Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Curr Gastroenterol Rep (2015) 17: 9

DOI 10.1007/s11894-015-0429-1

NEUROGASTROENTEROLOGY AND MOTILITY DISORDERS OF THE GASTROINTESTINAL TRACT (S RAO, SECTION EDITOR)

The Role of Cannabinoids in Regulation of Nausea and Vomiting,


and Visceral Pain
Zubair Malik & Daniel Baik & Ron Schey

Published online: 26 February 2015


# Springer Science+Business Media New York 2015

Abstract Marijuana derived from the plant Cannabis sativa Introduction


has been used for the treatment of many gastrointestinal (GI)
disorders, including anorexia, emesis, abdominal pain, diar- The marijuana plant Cannabis sativa is one of the most com-
rhea, and others. However, its psychotropic side effects have monly used illicit drug today with over 16 million users in the
often limited its use. Several cannabinoid receptors, which USA; the largest demographic group is the 18–25 year olds
include the cannabinoid receptor 1 (CB1), CB2, and possibly [1]. The plant contains at least 70 different cannabinoids that
GPR55, have been identified throughout the GI tract. These are a class of diverse chemical compounds of Cannabis that
receptors may play a role in the regulation of food intake, act on cannabinoid receptors located on cells that repress neu-
nausea and emesis, gastric secretion and gastroprotection, GI rotransmitter release in the brain. These receptor proteins in-
motility, ion transport, visceral sensation, intestinal inflamma- clude 1. endocannabinoids, which are produced naturally in
tion, and cell proliferation in the gut. However, the regulation the body by humans and animals and are a part of the
of nausea and vomiting by cannabinoids and the endocannabinoid system that also consists of cannabinoid re-
endocannabinoid system has shed new knowledge in this ceptors and the enzymes that synthesize and degrade the li-
field. Thus far, despite evidence of visceral sensitivity inhibi- gands, 2. phytocannabinoids, which are found in cannabis and
tion in animal models, data in irritable bowel syndrome (IBS) some other plants (Cannabis delta-9-tetrahydrocannabinol
patients is scarce and not supportive. Furthermore, many com- (Δ9-THC) and cannabidiol), and 3. synthetic cannabinoids,
pounds that either act directly at the receptor or increase (or which are manufactured chemically (Table 1) [2].
reduce) ligand availability have the potential to affect other Cannabinoids have been used to treat many health prob-
brain functions and cause side effects. Novel drug targets such lems, including anorexia, emesis, abdominal pain, and diar-
as FAAH and monoacylglycerol lipase (MAGL) inhibitors rhea. Their role in the treatment of gastrointestinal (GI) disor-
appear to be promising in animal models, but more studies ders particularly visceral pain, nausea, and vomiting will be
are necessary to prove their efficiency. The promise of emerg- the focus of this review.
ing drugs that are more selective and peripherally acting sug-
gest that, in the near future, cannabinoids will play a major
role in managing an array of GI diseases. The Cannabinoid Receptors

Cannabinoid receptors are present throughout the GI tract,


Keywords Cannabis . CB1 . CB2 . Endocannabinoids . including the liver, pancreas, stomach, and the small and large
Nausea and vomiting . Visceral pain intestines. In 1990, the first cannabinoid receptor was cloned
by Matsuda et al. and named it as cannabinoid receptor 1
(CB1) [3]. Subsequently, a second receptor (CB2) was iden-
This article is part of the Topical Collection on Neurogastroenterology
and Motility Disorders of the Gastrointestinal Tract tified [4]. The discovery of the endogenous ligands for these
receptors provided the basis for the establishment of the
Z. Malik : D. Baik : R. Schey (*)
endocannabinoid system [3, 5]. The established CB receptors
Section of Gastroenterology, Department of Medicine, Temple
University School of Medicine, Philadelphia, PA, USA show a distinct distribution in the gastrointestinal tract with
e-mail: Ron.schey@tuhs.temple.edu CB1 and CB2 receptors found on macrophages, plasma cells,
9 Page 2 of 9 Curr Gastroenterol Rep (2015) 17: 9

Table 1 Cannabinoid receptor compounds The recently discovered G-protein-coupled receptor,


