You are on page 1of 19

Gastroenterology 2020;159:62–80

REVIEWS IN BASIC AND CLINICAL GASTROENTEROLOGY


PERSPECTIVES
REVIEWS AND

AND HEPATOLOGY
Role of Cannabis and Its Derivatives in Gastrointestinal and
Hepatic Disease
Jonathan Gotfried,1 Timna Naftali,2 and Ron Schey3
1
Section of Gastroenterology, Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia,
Pennsylvania; 2Division of Gastroenterology and Hepatology, Meir Medical Center, affiliated with the Sackler School of
Medicine, Tel Aviv University, Tel Aviv, Israel; and 3Division of Gastroenterology/Hepatology Department of Internal Medicine,
University of Florida College of Medicine, Jacksonville, Florida

Medical and recreational cannabis use has increased GPR119.3 Enzymes that synthesize endocannabinoids include
dramatically over the last decade, resulting from main- diacylglycerol lipase, which synthesizes anandamide, and N-
stream cultural acceptance and legalization in several acyl-phosphatidylethanolamine-specific phospholipase D,
countries worldwide. Cannabis and its derivatives affect which synthesizes 2-arachidonoylglycerol. The enzymes fatty
many gastrointestinal processes via the endocannabinoid acid amide hydrolyase (FAAH) and monoacylglycerol lipase
system (ECS). The ECS influences gastrointestinal homeo- degrade endocannabinoids.4 The ECS can be activated by
stasis through anti-inflammatory, anti-nociceptive, and anti- exogenous cannabis or other phytocannabinoids, or synthetic
secretory effects. Some gastrointestinal disorders might compounds.
therefore be treated with cannabinoids. Despite numerous Cannabis affects many gastrointestinal processes
studies in cell lines and animals, few human studies have through its actions on the ECS (Figure 2). Cannabinoid re-
evaluated the therapeutic effects of cannabinoids. Cannabis’ ceptors and their ligands are distributed throughout the
schedule 1 drug status has limited its availability in
human gastrointestinal tract with regional variations in
research; cannabis has been legalized only recently, in some
expression.3 CB1 is expressed in the enteric nervous system
states, for medicinal and/or recreational use. Cannabinoids
in epithelial cells and myenteric and submucosal plexuses,
can alleviate chemotherapy-induced nausea and emesis and
chronic pain. Studies have demonstrated the important and is found adjacent to motoneurons, interneurons, and
roles of the ECS in metabolism, obesity, and nonalcoholic primary afferent neurons. CB2 is frequently expressed by
fatty liver disease and the anti-inflammatory effects of immune cells and the peripheral nervous system.5,6 The ECS
cannabis have been investigated in patients with inflam- maintains gut homeostasis by modulating immune toler-
matory bowel diseases. Despite its potential benefits, un- ance,7 gastrointestinal motility,5 and visceral pain and
desired or even detrimental effects of cannabis can limit its inflammation.8 Activation leads to increased food intake and
use. Side effects such as cannabinoid hyperemesis syndrome other metabolic processes that affect energy balance,
affect some users. We review the ECS and the effects of including lipolysis and glucose metabolism.3,8,9
cannabis and its derivatives on gastrointestinal and hepatic Medical and recreational cannabis use has increased
function in health and disease. nationwide, especially after legalization in several states.
Although cannabis can have beneficial effects in patients
with some disorders, there has also been a commensurate
Keywords: CHS; NAFLD; drug; THC. increase in treatment of cannabis-use disorders.10 It is
therefore important to review cannabis’ salutary and po-

C annabis contains numerous chemically active com-


pounds, including cannabinoids, terpenoids, flavo-
noids, and alkaloids. The most prominent and best
tential harmful side effects.

