-Gastaut syndrome. CBD (20 mg/kg/d) reduced related lipids. Vitam Horm 2009;81:25e53.
9. Mikuriya TH. Marijuana in medicine: past, present and future. Calif
the median percentage of seizures per month to 43.9%. More Med 1969;110:34e40.
studies have been conducted (131), all of them supported 10. Russo EB, Guy GW, Robson PJ. Cannabis, pain, and sleep: lessons
CBD anti-seizure effects. Regarding adverse effects, they from therapeutic clinical trials of Sativex, a cannabis-based medicine.
have been somnolence, diarrhea, decreased appetite, leth- Chem Biodivers 2007;4:1729e1743.
argy, fatigue, vomiting, and even convulsions that became 11. Flach AJ. Delta-9-tetrahydrocannabinol (THC) in the treatment of
end-stage open-angle glaucoma. Trans Am Ophthalmol Soc 2002;
severe (129e132). Elevated transaminase was common for 100:215e222. discussion 22e24.
patients using valproate medication and high doses of 12. Maldonado R, Valverde O, Berrendero F. Involvement of the endo-
CBD (133). Based on reports from the clinical trials, the cannabinoid system in drug addiction. Trends Neurosci 2006;29:
FDA approved Epidiolex as a first-line CBD medication 225e232.
for Dravet and Lennox-Gastaut syndrome (133). 13. Gilron I, Coderre TJ. Emerging drugs in neuropathic pain. Expert
Opin Emerg Drugs 2007;12:113e126.
14. Devinsky O, Nabbout R, Miller I, et al. Long-term cannabidiol treat-
ment in patients with Dravet syndrome: an open-label extension trial.
Conclusion Epilepsia 2019;60:294e302.
15. Urits I, Borchart M, Hasegawa M, et al. An update of current
In this context, it is clear that the eCB system can be a target to Cannabis-based pharmaceuticals in pain medicine. Pain Ther 2019;
control some debilitating diseases, that is insomnia and hyper- 8:41e51.
somnia, learning and memory, anxiety, cancer and epilepsy. In 16. Machado Rocha FC, Stefano SC, De Cassia Haiek R, et al. Therapeu-
tic use of Cannabis sativa on chemotherapy-induced nausea and vom-
the meantime, we believe marijuana’s derivatives may improve
iting among cancer patients: systematic review and meta-analysis.
the quality of life for many people suffering a terminal disease. Eur J Cancer Care (Engl) 2008;17:431e443.
Although cannabinoids induce dependence, see above, we 17. Fogarty A, Rawstorne P, Prestage G, et al. Marijuana as therapy for
believe in terminal diseases this issue should not be a concern. people living with HIV/AIDS: social and health aspects. AIDS care
In other types of diseases, such as insomnia or anorexia, or pain, 2007;19:295e301.
18. Leussink VI, Husseini L, Warnke C, et al. Symptomatic therapy in
molecules that target the eCB system and that have no abuse po-
multiple sclerosis: the role of cannabinoids in treating spasticity. Ther
tential may prove to be efficacious medical treatments. Adv Neurol Disord 2012;5:255e266.
19. Scherma M, Panlilio LV, Fadda P, et al. Inhibition of ananda-
mide hydrolysis by cyclohexyl carbamic acid 3’-carbamoyl-3-yl
ester (URB597) reverses abuse-related behavioral and neuro-
Acknowledgment
chemical effects of nicotine in rats. J Pharmacol Exp Ther
This work received support from Grant IN215218, IN217918, 2008;327:482e490.
IA205218 from DGAPA-UNAM to OPG, AERC and MMD, 20. Cippitelli A, Cannella N, Braconi S, et al. Increase of brain en-
respectively. The authors are grateful to Edith Monroy for review- docannabinoid anandamide levels by FAAH inhibition and
ing the language of the manuscript. We thank Dr. George Koob, alcohol abuse behaviours in the rat. Psychopharmacology 2008;
Professor, The Scripps Research Institute for his critique of an 198:449e460.
early draft of the manuscript. 21. Merritt LL, Martin BR, Walters C, et al. The endogenous cannabi-
noid system modulates nicotine reward and dependence. J Pharmacol
Exp Ther 2008;326:483e492.