Agonists GPR55, has been proposed to be the ‘third’ cannabinoid re-
Plant derived ceptor. Although gene expression of GPR55 is evident in the
Δ9-THC Main psychoactive cannabinoid in the marijuana
gut, functional evidence for GPR55 in the gut is lacking. A
plant recent study found that GPR55 activation inhibits neurogenic
Δ8-THC Slightly less potent than Δ9-THC contractions in the gut [14].
11-OH-Δ9-THC Bioactive compound formed when the body Effects of Cannabinoids: Activation of CB1 and CB2
breaks down Δ9-THC receptors may play a role in the regulation of GI function.
Animal derived CB1 receptor activation has been shown to inhibit the
Anandamide 2-AG peristaltic reflex and slow down GI and colonic transit,
THC analogues while endocannabinoids may play a role in transient lower
Dronabinol Nabilone CP 55,940 HU-210 Levonantradol esophageal sphincter relaxations (TLESRs) [15]. The en-
Different chemical structure dogenous cannabinoid system also modulates the nerve
WIN-55,212 Binds to both cannabinoid receptors. growth factor-mediated components of inflammatory re-
sponse [16]. A recent study demonstrated that Δ9-THC
Antagonists (receptor blockers) blocks diclofenac-induced gastric inflammatory damage
SR 141716A Synthetic CB1 antagonist in rats at doses insufficient to cause common cannabinoid
SR 144528 Synthetic CB2 antagonist side effects [17].
Cannabinoid receptor agonists inhibit gastric emptying and
intestinal motility in humans. There is good evidence to sup-
port the role of CB1 receptors in the control of gastrointestinal
sensation, since these receptors have been identified in the
enteric neurons, nerve fibers, and terminals throughout the neuronal circuitry of the transmission of visceral pain. CB2
enteric nervous system [6–9]. CB1 receptors are also localized receptor activation reduces nociception in a variety of preclin-
on epithelial cells, and CB2 receptors are present on immune ical models, including those involved in tactile and thermal
cells [6, 10]. Both receptors are coupled negatively through allodynia, mechanical and thermal hyperalgesia, and writhing
Gi/Go-type G proteins to adenylate cyclase and positively to [18]. Experimental data show that cannabinoid receptor ago-
mitogen-activated protein kinase (MAP kinase), but little is nists have a visceral anti-receptive effect. In general, the phar-
known regarding the exact cellular mechanisms involved after macological effects of cannabis consumption on the GI tract
their activation in the gastrointestinal tract [10]. CB1 receptors include decreased motility and secretion and slowing of
are mainly expressed in the central and peripheral nervous gastric/colonic emptying as well as anti-inflammatory [9].
system, including the enteric nervous system, whereas the The m ajor constituent of cannabis is delta-9-
CB2 re ce pto rs a re s ee n in immu ne cell s [7 , 8 ]. tetrahydrocannabinol (Δ9-THC) which acts as a partial ago-
Endocannabinoid and CB1 receptors have been identified in nist at of both cannabinoid receptors. Its prototypes are
key areas of the GI tract, such as the cholinergic neurons. The nabilone and dronabinol. Dronabinol is marketed as an appe-
endogenous arachidonate-based lipids, anandamide (N- tite stimulant and as an anti-emetic in many countries. One
arachidonoylethanolamide, AEA) and 2- study has shown that it also activates non-CB receptors, and
arachidonoylglycerol (2-AG), are known as this may be responsible for the psychoactive effects mainly
“endocannabinoids” and are physiological ligands for the can- through its actions on the CB1 receptor [19, 20]. Other com-
nabinoid receptors. The endocannabinoid system has been pounds have also been identified that may play a role as phar-
shown to have a role in regulation of food intake, nausea macologic agents, such as cannabidiol, oleoylethanolamide
and emesis, gastric secretion and gastroprotection, GI motility, (OEA), and salvinorin [21]. It has been suggested that these
ion transport, visceral sensation, intestinal inflammation, and other compounds interact in a complex manner to modulate
cell proliferation in the gut [9, 11]. Unlike traditional neuro- pain [22].
transmitters, endogenous cannabinoids are not stored in vesi- The use of cannabinoids is often limited by their side ef-
cles after synthesis but are synthesized on demand. However, fects, which can include tachycardia, hypotension, muscle re-
some evidence suggests that a pool of synthesized laxation, bloodshot eyes, gastroparesis, dizziness, depression,
endocannabinoids (namely 2-AG) may exist without the re- hallucinations, and paranoia, and these often occur at higher
quirement of on-demand synthesis. CB2 receptors are thought doses [23–28].
to serve as an important role in immune function and inflam- Cannabis may exacerbate psychiatric disorders and de-
mation, but it has been shown that they also have a role in crease motivation. Other side effects may include moderate
regulating abnormal motility, modulating intestinal inflamma- driving impairment, gynecomastia, impairment of fetal
tion, and reducing visceral sensitivity and pain [12, 13]. growth, and reduction in fertility and immune function.
Curr Gastroenterol Rep (2015) 17: 9 Page 3 of 9 9