characterized are D9-tetrahydrocannabinol (THC) and can-


nabidiol (CBD). There are, however, more than 100 active Abbreviations used in this paper: AP, acute pancreatitis; CBD, cannabi-
cannabinoids, each with capacity to modulate the endo- diol; CDAI, Crohn’s Disease Activity Index; CHS, cannabinoid hyperemesis
syndrome; CI, confidence interval; CNR1, cannabinoid receptor 1; CNS,
cannabinoid system (ECS).1 The ECS is a network of canna- central nervous system; CP, chronic pancreatitis; CVS, cyclical vomiting
binoid receptors, their ligands, and regulating synthesizing syndrome; ECS, endocannabinoid system; FAAH, fatty acid amide
and degrading enzymes that function on demand (Figure 1). hydrolyase; GE, gastric emptying; GERD, gastroesophageal reflux dis-
ease; IBD, inflammatory bowel disease; IBS, irritable bowel syndrome;
Ligands include arachidonic acid-derived lipids anandamide IBS-D, irritable bowel syndrome with diarrhea; IL, interleukin; LES, lower
and 2-arachidonoylglycerol, although others exist.2 Corre- esophageal sphincter; LPS, lipopolysaccharide; NAFLD, nonalcoholic fatty
liver disease; STC, slow transit constipation; THC, D9-tetrahydrocannab-
sponding receptors are cannabinoid receptor 1 (CNR1, also inol; WAT, white adipose tissue.
called CB1) and cannabinoid receptor 2 (also called CB2), as
Most current article
well as transient receptor potential cation channel subfamily
© 2020 by the AGA Institute
V member 1, peroxisome proliferator–activated receptor-a, 0016-5085/$36.00
and the orphan G-protein coupled receptors GPR55 and https://doi.org/10.1053/j.gastro.2020.03.087
July 2020 Role of Cannabis and Its Derivatives in Gastrointestinal and Hepatic Disease 63

PERSPECTIVES
REVIEWS AND
Figure 1. ECS. The major
receptors, ligands, and
synthesizing and degrading
enzymes in the ECS. AEA,
arachidonoylethanolamide;
2-AG, 2-
arachidonoylglycerol; DAGL,
diacylglycerol lipase; ER,
endoplasmic reticulum;
MGL, monoacylglycerol
lipase; NAPE, N-arach-
idonoylphosphatidylethanol-
amine. Adapted from Vemuri
and Makriyannis,145 with
permission.

Gastrointestinal Motility known about the roles of CB2 in health, and its effects on
In animal studies, CB1 agonists reduced motility, motility are better studied in inflammatory conditions.
whereas CB1 antagonists had promotility effects.11 CB1 is
found on presynaptic neurons in the myenteric plexus and Esophageal Function
submucosal neurons. Agonists inhibit excitatory cholinergic Few studies have evaluated cannabis’ effects on
neurons, leading to reduced contractile activity subse- esophageal motility and gastroesophageal reflux disease
quently decreasing peristalsis.12 This occurs through a (GERD) pathogenesis. Two studies in humans found lower
dampening effect on the spread of junction potentials by esophageal sphincter (LES) relaxation to be affected by
inhibiting interneuron-mediated neurotransmission.13 cannabinoids; short-term THC administration relaxed LES
Furthermore, CB1 modulates peristaltic reflexes by inhibit- pressures, whereas the CB1 antagonist rimonabant
ing substance P and VIP release.5,12,13 These actions seem to increased postprandial LES pressures.14,15 Conversely, a
occur in a dose-dependent manner, independently from limited retrospective study reported higher prevalence of
pacemaker cells (such as interstitial cells of Cajal).13 Less is hypertensive LES in chronic cannabis users, and further
64 Gotfried et al Gastroenterology Vol. 159, No. 1
PERSPECTIVES
REVIEWS AND

Figure 2. Effects of
cannabis and its de-
rivatives on gastrointes-
tinal organs and functions.
TLESR, transient lower
esophageal sphincter
relaxation.