22. Maldonado R, Berrendero F, Ozaita A, et al. Neurochemical basis of
References cannabis addiction. Neurosci 2011;181:1e17.
1. Gaoni Y, Mechoulam R. Isolation, structure, and partial synthesis of 23. Xi ZX, Peng XQ, Li X, et al. Brain cannabinoid CB(2) receptors
an active constituent of hashish. J Am Chem Soc 1964;86: modulate cocaine’s actions in mice. Nat Neurosci 2011;14:
1646e1647. 1160e1166.
524 Prospero-Garcıa et al./ Archives of Medical Research 50 (2019) 518e526

24. Navarrete F, Rodriguez-Arias M, Martin-Garcia E, et al. Role of 45. Di Marzo V, Matias I. Endocannabinoid control of food intake and
CB2 cannabinoid receptors in the rewarding, reinforcing, and energy balance. Nat Neurosci 2005;8:585e589.
physical effects of nicotine. Neuropsychopharmacology 2013;38: 46. Soria-Gomez E, Matias I, Rueda-Orozco PE, et al. Pharmacological
2515e2524. enhancement of the endocannabinoid system in the nucleus accum-
25. American Psychiatric Association. DSM-5: Diagnostic and statistical bens shell stimulates food intake and increases c-Fos expression in
manual of mental disorders. 5th ed Washington: American Psychiat- the hypothalamus. Br J Pharmacol 2007;151:1109e1116.
ric Publishing; 2013. 47. Mendez-Diaz M, Rueda-Orozco PE, Ruiz-Contreras AE, et al. The
26. Haney M, Ward AS, Comer SD, et al. Abstinence symptoms endocannabinoid system modulates the valence of the emotion asso-
following smoked marijuana in humans. Psychopharmacology ciated to food ingestion. Addict Biol 2012;17:725e735.
1999;141:395e404. 48. Kirkham TC, Williams CM, Fezza F, et al. Endocannabinoid levels in
27. Gruber SA, Dahlgren MK, Sagar KA, et al. Worth the wait: effects of rat limbic forebrain and hypothalamus in relation to fasting, feeding
age of onset of marijuana use on white matter and impulsivity. Psy- and satiation: stimulation of eating by 2-arachidonoyl glycerol. Br J
chopharmacology 2014;231:1455e1465. Pharmacol 2002;136:550e557.
28. Lubman DI, Cheetham A, Yucel M. Cannabis and adolescent brain 49. Babor TF, Mendelson JH, Kuehnle J. Marihuana and human physical
development. Pharmacol Ther 2015;148:1e16. activity. Psychopharmacology 1976;50:11e19.
29. Camchong J, Lim KO, Kumra S. Adverse effects of Cannabis on 50. Crippa JA, Zuardi AW, Martin-Santos R, et al. Cannabis and anxiety:
adolescent brain development: a longitudinal study. Cereb Cortex a critical review of the evidence. Hum Psychopharmacol 2009;24:
2017;27:1922e1930. 515e523.
30. Fried P, Watkinson B, James D, et al. Current and former marijuana 51. Prospero-Garcia O, Amancio-Belmont O, Becerril Melendez AL,
use: preliminary findings of a longitudinal study of effects on IQ in et al. Endocannabinoids and sleep. Neurosci Biobehav Rev 2016;
young adults. CMAJ 2002;166:887e891. 71:671e679.
31. Meier MH, Caspi A, Ambler A, et al. Persistent cannabis users show 52. Reyes Prieto NM, Romano Lopez A, Perez Morales M, et al. Olea-
neuropsychological decline from childhood to midlife. Proc Natl mide restores sleep in adult rats that were subjected to maternal sep-
Acad Sci U S A 2012;109:E2657eE2664. aration. Pharmacol Biochem Behav 2012;103:308e312.