Coadministration of opioids should be done cautiously be- type 1 receptor (TRPV1), which plays a pivotal role in the
cause cannabinoids may increase the synthesis or release of development of inflammatory heat hyperalgesia and visceral
endogenous opioids and may upregulate opioid gene expres- hyperreflexia, can be activated by cannabinoids (Fig. 1) [40•].
sion in the brain and spinal cord and regions that regulate pain In other rat models of acid-induced colitis, CB1 and CB2
sensation, motor activity, and pituitary secretion. A systematic antagonists resulted in an increased visceral hypersensitivity
review of safety studies of medical cannabinoids reported that to rectal distension, whereas CB1 and CB2 agonists reduced
the most common serious adverse event was relapse of mul- basal sensitivity and colitis-induced hypersensitivity [39, 41].
tiple sclerosis, vomiting, and urinary tract infections as well as Bingham et al. showed that a CB2 agonist was effective in the
cyclical vomiting-like syndrome also called cannabinoid relief of visceral pain in rats [42]. Put together, these studies
hyperemesis. Dizziness was the most commonly non-serious provide evidence for a role of cannabis in visceral sensation.
adverse event among people exposed to cannabinoids [29, Few clinical trials have studied the role of cannabinoids in
30]. the control of intestinal motility and sensory function in pa-
tients. Dronabinol decreased fasting colonic motility and in-
creased compliance to colorectal distention in healthy volun-
Cannabinoids and Visceral Pain teers. However, IBS patients who received dronabinol report-
ed increased pain during colorectal distention. It was hypoth-
Visceral pain results from the activation of nociceptors located esized that this phenomenon was possibly due to the central
in the thoracic, pelvic, or abdominal viscera, which are sensi- side effect of increased awareness [43].
tive to distension, ischemia, and inflammation. The pain is Klooker et al. examined the effect of Δ9-THC on rectal
diffuse, often difficult to localize, and usually accompanied sensitivity. They compared the effect of placebo and a Δ9-
by referred pain. Patients with visceral pain often fall into THC agonist (5 and 10 mg) on rectal sensitivity using a rectal
the category of functional GI disorders, with the two major barostat in 10 patients with IBS and 12 healthy volunteers.
disorders being functional dyspepsia and irritable bowel syn- The cannabinoid agonist did not alter baseline rectal percep-
drome (IBS). IBS is the most common disorder seen in the GI tion to distension compared to placebo in both groups. Simi-
outpatient practice [31]. The mechanism of pain in IBS is larly, after sigmoid stimulation, there was no significant dif-
attributed to visceral hypersensitivity or enhanced perception ference compared to placebo. The authors concluded that Δ9-
to distention of colon or rectosigmoid, in approximately 70 % THC agonist does not modify visceral rectal perception and
of patients [32, 33]. It is estimated that there are approximately suggested that CB agonists are not useful for treatment of
3.65 million physician visits annually for IBS with a direct visceral hypersensitivity in IBS patients [44•]. This study is
cost of $1.5 billion and an indirect cost of $20 billion annually hampered by the fact that anxiety was not evaluated, although
[34, 35]. all participants reported central side effects including in-
Cannabis may be used for its analgesic properties, but its creased awareness of the surrounding, light-headedness, and
use is limited by its psychotropic side effects. Cannabinoids sleepiness with the highest dose of Δ9-THC agonist
have been demonstrated to have an analgesic effect at both the (dronabinol 10 mg), whereas no side effects were reported
spinal and peripheral levels in both the GI tract as well as other with placebo. Although BP was stable, the heart rate increased
areas of the body [36, 37, 38•]. in both groups, substantially more in IBS patients. As men-
Intestinal peristalsis is mediated by intrinsic sensory neu- tioned, the central activation of CB1 receptor induces anxiety
rons and interneurons, as well as by excitatory and inhibitory and increased awareness of physical stimuli. All medications
motor neurons. Acetylcholine (ACh) acting through both were given in a single oral administration 30–100 min prior to
muscarinic and nicotinic receptors and tachykinins serve as evaluation. These results are disappointing given the evidence
excitatory neurotransmitters for peristalsis, whereas VIP, nitric in animal models for an analgesic role of cannabinoids. In
oxide (NO), and ATP (or related purine) acts as inhibitory addition, these patients were noted to have central side effects
mediators. Cannabinoids may affect the intrinsic sensory neu- such as drowsiness and heightened awareness [44•].
rons. CB1 receptor immunoreactivity was identified on the Wong et al. demonstrated that dronabinol decreased fasting
intrinsic sensory neurons, ascending neurons, as well as the colonic motility and increased colonic compliance selectively
excitatory motor neurons that project into the longitudinal and in patients with IBS-D or IBS-A. Participants were random-
circular muscles (Fig. 1) [28]. Brusberg et al. showed that CB1 ized to one oral administration of placebo, dronabinol 2.5 mg
receptors, but not CB2, are involved in the modulation of or dronabinol 5 mg, taken at the study center under supervi-
basal visceral sensation in a rodent model of visceral pain sion of a study staff. The effects were influenced by the pres-
induced by colorectal distension [7]. This did not support a ence of genetic polymorphisms in fatty acid amide hydrolase
previous study that demonstrated an anti-nociceptive effect of (FAAH) or CB1. However, pain scores during colorectal dis-
a putative CB2 receptor agonist in a rat model [39]. Another tention were unaffected [45]. Overall, these data suggest that
rat model showed that transient receptor potential vanilloid the endocannabinoid system does play a role in the
9 Page 4 of 9 Curr Gastroenterol Rep (2015) 17: 9

Fig. 1 Sites of action of


cannabinoids in the enteric
nervous system (adapted with the
permission from Aviello G et al.,
Verducci publishers [28])

pathophysiology of IBS in animal models: further studies are models mimicking functional GI disorders. The effects of PF-
needed to determine whether pharmacologic manipulation of 3845 were mediated by endogenous CBs and non-CB lipo-
this system can be clinically beneficial [7, 38•, 39, 40•, 41, 43, philic compounds via classical CB1 and atypical CB recep-
44•, 45]. tors. The anti-nociceptive action of PF-3845 was evaluated on
Recently, we evaluated visceral pain thresholds in patients the basis of behavioral pain models [47•].
with non-GERD-related non-cardiac chest pain (NCCP). Thir-
teen patients were randomized to receive a Δ9-THC agonist Cannabinoids and Nausea and Vomiting
(dronabinol 5 mg bid) or placebo for 4 weeks. An esophageal
balloon distention test (EBDT) was performed by incremental Nausea is an aversive experience that often precedes
balloon distension with water in the mid esophagus. First sen- emesis, which is a forceful expulsion of gastric and up-
sation and maximum tolerance on a 0–4 scale were assessed at per intestinal contents. Nausea and vomiting (N&V) are
baseline and on the last day of treatment (day 28). Chest pain important defense mechanisms that protect the gut from
symptoms and esophageal sensorimotor properties were the ingestion of potentially harmful substance [48]. How-
assessed pre and on day 28 of treatment using questionnaires ever, the sensitivity of this reflex is extremely low and
and a symptom diary. The threshold for the first sensation was triggered with marginal stimuli, leading to improper
significantly higher after treatment compared to pretreatment stimulation in many disease states [48]. Similarly, N&V
in the dronabinol group and non-significant in the placebo are some of the most common adverse effects of medi-
group. In addition, the dronabinol group demonstrated a sig- cations, notoriously associated with many chemothera-
nificant improvement in pain frequency and intensity on day peutic agents [48]. Other common causes of N&V in-
28 when compared to pretreatment. Interestingly, no signifi- clude pregnancy (with the extreme health-compromising
cant adverse effects were noted despite the fact that the med- situation of hyperemesis gravidarum) and motion sick-
ication was given twice daily for a period of 4 weeks [46•]. ness (caused by conflict between the vestibular, visual,
Fichna et al. reported that PF-3845 (selective FAAH inhib- and other proprioceptive systems) [49, 50].
itor) significantly inhibited mouse colonic motility in vitro and The brain centers that control nausea are located in the
in vivo. It reversed hypermotility and reduced pain in mouse forebrain, and the receptors which are located in the floor of
Curr Gastroenterol Rep (2015) 17: 9 Page 5 of 9 9