research is needed to understand its effects on LES gastroparetics using oral dronabinol, potentially from
pressure as it relates to GERD.16 Cannabis might also decreased bioavailability compared to inhaled cannabis.
affect GERD through action on rates of transient LES These findings indicate that dose and cannabis’ effects on
relaxation. pathways beyond GE might contribute to development of
THC administration transiently reduced transient LES gastroparesis.22 Further studies are needed to determine
relaxation frequency and acid reflux episodes, although this benefits in certain subgroups (idiopathic vs diabetes vs
reduction was not statistically significant.14 There are postsurgical) before recommendations for clinical use.
limited data on cannabinoids’ role in functional chest pain.
In a prospective study, 4 weeks’ administration of the CB1
agonist dronabinol increased pain thresholds and reduced Colon Transit
pain intensity and odynophagia, compared to placebo, Cannabinoid administration delays colonic transit as
without significant side effects.17 Cannabis might therefore shown in animal and human studies.20 These studies
improve esophageal function, reduce symptoms of GERD postulate increased ECS tone inhibits cholinergic contrac-
and noncardiac chest pain, but further studies are needed. tility, subsequently decreasing colonic transit.23 Human
studies have evaluated exogenous cannabinoid derivatives
on colonic motility. In a randomized, placebo-controlled
Gastric Emptying and Gastroparesis trial, dronabinol reduced colonic contractile activity in
Gastric emptying (GE) decreases after cannabinoid fasting and postprandial periods.20 Additionally, in a retro-
administration, based on findings from animal and limited spective case series of 6 patients with refractory diarrhea,
human studies, mainly via the effects of CB1 agonists on patients receiving CB1 agonist nabilone experienced
peripheral and central nervous system (CNS) pathways.18 decreased stool frequency and improved weight gain. Only 1
THC and dronabinol delayed GE in 2 placebo-controlled patient experienced significant side effects, which resolved
trials. Patients given THC had delayed GE compared to pa- after nabilone cessation.24 By extension, CB1 antagonists
tients given placebo, especially at 120 minutes, determined would be expected to increase colonic motility, as confirmed
by scintigraphy.19 Similar findings occurred with dronabi- through a meta-analysis demonstrating increased incidence
nol.20 Notably, sex-specific responses in the dronabinol of diarrhea from rimonabant or taranabant in clinical
group were observed; women had longer times of GE and trials.25
men had higher fasting gastric volumes, possibly due to Dysregulation of enzymes that synthesize and degrade
hormonal differences. endocannabinoids, such as FAAH, monoacylglycerol lipase,
Unexpectedly, despite evidence that cannabis delays GE, and diacylglycerol lipase, might contribute to colonic
a recent survey of patients with gastroparesis symptoms motility disorders.26 Inhibiting these enzymes increases
found cannabis use was associated with symptom availability of endocannabinoids, thereby decreasing colon
improvement.21 This was not as pronounced in transit.27 A recent case series evaluated FAAH activity in
July 2020 Role of Cannabis and Its Derivatives in Gastrointestinal and Hepatic Disease 65

patients with slow transit constipation (STC). Compared FAAH and monoacylglycerol lipase had analgesic effects,
with controls, serum samples from patients with STC had reducing pain from inflammation, and increased distension-