32. Cunha PJ, Rosa PG, Ayres Ade M, et al. Cannabis use, cognition and 53. Perez-Morales M, Fajardo-Valdez A, Mendez-Diaz M, et al. 2-
brain structure in first-episode psychosis. Schizophr Res 2013;147: Arachidonoylglycerol into the lateral hypothalamus improves
209e215. reduced sleep in adult rats subjected to maternal separation. Neuro-
33. Foltin RW, Brady JV, Fischman MW. Behavioral analysis of mari- report 2014;25:1437e1441.
juana effects on food intake in humans. Pharmacol Biochem Behav 54. Santucci V, Storme JJ, Soubrie P, et al. Arousal-enhancing properties
1986;25:577e582. of the CB1 cannabinoid receptor antagonist SR 141716A in rats as
34. Foltin RW, Fischman MW, Byrne MF. Effects of smoked marijuana assessed by electroencephalographic spectral and sleep-waking cycle
on food intake and body weight of humans living in a residential lab- analysis. Life Sci 1996;58:PL103ePL110.
oratory. Appetite 1988;11:1e14. 55. Huitron-Resendiz S, Sanchez-Alavez M, Wills DN, et al. Character-
35. Koch JE, Matthews SM. Delta 9-tetrahydrocannabinol stimulates ization of the sleep-wake patterns in mice lacking fatty acid amide
palatable food intake in Lewis rats: effects of peripheral and central hydrolase. Sleep 2004;27:857e865.
administration. Nutr Neurosci 2001;4:179e187. 56. Silvani A, Berteotti C, Bastianini S, et al. Multiple sleep alterations
36. Koch JE. Delta (9)-THC stimulates food intake in Lewis rats: effects in mice lacking cannabinoid type 1 receptors. PLoS One 2014;9:
on chow, high-fat and sweet high-fat diets. Pharmacol Biochem Be- e89432.
hav 2001;68:539e543. 57. Pava MJ, den Hartog CR, Blanco-Centurion C, et al. Endocannabi-
37. Cooper SJ. Endocannabinoids and food consumption: comparisons noid modulation of cortical up-states and NREM sleep. PLoS One
with benzodiazepine and opioid palatability-dependent appetite. 2014;9:e88672.
Eur J Pharmacol 2004;500:37e49. 58. Thomas A, Baillie GL, Phillips AM, et al. Cannabidiol displays un-
38. Van Gaal LF, Rissanen AM, Scheen AJ, et al. Effects of the expectedly high potency as an antagonist of CB1 and CB2 receptor
cannabinoid-1 receptor blocker rimonabant on weight reduction agonists in vitro. Br J Pharmacol 2007;150:613e623.
and cardiovascular risk factors in overweight patients: 1-year experi- 59. Monti JM. Hypnoticlike effects of cannabidiol in the rat. Psychophar-
ence from the RIO-Europe study. Lancet 2005;365:1389e1397. macology 1977;55:263e265.
39. Deveaux V, Cadoudal T, Ichigotani Y, et al. Cannabinoid CB2 recep- 60. Chagas MH, Crippa JA, Zuardi AW, et al. Effects of acute systemic
tor potentiates obesity-associated inflammation, insulin resistance administration of cannabidiol on sleep-wake cycle in rats. J Psycho-
and hepatic steatosis. PLoS One 2009;4:e5844. pharmacol 2013;27:312e316.
40. Agudo J, Martin M, Roca C, et al. Deficiency of CB2 cannabinoid 61. Murillo-Rodriguez E, Millan-Aldaco D, Palomero-Rivero M, et al.
receptor in mice improves insulin sensitivity but increases food Cannabidiol, a constituent of Cannabis sativa, modulates sleep in
intake and obesity with age. Diabetologia 2010;53:2629e2640. rats. FEBS Lett 2006;580:4337e4345.