the fourth ventricle of the brain represent a chemoreceptor Cannabis prevents N&V triggered by many causes [56].
trigger zone that, when stimulated, leads to vomiting. The However, its therapeutic value is overshadowed by its central
chemoreceptor trigger zone has dopamine D2 receptors, sero- nervous system side effects. Recent research has focused on
tonin 5-HT3 receptors, opioid receptors, acetylcholine recep- finding selective ligands for the CB2 receptor or on develop-
tors, and receptors for substance P (Fig. 2). The pathophysi- ing peripherally restricted CB1/CB2 ligands [57]. Many stud-
ology for most cases of emesis are well studied and include a ies have confirmed that cannabinoids block both acute and
trigger (serotonin e.g., 5-HT) released from enterochromaffin delayed emesis, mediated by its effects of the CB1 receptors
cells in the GI epithelium, activating 5-HT3/5-HT4 receptors in the DVC [58–61]. Additionally, the administration of CB1
on vagal primary afferent nerves [51]. These stimulate circuits receptor antagonists in humans has actually evoked nausea
to the dorsal vagal complex (DVC) of the brainstem, which and emesis in multiple studies [62–64].
includes the nucleus of the solitary tract, area postrema, and I n t e r e s t i n g l y, C h o u k e r e t a l . r e v i e w e d b l o o d
dorsal motor nucleus of the vagus. The solitary nucleus acti- endocannabinoid levels in humans undergoing parabolic
vates the motor responses that elicit the characteristic emesis flight maneuvers and noted that those who experienced mo-
[48]. tion sickness had lower levels of anandamide and 2-AG, while
The brain circuitry responsible for evoking nausea is not those who did not experience motion sickness had higher
well understood. A recent study triggered nausea by visual levels of the endocannabinoids [65]. In addition, CB1 receptor
stimulation and performed functional magnetic resonance im- expression was also reduced in those experiencing more
aging simultaneously. This study revealed an elaborate net- symptoms, and the level of arachidonic acid, the downstream
work in the brain activated by nausea, including areas respon- metabolite of endocannabinoids, was significantly increased
sible for interceptive, limbic, somatosensory, and cognitive in patients experiencing N&V upon acceleration than those
processing [52]. who did not [61, 65, 66].
As mentioned previously, CB1 receptors are widely distrib- Conversely, the role of CB2 receptors in preventing N&V
uted in virtually all brain regions, including the DVC involved is ill defined, although one study did show that there are CB2
in emesis and the brain regions described above [53]. The receptors in the DVC of a ferret [61]. CB2 receptors are post-
highest density of CB1 receptors are in the cortex, amygdala, synaptically localized and may regulate neuronal excitability
and basal ganglia, with lower densities in the nucleus accum- by unique mechanisms, as well as through more traditional
bens, ventral tegmental area, and brainstem regions [54]. CB1 cannabinoid signaling. CB2 receptors in the prefrontal cortex
receptors are also located on dopaminergic, noradrenergic, are intracellular and regulate neuronal excitability through
and other transmitter-containing neurons in the brain regions calcium-activated chloride channels and are recently reported
involved in the control of nausea and vomiting [55]. to form functional heteromers with the CB1 receptor [67]. The
distribution of endocannabinoid biosynthesis enzymes needs
further evaluation, though FAAH and monoacylglycerol li-
pase (MAGL) are present in some of these regions, such as
the nucleus accumbens and the amygdala [68].
The CB2 receptor’s role in anti-emesis merits further re-
search because of its lack of psychotropic effects [48]. It is
also important to note that the entire endocannabinoid system
likely communicates with other receptor systems, including
the 5-HT3 and the TRPV1 systems, and their interactions
require further study.
Chemotherapy-induced nausea and vomiting (CINV) is as-
sociated with a significant deterioration in quality of life [51].
CB1-
The orally active, synthetic analogue of Δ9-THC, nabilone,
has been shown to lower the vomiting episodes compared
with D2 receptor antagonists such as domperidone and
D2+ metoclopramide in patients taking moderately toxic chemo-
therapy treatments [69]. However, there was no advantage
NK-1+ over D2 antagonists when given to cancer patients receiving
cisplatin (highly emetogenic agent) chemotherapy. The same
effect was seen with another synthetic analogue of Δ9-THC,
dronabinol [70]. Namisol is a novel oral formulation of pure
5-HT3+ Δ9-THC that has shown promising pharmacokinetic and
Fig. 2 Chemoreceptor trigger zone agonists and antagonists pharmacodynamic characteristics. Variability and t (max) of
9 Page 6 of 9 Curr Gastroenterol Rep (2015) 17: 9