PERSPECTIVES
REVIEWS AND
higher amounts of anandamide, 2-arachidonoylglycerol, and induced visceral pain thresholds after FAAH inhibition.34
palmitoylethenolamide (inversely associated with FAAH), The ECS therefore controls pain sensation in physiologic
indicating that low FAAH contributes to STC. Additionally, and inflammatory states.
patients with STC have increased CB1 expression in myen-
teric nerve fibers, indicating increased sensitivity to endo-
cannabinoid action. CB2 appears to have anti-kinetic effects Humans
during intestinal inflammation, observed in patients with There have been few studies of the role of the ECS in
inflammatory bowel disease (IBD), or immune activation. patients with IBS. Small bowel and colonic transit were
Rats with intestinal hypermotility, induced by administra- studied, using scintigraphy and sensation to isobaric
tion of lipopolysaccharide (LPS), had slower motility after distension, in patients with IBS with 2 variants in CNR1 and
CB2R activation.6 subjects without IBS (controls). The researchers found a
Despite these findings, cannabis use was associated with significant association between CNR1 rs806378 and
decreased constipation in a nationwide database survey.28 increased colonic transit in patients with IBS and diarrhea
This discrepancy could be attributed to the survey not (IBS-D).35 There was also an association between these gene
evaluating delivery method (inhaled or ingested) or dose. variants and gas sensation, but not pain, indicating roles for
Additionally, CBD could inhibit CB1, whereby different for- cannabinoid receptors in motility and sensation. ECS mod-
mulations with altered CBD/THC ratios might attenuate ulation by dronabinol was evaluated in 75 patients with
CB1-mediated activities. Overall, data indicate that the ECS different IBS subtypes and polymorphisms in ECS genes.36
affects colonic motor activity and might be targeted for Regardless of IBS subtype, dronabinol decreased fasting
treatment of colonic motility disorders, although further proximal left colonic motility index compared with placebo,
research is needed. with the largest effects in patients with IBS-D or IBS-
alternating. Interestingly, dronabinol did not significantly
alter sensation or tone among subjects. Based on those
Irritable Bowel Syndrome findings, a randomized trial evaluated single nucleotide
IBS involves changes in gastrointestinal motility, polymorphisms CNR1 rs806378 and FAAH rs324420 in
inflammation and immune dysregulation, visceral hyper- patients with IBS-D.37 However, dronabinol did not produce
sensitivity, and possibly dysbiosis.29 Considering the in- statistically significant effects on transit. In persons without
teractions of the ECS with many of these processes, IBS, dronabinol reduced postprandial colonic motility, as
alterations in ECS tone might affect IBS pathogenesis. previously observed in patients with IBS, but the subjects
Visceral hypersensitivity is an important feature of IBS. had increased thresholds for pain and distension.38 So there
Studies of rodents have found direct or indirect activation of seems to be a difference between patients with IBS and
CB1R and likely CB2R, which might inhibit visceral sensi- controls in response to cannabinoids.
tivity and pain.30,31 Accordingly, expression of CB1 de- CB2 might modulate inflammation and pain in patients
creases in stress-induced states and visceral hyperalgesia with IBS. The CB2 ligand PF-03550096 increased the
occurs after administration of CB1 antagonist (WIN 55,212- visceral pain threshold in rats. In a functional, human cell-
2).32,33 CB1 activation also affects other pain pathways based assay, PF-03550096 bound CB2 with a similar affin-
outside the ECS. Low expression of CB1R in the dorsal root ity as in the rat studies.30 CB2 has been evaluated in only 1
ganglion results in increased expression of transient re- animal study, which investigated the effects of the dietary
ceptor potential cation channel subfamily V member 1.32 compounds polydatin and palmithoylethanolamide (struc-
This finding indicates that there are interactions between turally related to anandamide). These compounds have
the ECS and vanilloid system—a pathway that senses synergistic effects on mast cells.39 Patients with IBS given
noxious stimuli and pain, indicating a role for CB1 in pain these compounds for 12 weeks had lower severity of
sensation. abdominal pain compared with placebo. These patients had
The ECS appears to become sensitized during inflam- higher mucosal mast cell counts and higher levels of CB2R
matory or hyperalgesic states through modulation of CB2 expression.
expression. This is important because patients with IBS have A recent study by Dothel et al40 reported increased
been reported to have low-level inflammation. Studies of levels of m-opioid receptor, CB2 messenger RNA and protein,
rats with colitis support this observation; administration of and b-endorphin in colon mucosal biopsies from patients
a CB2 agonist (PF-03,550,096) increased pain thresholds, with IBS compared with asymptomatic individuals; biopsy
measured by colonic-distension, in a dose-dependent tissues from women had higher levels of CB2 messenger
manner.30 Activation of CB2 might down-regulate other in- RNA than men. In contrast, in the asymptomatic control
flammatory mediators, including bradykinin, indicating its group, men had higher expression than women. These
role in pain mediation during inflammation.34 findings indicate that cannabinoids, via CB2, contribute to
In addition to direct action on cannabinoid receptors, immune-related compensatory action in visceral pain.
modification of degrading enzymes may also affect IBS Although cannabinoids could have promising effects, they
symptoms. In mice with visceral inflammation (induced are not used in the management of dysmotility-associated
with acetic acid) and distension-induced pain, inhibitors of conditions—further studies are needed (Table 1).
PERSPECTIVES
REVIEWS AND

Table 1.Studies of Functional Gastrointestinal Disorders

66
Patients

Gotfried et al
Study Study, first
Study design duration n Age and sex Interventions Key results author