41. Williams CM, Kirkham TC. Anandamide induces overeating: medi- 62. Murillo-Rodriguez E, Palomero-Rivero M, Millan-Aldaco D, et al.
ation by central cannabinoid (CB1) receptors. Psychopharmacology Administration of URB597, oleoylethanolamide or palmitoylethano-
1999;143:315e317. lamide increases waking and dopamine in rats. PLoS One 2011;6:
42. Martinez-Gonzalez D, Bonilla-Jaime H, Morales-Otal A, et al. Ole- e20766.
amide and anandamide effects on food intake and sexual behavior 63. Zuardi AW, Guimaraes FS, Moreira AC. Effect of cannabidiol on
of rats. Neurosci Lett 2004;364:1e6. plasma prolactin, growth hormone and cortisol in human volunteers.
43. Sanchez-Fuentes A, Marichal-Cancino BA, Mendez-Diaz M, et al. Braz J Med Biol Res 1993;26:213e217.
mGluR1/5 activation in the lateral hypothalamus increases food 64. Nicholson AN, Turner C, Stone BM, et al. Effect of Delta-9-
intake via the endocannabinoid system. Neurosci Lett 2016;631: tetrahydrocannabinol and cannabidiol on nocturnal sleep and early-
104e108. morning behavior in young adults. J Clin Psychopharmacol 2004;
44. Cota D, Marsicano G, Tschop M, et al. The endogenous cannabinoid 24:305e313.
system affects energy balance via central orexigenic drive and pe- 65. Linares IMP, Guimaraes FS, Eckeli A, et al. No acute effects of can-
ripheral lipogenesis. J Endocrinol Invest 2003;112:423e431. nabidiol on the sleep-wake cycle of healthy subjects: a randomized,
Cannabinoids and Medicine 525

double-blind, placebo-controlled, crossover study. Front Pharmacol 86. Colizzi M, Bhattacharyya S. Does Cannabis composition matter? dif-
2018;9:315. ferential effects of delta-9-tetrahydrocannabinol and cannabidiol on
66. Ware MA, Fitzcharles MA, Joseph L, et al. The effects of nabilone on human cognition. Curr Addict Rep 2017;4:62e74.
sleep in fibromyalgia: results of a randomized controlled trial. Anesth 87. Morgan CJ, Schafer G, Freeman TP, et al. Impact of cannabidiol on
Analg 2010;110:604e610. the acute memory and psychotomimetic effects of smoked cannabis:
67. Abrams DI. The therapeutic effects of Cannabis and cannabinoids: an naturalistic study: naturalistic study corrected]. Br J Psychiatry 2010;
update from the National Academies of Sciences, Engineering and 197:285e290.
Medicine report. Eur J Intern Med 2018;49:7e11. 88. Calabrese EJ, Rubio-Casillas A. Biphasic effects of THC in memory
68. Babson KA, Sottile J, Morabito D. Cannabis, Cannabinoids, and and cognition. Eur J Clin Invest 2018;48:e12920.
sleep: a review of the literature. Curr Psychiatry Rep 2017;19:23. 89. Varvel SA, Hamm RJ, Martin BR, et al. Differential effects of delta
69. Nakamura EM, da Silva EA, Concilio GV, et al. Reversible effects of 9-THC on spatial reference and working memory in mice. Psycho-
acute and long-term administration of delta-9-tetrahydrocannabinol pharmacology 2001;157:142e150.
(THC) on memory in the rat. Drug Alcohol Depend 1991;28: 90. Suliman NA, Taib CNM, Moklas MAM, et al. Delta-9-
167e175. Tetrahydrocannabinol ((9)-THC) induce neurogenesis and improve
70. Da S, Takahashi RN. SR 141716A prevents delta cognitive performances of male sprague dawley rats. Neurotox Res
9-tetrahydrocannabinol-induced spatial learning deficit in a Morris- 2018;33:402e411.
type water maze in mice. Prog Neuropsychopharmacol Biol Psychi- 91. Ruiz-Contreras AE, Carrillo-Sanchez K, Ortega-Mora I, et al. Perfor-
atry 2002;26:321e325. mance in working memory and attentional control is associated with
71. Varvel SA, Lichtman AH. Evaluation of CB1 receptor knockout the rs2180619 SNP in the CNR1 gene. Genes Brain Behav 2014;13:
mice in the Morris water maze. J Pharmacol Exp Ther 2002; 173e178.