THC plasma concentrations were smaller than reported for activation as well as direct CB2 receptor activation inhibit
studies using oral dronabinol and nabilone [71]. visceral sensitivity and pain in rodents. Thus far, despite these
A recent study evaluated the safety and pharmacokinetic promising results in animal models, data in IBS patients is
profile of Namisol in healthy older subjects and found it to be scarce and not supportive. Furthermore, central side effects
safe and well tolerated by healthy older individuals [72]. Pre- such as drowsiness and heightened awareness were reported
viously, Meiri et al. compared dronabinol alone versus in these studies. The development of new CB2 ligands as well
ondansetron (5-HT3 receptor antagonist) versus a combina- as peripherally acting CB1/CB2 ligands has the potential to
tion in the treatment of CINV. In this study, dronabinol or alleviate symptoms without central nervous system side ef-
ondansetron were similarly effective (although dronabinol fects. Overall, further studies are needed to validate whether
had an advantage in mild-moderately severe nausea), but the pharmacologic manipulation of this system is clinically useful
combination of both was no more effective than either one in treating IBS.
alone [73]. Recently, a non-selective cannabinoid agonist Cannabinoids have a more proven role in N&V, with the
WIN 55 212-2 (WIN) aggravated gastric dysmotility caused recent progress in understanding the regulation of nausea and
by cisplatin but did prevent induced neuropathy in a rat model vomiting by cannabinoids and the endocannabinoid system.
[74]. Another study used cannabidiolic acid in rats, which also However, much research is needed regarding the fact that
showed improvement in nausea-induced behavior and compounds that either act directly at the receptor or increase
vomiting [75]. (or reduce) ligand availability have the potential to effect brain
The current CINV prevention strategy includes a combina- functions besides nausea and vomiting and lead to side effects,
tion of serotonin (5-HT3) receptor antagonists, corticoste- worsen GI motility disorders, and potentially to induce can-
roids, and/or neurokinin-1 receptor antagonists. Current nabinoid hyperemesis syndrome. Novel drug targets such as
guidelines recommend that D2 receptor antagonists be re- FAAH and MAGL inhibitors seem to be promising in animal
served for patients intolerant of or refractory to 5-HT3 recep- models, and more studies are necessary.
tor antagonists, but this study may cause extrapyramidal In conclusion, our review suggests that cannabinoids have
symptoms, which limits the use of these agents. Thus far, a the potential to play a vital role in the modulation of nausea,
clinical trial which compares the effect of cannabinoids versus vomiting, and possibly visceral pain. Although the jury is still
dopamine/serotonin receptor combination has not been done. out on determining the “magic drug,” it seems that with the
A phase II trial compared Sativex (combination of Δ9-THC development of newer ligands, the future appears promising.
and cannabidiol) taken with 5-HT3 receptor antagonists ver-
sus a combination of placebo and 5-HT3 receptor antagonists
Compliance with Ethics Guidelines
in patients with CINV. The addition of Sativex reduced the
incidence of delayed nausea and vomiting and was well toler- Conflict of Interest Zubair Malik, Daniel Baik, and Ron Schey declare
ated. This combination may be useful in managing delayed no conflict of interest.
nausea and vomiting in humans [76].
The “cannabinoid hyperemesis syndrome” (CHS) was first Human and Animal Rights and Informed Consent This article does
described in 2004 [77]. CHS is a syndrome of cyclic episodes not contain any studies with animal subjects performed by any of the
authors. With regard to the authors’ research cited in this paper, all pro-
of nausea and vomiting and abdominal pain, due to chronic cedures were followed in accordance with the ethical standards of the
cannabis usage. Patients adopt a learned behavior of hot baths responsible committee on human experimentation and with the Helsinki
or showers to improve symptoms [78]. Interestingly, standard Declaration of 1975, as revised in 2000 and 2008.
anti-emetics are markedly ineffective in its treatment [77, 79,
80]. The mechanism of this syndrome is entirely unknown,
although some believe a toxic metabolite from the cannabis
plant may be responsible. Another hypothesis is that the high References
exposure to the ligand may lead to the downregulation of
cannabinoid receptors [48]. Papers of particular interest, published recently, have been
highlighted as:
• Of importance
Conclusions

Cannabinoids are increasingly being prescribed for GI condi- 1. United States Department of Health and Human Services.
tions, particularly nausea and vomiting, as an appetite stimu- Substance Abuse and Mental Health Services Administration.
Office of Applied, Studies.
lant and anecdotally for visceral pain. However, treatment of 2. Pacher P, Batkai S, Kunos G. The endocannabinoid system as an
visceral pain has yet to be proven in humans, with some prom- emerging target of pharmacotherapy. Pharmacol Rev. 2006;58(3):
ising early results. It seems that direct or indirect CB1 receptor 389–462.
Curr Gastroenterol Rep (2015) 17: 9 Page 7 of 9 9

3. Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. 22. Booker L, Naidu PS, Razdan RK, Mahadevan A, Lichtman AH.
Structure of a cannabinoid receptor and functional expression of Evaluation of prevalent phytocannabinoids in the acetic acid model
the cloned cDNA. Nature. 1990;346(6284):561–4. doi:10.1038/ of visceral nociception. Drug Alcohol Depend. 2009;105(1–2):42–
346561a0. 7. doi:10.1016/j.drugalcdep.2009.06.009.
4. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of 23. Saez-Cassanelli JL, Fontanella GH, Delgado-Garcia JM, Carrion
a peripheral receptor for cannabinoids. Nature. 1993;365(6441): AM. Functional blockage of the cannabinoid receptor type 1 evokes
61–5. doi:10.1038/365061a0. a kappa-opiate-dependent analgesia. J Neurochem. 2007;103(6):
5. Di Marzo V, Fontana A. Anandamide, an endogenous 2629–39. doi:10.1111/j.1471-4159.2007.05000.x.
cannabinomimetic eicosanoid: ‘killing two birds with one stone’. 24. Gregg JM, Small EW, Moore R, Raft D, Toomey TC. Emotional
Prostaglandins Leukot Essent Fat Acids. 1995;53(1):1–11. response to intravenous delta9tetrahydrocannabinol during oral sur-
6. Wright K, Rooney N, Feeney M, et al. Differential expression of gery. J Oral Surg. 1976;34(4):301–13.
cannabinoid receptors in the human colon: cannabinoids promote 25. Bisogno T, Ligresti A, Di Marzo V. The endocannabinoid signalling
epithelial wound healing. Gastroenterology. 2005;129(2):437–53. system: biochemical aspects. Pharmacol Biochem Behav.
7. Brusberg M, Arvidsson S, Kang D, Larsson H, Lindstrom E, 2005;81(2):224–38.
Martinez V. CB1 receptors mediate the analgesic effects of canna- 26. Battista N, Di Tommaso M, Bari M, Maccarrone M. The
binoids on colorectal distension-induced visceral pain in rodents. J endocannabinoid system: an overview. Front Behav Neurosci.
Neurosci. 2009;29(5):1554–64. doi:10.1523/JNEUROSCI. 5166- 2012;6:9. doi:10.3389/fnbeh.2012.00009.
08.2009. 27. Sugiura T, Kishimoto S, Oka S, Gokoh M. Biochemistry, pharma-
8. Kulkarni-Narla A, Brown DR. Localization of CB1-cannabinoid cology and physiology of 2-arachidonoylglycerol, an endogenous
receptor immunoreactivity in the porcine enteric nervous system. cannabinoid receptor ligand. Prog Lipid Res. 2006;45(5):405–46.
Cell Tissue Res. 2000;302(1):73–80. 28. Aviello G, Romano B, Izzo AA. Cannabinoids and gastrointestinal
9. Izzo AA, Sharkey KA. Cannabinoids and the gut: new develop- motility: animal and human studies. Eur Rev Med Pharmacol Sci.
ments and emerging concepts. Pharmacol Ther. 2010;126(1):21– 2008;12 Suppl 1:81–93.
38. doi:10.1016/j.pharmthera.2009.12.005. 29. Anthony JC, Warner L, Kessler R. Comparative epidemiology of
10. Gruber SA, Rogowska J, Yurgelun-Todd DA. Altered affective re- dependence on tobacco, alcohol, controlled substances, and inhal-
sponse in marijuana smokers: an FMRI study. Drug Alcohol ants: basic findings from the national comorbidity survey. Exp Clin
Depend. 2009;105(1–2):139–53. doi:10.1016/j.drugalcdep.2009. Psychopharmacol. 1994;2:224–68.
06.019. 30. Wang T, Collet JP, Shapiro S, Ware MA. Adverse effects of medical
cannabinoids: a systematic review. CMAJ. 2008;178(13):1669–78.
11. Moreno-Sanz G, Sasso O, Guijarro A, et al. Pharmacological char-
doi:10.1503/cmaj.071178.
acterization of the peripheral FAAH inhibitor URB937 in female
31. Camilleri M. Management of the irritable bowel syndrome.
rodents: interaction with the Abcg2 transporter in the blood-
Gastroenterology. 2001;120(3):652–68.
placenta barrier. Br J Pharmacol. 2012;167(8):1620–8. doi:10.
32. Mertz H, Naliboff B, Munakata J, Niazi N, Mayer EA. Altered
1111/j.1476-5381.2012.02098.x.
rectal perception is a biological marker of patients with irritable
12. Wright KL, Duncan M, Sharkey KA. Cannabinoid CB2 receptors
bowel syndrome. Gastroenterology. 1995;109(1):40–52.
in the gastrointestinal tract: a regulatory system in states of inflam-
33. Posserud I, Syrous A, Lindstrom L, Tack J, Abrahamsson H,
mation. Br J Pharmacol. 2008;153(2):263–70.
Simren M. Altered rectal perception in irritable bowel syndrome
13. Izzo AA. The cannabinoid CB(2) receptor: a good friend in the gut.
is associated with symptom severity. Gastroenterology.
Neurogastroenterol Motil. 2007;19(9):704–8.
2007;133(4):1113–23.
14. Ross GR, Lichtman A, Dewey WL, Akbarali HI. Evidence for the 34. Talley NJ. Functional gastrointestinal disorders as a public health
putative cannabinoid receptor (GPR55)-mediated inhibitory effects problem. Neurogastroenterol Motil. 2008;20 Suppl 1:121–9. doi:
on intestinal contractility in mice. Pharmacology. 2012;90(1–2):55– 10.1111/j.1365-2982.2008.01097.x.
65. doi:10.1159/000339076. 35. Cash B, Sullivan S, Barghout V. Total costs of IBS: employer and
15. Fioramonti J, Bueno L. Role of cannabinoid receptors in the control managed care perspective. Am J Manage Care. 2005;11(1 Suppl):
of gastrointestinal motility and perception. Expert Rev S7–S16.
Gastroenterol Hepatol. 2008;2(3):385–97. doi:10.1586/17474124. 36. Rice AS, Farquhar-Smith WP, Nagy I. Endocannabinoids and pain:
2.3.385. spinal and peripheral analgesia in inflammation and neuropathy.
16. Farquhar-Smith WP, Jaggar SI, Rice AS. Attenuation of nerve Prostaglandins Leukot Essent Fat Acids. 2002;66(2–3):243–56.
growth factor-induced visceral hyperalgesia via cannabinoid doi:10.1054/plef.2001.0362.
CB(1) and CB(2)-like receptors. Pain. 2002;97(1–2):11–21. 37. Farquhar-Smith WP, Rice AS. Administration of endocannabinoids
17. Kinsey SG, Cole EC. Acute delta(9)-tetrahydrocannabinol blocks prevents a referred hyperalgesia associated with inflammation of
gastric hemorrhages induced by the nonsteroidal anti-inflammatory the urinary bladder. Anesthesiology. 2001;94(3):507–13. discus-
drug diclofenac sodium in mice. Eur J Pharmacol. 2013;715(1–3): sion 6A.
111–6. doi:10.1016/j.ejphar.2013.06.001. 38.• Jaggar SI, Hasnie FS, Sellaturay S, Rice AS. The anti-hyperalgesic
18. Beltramo M. Cannabinoid type 2 receptor as a target for chronic actions of the cannabinoid anandamide and the putative CB2 recep-
pain. Mini-Rev Med Chem. 2009;9(1):11–25. tor agonist palmitoylethanolamide in visceral and somatic inflam-
19. Govaerts SJ, Hermans E, Lambert DM. Comparison of cannabinoid matory pain. Pain. 1998;76(1–2):189–99. The first study to demon-
ligands affinities and efficacies in murine tissues and in transfected strate anti-nociceptive effect of a CB2 receptor agonist.
cells expressing human recombinant cannabinoid receptors. Eur J 39. Sanson M, Bueno L, Fioramonti J. Involvement of cannabinoid
Pharm Sci. 2004;23(3):233–43. receptors in inflammatory hypersensitivity to colonic distension in
20. Pertwee RG. Receptors and channels targeted by synthetic canna- rats. Neurogastroenterol Motil. 2006;18(10):949–56.
binoid receptor agonists and antagonists. Curr Med Chem. 40.• Mahmud A, Santha P, Paule CC, Nagy I. Cannabinoid 1 receptor
2010;17(14):1360–81. activation inhibits transient receptor potential vanilloid type 1
21. Di Marzo V, Piscitelli F. Gut feelings about the endocannabinoid receptor-mediated cationic influx into rat cultured primary sensory
system. Neurogastroenterol Motil. 2011;23(5):391–8. doi:10.1111/ neurons. Neuroscience. 2009;162(4):1202–11. doi:10.1016/j.
j.1365-2982.2011.01689.x. neuroscience.2009.05.024. Demonstrated that potential vanilloid
9 Page 8 of 9 Curr Gastroenterol Rep (2015) 17: 9