IBS-C, IBS-D, IBS-M Single dose 75 Approximately 40 y, Assessed left colonic IBS-D or IBS-mixed, Wong36
Double-blind, randomized, placebo-controlled, 92% women compliance, motility index, dronabinol reduced
parallel-group study tone, and sensation during fasting colonic motility
Dronabinol (2.5 mg or 5 mg) or placebo fasting and after a meal FAAH and CNR1 variants
Analyzed the single nucleotide could influence the
polymorphisms CNR1 effects of this drug on
rs806378, FAAH rs324420, colonic motility
MGLL rs11538700
IBS-D 2d 36 Approximately 41 y, Gastric, small bowel, and Dronabinol had no effect Wong37
Double-blind, randomized, placebo-controlled, 2% men colonic transit on gut transit in patients
parallel-group study Genotype, the single with IBS-D, dronabinol
Dronabinol (2.5 mg or 5 mg) twice daily for 2 d vs nucleotide polymorphisms delayed colonic transit
placebo CNR1 rs806378, FAAH in patients with the CB1
rs324420 rs806378 CT/TT
polymorphism
Healthy volunteers Single dose 52 34 y, 42% men Volonic compliance, motility, Significant increase in Esfandyari38
Double-blind, randomized, placebo-controlled, tone, and sensation 1 h colonic compliance,
parallel-group study before and after a 1000- borderline effect of
Dronabinol (7.5 mg) vs placebo kcal meal relaxation in fasting
colonic tone, inhibition
of postprandial colonic
tone and fasting and
postprandial phasic
pressure
IBS 12 wk 68 40 y, 41.2% men Questionnaires, biopsy, and Did not significantly modify Cremon39
Randomized, double-blind, placebo-controlled trial IBS biologic profile (ELISA, IBS biological profile,
palmithoylethanolamide or polydatin vs placebo immunoblot, including mast cell
immunohistochemistry) count
Reduced severity of
abdominal pain
IBS-C, IBS-D, or IBS-M vs healthy individuals 1 injection 63 Age not available, Left colon mucosal biopsies IBS patients had increased Dothel40
Expression of MOR ligand b-endorphin, CB2R 55% men colonic mucosal
Correlation with sex and symptom perception expression of MOR,

Gastroenterology Vol. 159, No. 1


b-endorphin, and
CB2R, localized
to immune cells

ELISA, enzyme-linked immunsorbent assay; IBS-C, IBS with constipation; IBS-D, IBS with diarrhea; IBS-M, IBS mixed.
July 2020 Role of Cannabis and Its Derivatives in Gastrointestinal and Hepatic Disease 67

Salutary Effects Against Nausea and headaches, lack of hot water symptom relief, and female
predilection. It is important to note some patients with CVS