301:915e924. 92. Ruiz-Contreras AE, Roman-Lopez TV, Caballero-Sanchez U, et al.
72. Kruk-Slomka M, Dzik A, Budzynska B, et al. Endocannabinoid sys- Because difficulty is not the same for everyone: the impact of
tem: the direct and indirect involvement in the memory and learning complexity in working memory is associated with cannabinoid 1 re-
processes-a short review. Mol Neurobiol 2017;54:8332e8347. ceptor genetic variation in young adults. Memory 2017;25:335e343.
73. Lichtman AH, Dimen KR, Martin BR. Systemic or intrahippocampal 93. Heitland I, Klumpers F, Oosting RS, et al. Failure to extinguish fear
cannabinoid administration impairs spatial memory in rats. Psycho- and genetic variability in the human cannabinoid receptor 1. Transl
pharmacology 1995;119:282e290. Psychiatry 2012;2:e162.
74. Jentsch JD, Andrusiak E, Tran A, et al. Delta 9-tetrahydrocannabinol 94. Murillo-Rodriguez E, Sanchez-Alavez M, Navarro L, et al. Ananda-
increases prefrontal cortical catecholaminergic utilization and im- mide modulates sleep and memory in rats. Brain Res 1998;812:
pairs spatial working memory in the rat: blockade of dopaminergic 270e274.
effects with HA966. Neuropsychopharmacology 1997;16:426e432. 95. Marsicano G, Wotjak CT, Azad SC, et al. The endogenous cannabi-
75. Hampson RE, Deadwyler SA. Cannabinoids reveal the necessity of noid system controls extinction of aversive memories. Nature 2002;
hippocampal neural encoding for short-term memory in rats. J Neu- 418:530e534.
rosci 2000;20:8932e8942. 96. Rueda-Orozco PE, Soria-Gomez E, Montes-Rodriguez CJ, et al.
76. Mallet PE, Beninger RJ. The cannabinoid CB1 receptor antagonist A potential function of endocannabinoids in the selection of a
SR141716A attenuates the memory impairment produced by navigation strategy by rats. Psychopharmacology 2008;198:
delta9-tetrahydrocannabinol or anandamide. Psychopharmacology 565e576.
1998;140:11e19. 97. Rammes G, Eder M, Dodt HU, et al. Long-term depression in the ba-
77. Takahashi RN, Pamplona FA, Fernandes MS. The cannabinoid antag- solateral amygdala of the mouse involves the activation of interneu-
onist SR141716A facilitates memory acquisition and consolidation in rons. Neuroscience 2001;107:85e97.
the mouse elevated T-maze. Neurosci Lett 2005;380:270e275. 98. Ganon-Elazar E, Akirav I. Cannabinoids prevent the development of
78. Pamplona FA, Bitencourt RM, Takahashi RN. Short- and long-term behavioral and endocrine alterations in a rat model of intense stress.
effects of cannabinoids on the extinction of contextual fear memory Neuropsychopharmacology 2012;37:456e466.
in rats. Neurobiol Learn Mem 2008;90:290e293. 99. Stern CAJ, da Silva TR, Raymundi AM, et al. Cannabidiol disrupts
79. Dougherty DM, Mathias CW, Dawes MA, et al. Impulsivity, atten- the consolidation of specific and generalized fear memories via dor-
tion, memory, and decision-making among adolescent marijuana sal hippocampus CB1 and CB2 receptors. Neuropharmacology 2017;
users. Psychopharmacology 2013;226:307e319. 125:220e230.