type 1 receptor (TRPV1), which plays a pivotal role in the transmission. Physiol Rev. 2009;89(1):309–80. doi:10.1152/
development of inflammatory heat hyperalgesia and visceral physrev.00019.2008.
hyper-reflexia, can be activated by cannabinoids. 56. Mechoulam R. Plant cannabinoids: a neglected pharmacological
41. Kikuchi A, Ohashi K, Sugie Y, Sugimoto H, Omura H. treasure trove. Br J Pharmacol. 2005;146(7):913–5.
Pharmacological evaluation of a novel cannabinoid 2 (CB2) ligand, 57. Nevalainen T. Recent development of CB2 selective and peripheral
PF-03550096, in vitro and in vivo by using a rat model of visceral CB1/CB2 cannabinoid receptor ligands. Curr Med Chem.
hypersensitivity. J Pharmacol Sci. 2008;106(2):219–24. 2014;21(2):187–203.
42. Bingham B, Jones PG, Uveges AJ, et al. Species-specific in vitro 58. Darmani NA. Delta(9)-tetrahydrocannabinol and synthetic canna-
pharmacological effects of the cannabinoid receptor 2 (CB2) selec- binoids prevent emesis produced by the cannabinoid CB(1) recep-
tive ligand AM1241 and its resolved enantiomers. Br J Pharmacol. tor antagonist/inverse agonist SR 141716A.
2007;151(7):1061–70. Neuropsychopharmacology. 2001;24(2):198–203.
43. Esfandyari T, Camilleri M, Busciglio I, Burton D, Baxter K, 59. Darmani NA. Delta-9-tetrahydrocannabinol differentially sup-
Zinsmeister AR. Effects of a cannabinoid receptor agonist on co- presses cisplatin-induced emesis and indices of motor function via
lonic motor and sensory functions in humans: a randomized, cannabinoid CB(1) receptors in the least shrew. Pharmacol
placebo-controlled study. Am J Physiol Gastrointest Liver Biochem Behav. 2001;69(1–2):239–49.
Physiol. 2007;293(1):G137–45. 60. Ray AP, Chebolu S, Darmani NA. Receptor-selective agonists in-
44.• Klooker TK, Leliefeld KE, Van Den Wijngaard RM, Boeckxstaens duce emesis and fos expression in the brain and enteric nervous
GE. The cannabinoid receptor agonist delta-9-tetrahydrocannabinol system of the least shrew (Cryptotis parva). Pharmacol Biochem
does not affect visceral sensitivity to rectal distension in healthy Behav. 2009;94(1):211–8. doi:10.1016/j.pbb.2009.08.010.
volunteers and IBS patients. Neurogastroenterol Motil. 61. Van Sickle MD, Duncan M, Kingsley PJ, et al. Identification and
2011;23(1):30–5, e2. doi: 10.1111/j.1365-2982.2010.01587.x. functional characterization of brainstem cannabinoid CB2 recep-
This study concluded that the CB agonists are not a useful drug tors. Science. 2005;310(5746):329–32.
for treatment of visceral hypersensitivity in IBS patients. 62. Despres JP, Ross R, Boka G, Almeras N, Lemieux I, ADAGIO-
45. Wong BS, Camilleri M, Busciglio I, et al. Pharmacogenetic trial of a Lipids Investigators. Effect of rimonabant on the high-triglyceride/
cannabinoid agonist shows reduced fasting colonic motility in pa- low-HDL-cholesterol dyslipidemia, intraabdominal adiposity, and
tients with nonconstipated irritable bowel syndrome. liver fat: the ADAGIO-lipids trial. Arterioscler Thromb Vasc Biol.
Gastroenterology. 2011;141(5):1638–47.e1-7. doi:10.1053/j. 2009;29(3):416–23. doi:10.1161/ATVBAHA.108.176362.
gastro.2011.07.036. 63. Kipnes MS, Hollander P, Fujioka K, et al. A one-year study to
46.• Malik Z, Bayman L, Valestin J, Schey R. Dronabinol increases pain assess the safety and efficacy of the CB1R inverse agonist
threshold in non-cardiac chest pain: A double blind placebo con- taranabant in overweight and obese patients with type 2 diabetes.
trolled trial. 2014. First study to demonstrate that dronabinol in- Diabetes Obes Metab. 2010;12(6):517–31. doi:10.1111/j.1463-
creases NCCP pain threshold using an objective parameter 1326.2009.01188.x.
(EBDT). 64. Pi-Sunyer FX, Aronne LJ, Heshmati HM, Devin J, Rosenstock J,
RIO-North America Study Group. Effect of rimonabant, a
47.• Fichna J, Salaga M, Stuart J, et al. Selective inhibition of FAAH
cannabinoid-1 receptor blocker, on weight and cardiometabolic risk
produces antidiarrheal and antinociceptive effect mediated by
factors in overweight or obese patients: RIO-north america: a ran-
endocannabinoids and cannabinoid-like fatty acid amides.
domized controlled trial. JAMA. 2006;295(7):761–75.
Neurogastroenterol Motil. 2014;26(4):470–81. doi:10.1111/nmo.
65. Chouker A, Kaufmann I, Kreth S, et al. Motion sickness, stress and
12272. One of the first studies evaluating the effect of selective
the endocannabinoid system. PLoS One. 2010;5(5):e10752. doi:10.
FAAH inhibitor on mouse colonic motility and pain.
1371/journal.pone.0010752.
48. Sharkey KA, Darmani NA, Parker LA. Regulation of nausea and
66. Mo FF, Qin HH, Wang XL, et al. Acute hyperglycemia is related to
vomiting by cannabinoids and the endocannabinoid system. Eur J
gastrointestinal symptoms in motion sickness: an experimental
Pharmacol. 2014;722:134–46. doi:10.1016/j.ejphar.2013.09.068.
study. Physiol Behav. 2012;105(2):394–401. doi:10.1016/j.
49. Patil CL, Abrams ET, Steinmetz AR, Young SL. Appetite sensa- physbeh.2011.08.024.
tions and nausea and vomiting in pregnancy: an overview of the 67. Callen L, Moreno E, Barroso-Chinea P, et al. Cannabinoid receptors
explanations. Ecol Food Nutr. 2012;51(5):394–417. doi:10.1080/ CB1 and CB2 form functional heteromers in brain. J Biol Chem.
03670244.2012.696010. 2012;287(25):20851–65. doi:10.1074/jbc.M111.335273.
50. Brainard A, Gresham C. Prevention and treatment of motion sick- 68. Gulyas AI, Cravatt BF, Bracey MH, et al. Segregation of two
ness. Am Fam Physician. 2014;90(1):41–6. endocannabinoid-hydrolyzing enzymes into pre- and postsynaptic
51. Rutkowska M, Gliniak H. The influence of ACEA—a selective compartments in the rat hippocampus, cerebellum and amygdala.
cannabinoid CB1 receptor agonist on whole blood and platelet- Eur J Neurosci. 2004;20(2):441–58. doi:10.1111/j.1460-9568.
poor plasma serotonin concentrations. Pharmazie. 2009;64(9): 2004.03428.x.
598–601. 69. Dalzell AM, Bartlett H, Lilleyman JS. Nabilone: an alternative an-
52. Napadow V, Lee J, Kim J, et al. Brain correlates of phasic autonom- tiemetic for cancer chemotherapy. Arch Dis Child. 1986;61(5):502–
ic response to acupuncture stimulation: an event-related fMRI 5.
study. Hum Brain Mapp. 2013;34(10):2592–606. doi:10.1002/ 70. Lane M, Vogel CL, Ferguson J, et al. Dronabinol and
hbm.22091. prochlorperazine in combination for treatment of cancer
53. Sharkey KA, Cristino L, Oland LD, et al. Arvanil, anandamide and chemotherapy-induced nausea and vomiting. J Pain Symptom
N-arachidonoyl-dopamine (NADA) inhibit emesis through canna- Manag. 1991;6(6):352–9.
binoid CB1 and vanilloid TRPV1 receptors in the ferret. Eur J 71. Klumpers LE, Beumer TL, van Hasselt JG, et al. Novel delta(9)-
Neurosci. 2007;25(9):2773–82. tetrahydrocannabinol formulation Namisol(R) has beneficial phar-
54. Mackie K. Distribution of cannabinoid receptors in the central and macokinetics and promising pharmacodynamic effects. Br J Clin
peripheral nervous system. Handb Exp Pharmacol. 2005;(168): Pharmacol. 2012;74(1):42–53. doi:10.1111/j.1365-2125.2012.
299–325. 04164.x.
55. Kano M, Ohno-Shosaku T, Hashimotodani Y, Uchigashima M, 72. Ahmed AI, van den Elsen GA, Colbers A, et al. Safety and phar-
Watanabe M. Endocannabinoid-mediated control of synaptic macokinetics of oral delta-9-tetrahydrocannabinol in healthy older
Curr Gastroenterol Rep (2015) 17: 9 Page 9 of 9 9

subjects: a randomized controlled trial. Eur chemotherapy-induced nausea and vomiting. Br J Clin
Neuropsychopharmacol. 2014. Pharmacol. 2010;70(5):656–63. doi:10.1111/j.1365-2125.2010.
73. Meiri E, Jhangiani H, Vredenburgh JJ, et al. Efficacy of dronabinol 03743.x.
alone and in combination with ondansetron versus ondansetron 77. Galli JA, Sawaya RA, Friedenberg FK. Cannabinoid
alone for delayed chemotherapy-induced nausea and vomiting. hyperemesis syndrome. Curr Drug Abus Rev. 2011;4(4):
Curr Med Res Opin. 2007;23(3):533–43. doi:10.1185/ 241–9.
030079907X167525. 78. Allen JH, de Moore GM, Heddle R, Twartz JC. Cannabinoid
74. Abalo R, Cabezos PA, Vera G, Lopez-Perez AE, Martin MI. hyperemesis: cyclical hyperemesis in association with chronic can-
Cannabinoids may worsen gastric dysmotility induced by chronic nabis abuse. Gut. 2004;53(11):1566–70.
cisplatin in the rat. Neurogastroenterol Motil. 2013;25(5):373. doi: 79. Nicolson SE, Denysenko L, Mulcare JL, Vito JP, Chabon B.
10.1111/nmo.12073. –82, e292. Cannabinoid hyperemesis syndrome: a case series and review of
75. Bolognini D, Rock EM, Cluny NL, et al. Cannabidiolic acid pre- previous reports. Psychosomatics. 2012;53(3):212–9. doi:10.1016/
vents vomiting in Suncus murinus and nausea-induced behaviour in j.psym.2012.01.003.
rats by enhancing 5-HT1A receptor activation. Br J Pharmacol. 80. Simonetto DA, Oxentenko AS, Herman ML, Szostek JH.
2013;168(6):1456–70. doi:10.1111/bph.12043. Cannabinoid hyperemesis: a case series of 98 patients.
76. Duran M, Perez E, Abanades S, et al. Preliminary efficacy and Mayo Clin Proc. 2012;87(2):114–9. doi:10.1016/j.mayocp.
safety of an oromucosal standardized cannabis extract in 2011.10.005.

You might also like