80. Solowij N, Battisti R. The chronic effects of cannabis on memory in 100. Bilkei-Gorzo A, Albayram O, Draffehn A, et al. A chronic low dose
humans: a review. Curr Drug Abuse Rev 2008;1:81e98. of Delta (9)-tetrahydrocannabinol (THC) restores cognitive function
81. Bossong MG, Jansma JM, van Hell HH, et al. Effects of delta9- in old mice. Nat Med 2017;23:782e787.
tetrahydrocannabinol on human working memory function. Biol Psy- 101. Zuurman L, Ippel AE, Moin E, et al. Biomarkers for the effects of
chiatry 2012;71:693e699. cannabis and THC in healthy volunteers. Br J Clin Pharmacol
82. Tervo-Clemmens B, Simmonds D, Calabro FJ, et al. Adolescent 2009;67:5e21.
cannabis use and brain systems supporting adult working memory 102. Pernia-Andrade AJ, Kato A, Witschi R, et al. Spinal endocannabi-
encoding, maintenance, and retrieval. Neuroimage 2018;169: noids and CB1 receptors mediate C-fiber-induced heterosynaptic
496e509. pain sensitization. Science 2009;325:760e764.
83. Pope HG Jr, Gruber AJ, Hudson JI, et al. Neuropsychological perfor- 103. Starowicz K, Przewlocka B. Modulation of neuropathic-pain-related
mance in long-term cannabis users. Arch Gen Psychiatry 2001;58: behaviour by the spinal endocannabinoid/endovanilloid system.
909e915. Philos Trans R Soc Lond B Biol Sci 2012;367:3286e3299.
84. Jager G, Kahn RS, Van Den Brink W, et al. Long-term effects of 104. Grotenhermen F, Muller-Vahl K. The therapeutic potential of
frequent cannabis use on working memory and attention: an fMRI cannabis and cannabinoids. Dtsch Arztebl Int 2012;109:495e501.
study. Psychopharmacology 2006;185:358e368. 105. Lopez-Ortiz M, Herrera-Solis A, Luviano-Jardon A, et al. Chemoen-
85. Smith JL, De Blasio FM, Iredale JM, et al. Verbal learning and mem- zymatic synthesis and cannabinoid activity of a new diazabicyclic
ory in cannabis and alcohol users: an event-related potential investi- amide of phenylacetylricinoleic acid. Bioorg Med Chem Lett 2010;
gation. Front Psychol 2017;8:2129. 20:3231e3234.
526 Prospero-Garcıa et al./ Archives of Medical Research 50 (2019) 518e526

106. Michalski CW, Oti FE, Erkan M, et al. Cannabinoids in pancreatic 119. Perez-Gomez E, Andradas C, Flores JM, et al. The orphan receptor
cancer: correlation with survival and pain. Int J Oncol 2008;122: GPR55 drives skin carcinogenesis and is upregulated in human squa-
742e750. mous cell carcinomas. Oncogene 2013;32:2534e2542.
107. Ramer R, Hinz B. Antitumorigenic targets of cannabinoids - current 120. De Petrocellis L, Melck D, Palmisano A, et al. The endogenous
status and implications. Expert Opin Ther Targets 2016;20: cannabinoid anandamide inhibits human breast cancer cell prolifera-
1219e1235. tion. Proc Natl Acad Sci U S A 1998;95:8375e8380.
108. Tam J, Hinden L, Drori A, et al. The therapeutic potential of targeting 121. Bifulco M, Laezza C, Valenti M, et al. A new strategy to block tumor
the peripheral endocannabinoid/CB1 receptor system. Eur J Intern growth by inhibiting endocannabinoid inactivation. FASEB J 2004;
Med 2018;49:23e29. 18:1606e1608.
109. Nguyen T, Li JX, Thomas BF, et al. Allosteric modulation: an alter- 122. Kwan P, Brodie MJ. Early identification of refractory epilepsy. N
nate approach targeting the Cannabinoid CB1 receptor. Med Res Rev Engl J Med 2000;342:314e319.
2017;37:441e474. 123. Friedman D, Sirven JI. Historical perspective on the medical use of
110. Ignatowska-Jankowska BM, Baillie GL, Kinsey S, et al. A Cannabi- cannabis for epilepsy: ancient times to the 1980s. Epilepsy Behav
noid CB1 receptor-positive allosteric modulator reduces neuropathic 2017;70:298e301.
pain in the mouse with no psychoactive effects. Neuropsychopharma- 124. Russo EB, Jiang HE, Li X, et al. Phytochemical and genetic analyses
cology 2015;40:2948e2959. of ancient cannabis from Central Asia. J Exp Bot 2008;59:
111. Slivicki RA, Xu Z, Kulkarni PM, et al. Positive allosteric modulation 4171e4182.
of cannabinoid receptor type 1 suppresses pathological pain without 125. O0 Shaughnessy WB. On the preparations of the Indian Hemp, or
producing tolerance or dependence. Biol Psychiatry 2018;84: Gunjah (Cannabis Indica), their effects on the animal system in
722e733. health, and their utility in the treatment of tetanus and other convul-
112. Munson AE, Harris LS, Friedman MA, et al. Antineoplastic activity sive diseases. Prov Med J Retrosp Med Sci 1843;5:363e369.
of cannabinoids. J Natl Cancer Inst 1975;55:597e602. 126. Gowers WR. Epilepsy and other chronic convulsive diseases: their
113. Guindon J, Hohmann AG. The endocannabinoid system and cancer: causes, symptoms, and treatment. London: London: Churchill; 1881.
therapeutic implication. Br J Pharmacol 2011;163:1447e1463. 127. Rosenberg EC, Patra PH, Whalley BJ. Therapeutic effects of
114. Malfitano AM, Ciaglia E, Gangemi G, et al. Update on the endocan- cannabinoids in animal models of seizures, epilepsy, epileptogen-
nabinoid system as an anticancer target. Expert Opin Ther Targets esis, and epilepsy-related neuroprotection. Epilepsy Behav 2017;
2011;15:297e308. 70:319e327.
115. Jhawer M, Goel S, Wilson AJ, et al. PIK3CA mutation/PTEN expres- 128. Maa E, Figi P. The case for medical marijuana in epilepsy. Epilepsia
sion status predicts response of colon cancer cells to the epidermal 2014;55:783e786.
growth factor receptor inhibitor cetuximab. Cancer Res 2008;68: 129. Cunha JM, Carlini EA, Pereira AE, et al. Chronic administration of
1953e1961. cannabidiol to healthy volunteers and epileptic patients. Pharma-
116. Buck E, Eyzaguirre A, Barr S, et al. Loss of homotypic cell adhesion cology 1980;21:175e185.
by epithelial-mesenchymal transition or mutation limits sensitivity to 130. Devinsky O, Marsh E, Friedman D, et al. Cannabidiol in patients
epidermal growth factor receptor inhibition. Mol Cancer Ther 2007; with treatment-resistant epilepsy: an open-label interventional trial.
6:532e541. Lancet Neurol 2016;15:270e278.
117. Santoro A, Pisanti S, Grimaldi C, et al. Rimonabant inhibits hu- 131. Devinsky O, Cross JH, Laux L, et al. Trial of Cannabidiol for Drug-
man colon cancer cell growth and reduces the formation of pre- Resistant Seizures in the Dravet Syndrome. N Engl J Med 2017;376:
cancerous lesions in the mouse colon. Int J Cancer 2009;125: 2011e2020.
996e1003. 132. Devinsky O, Patel AD, Thiele EA, et al. Randomized, dose-ranging
118. Ortega A, Garcia-Hernandez VM, Ruiz-Garcia E, et al. safety trial of cannabidiol in Dravet syndrome. Neurology 2018;90:
Comparing the effects of endogenous and synthetic cannabinoid e1204ee1211.
receptor agonists on survival of gastric cancer cells. Life Sci 133. Ali S, Scheffer IE, Sadleir LG. Efficacy of cannabinoids in paediatric
2016;165:56e62. epilepsy. Dev Med Child Neurol 2019;61:13e18.

You might also like