PERSPECTIVES
REVIEWS AND
Emesis have improvement after self-medication with cannabis.
Cannabinoid receptors are found along emetic pathways Furthermore, abortive anti-emetics used for CVS have been
in peripheral and CNS, involving areas associated with tried in CHS with varying results.52
generation of nausea and emesis, including area postrema In patients with CHS, symptoms are episodic and may
and dorsal vagal complex. Cannabinoid agonists likely sup- cease, classically, after a hot shower. Symptom resolution
press the emetic reflex. Studies of animals have shown that after cannabis cessation strongly supports the diagnosis and
ECS activation down-regulates enterochromaffin release of is recommended first-line therapy. There is no defined time
serotonin and inhibits substance P-induced neurokinin for resolution of symptoms after cessation. By the authors’
pathways, indicating anti-emetic properties.41,42 Enzymes experience, symptoms resolve several days and up to
that regulate the ECS, such as FAAH, diacylglycerol lipase, months after cannabis weaning. It is imperative to caution
and N-acyl-phosphatidylethanolamine-specific phospholi- patients that symptoms may return with re-exposure.
pase D, might also affect CNS processes, although data to Tricyclic antidepressants are the most commonly pre-
support this come from only animal studies.41 scribed long-term treatment in patients requiring medical
Use of cannabis and cannabinoids as anti-emetics has therapy.55 Benzodiazepines, haloperidol, and capsaicin are
been mostly studied in patients with chemotherapy-induced preferred acute treatments. The rationale for capsaicin use
nausea and vomiting. Different formulations and delivery follows similar physiologic response to exposure to hot
methods have been used to treat chemotherapy-induced water relieving symptoms in CHS. Capsaicin affects transient
nausea and vomiting, sometimes with other anti-emetics, receptor potential cation channel subfamily V member 1
with varying results. In a meta-analysis of 28 studies receptors, found adjacent to CB1, and may modulate nausea
(including nabilone [n ¼ 14]; dronabinol [n ¼ 3]; and lev- and emesis through action on the medullary vomiting center
onantradol [n ¼ 4], cannabis extract nabiximols [n ¼ 1], and and gastrointestinal tract, possibly through release of sub-
THC [n ¼ 6]), use of cannabinoids appeared superior to stance P.3,56,57 However, a recent systematic review
placebo and active comparators (alizapride, hydroxyzine, concluded that the quality of data for capsaicin treatment of
metoclopramide, and ondansetron), although the results CHS is low, although the limited availability of alternative
were not statistically significant.43 Importantly, pharmaco- antiemetic therapies and capsaicin’s favorable risk-to-
dynamics and pharmacokinetics of these compounds might benefit profile make it a reasonable, adjunctive treatment
influence their effectiveness—newer formulations might option.58 Clinicians should be vigilant eliciting patient’s
have better outcomes.44,45 Recent oncology guidelines history of cannabis use when evaluating patients with
recommend dronabinol as rescue therapy for episodic emesis and recognize the varying response to
chemotherapy-induced nausea and vomiting, but not THC or available therapies in CHS.
CBD, citing insufficient data and potential side effects.46,47
There have been fewer studies of cannabis as an anti-
emetic agent during pregnancy. Obstetric guidelines Gut Microbiome
discourage cannabis use during pregnancy, citing lack of Cannabis has been reported to modify intestinal micro-
evidence for benefits and safety.48 Nonetheless, a recent biome, so it might be used in treatment of various condi-
phone survey found that many medical dispensaries in tions. For example, in an analysis of a nationwide inpatient
Colorado still recommend cannabis—clinicians with preg- database using ICD-9-CM codes, cannabis use (including
nant patients should be aware of this activity.49 dependent and nondependent use) was associated with a
significant reduction in risk (28%) of Clostridioides difficile
Cannabinoid Hyperemesis Syndrome infection in hospitalized patients compared with nonuse.59
Increased incidence of cannabinoid hyperemesis syn- However, there are few data on its overall effects on the
drome (CHS) has been reported in states after state-wide microbiome, especially because preclinical studies of
legalization of medicinal and recreational cannabis, high- cannabinoid receptor agonists and antagonists have pro-
lighting potential side effects in some users.50,51 CHS likely duced inconsistent results.60–62 Furthermore, due to limited
occurs in long-term users (near daily use for 1 year), most oversight and lack of standardization among dispensaries,
often in adolescent males or young adults.52 Cannabis and there have been reports of medicinal cannabis contaminated
synthetic formulations can cause CHS53 by unknown with bacterial and fungal pathogens, which have raised
mechanisms. Chronic use might down-regulate CB1 in per- concerns about its negative effects on microbiome
sons with specific genetic variants, lowering emetic pathway composition.63
thresholds. Other theories attribute relative potency or Despite barriers and limitations of its study, several
formulation differences of cannabis products (THC/CBD studies of the microbiome and its association with cannabis
ratio).51 Lastly, delayed GE from cannabis use is described are worth noting. The ECS could have an important role in
but the extent this contributes to CHS is unknown. modulating visceral pain sensation in patients with dys-
Patients with CHS present with features similar to biosis, a hallmark feature of some patients with functional
cyclical vomiting syndrome (CVS), so CHS might be a CVS gastrointestinal disorders.64 Ingestion of Lactobacillus aci-
subtype.51,54 However, unlike CHS, CVS is associated with dophilus strains increased endocannabinoid (CB2) and
psychologic comorbidities, anxiety and dysphoria, migraine m-opioid receptor expression in intestinal epithelial cells in a
68 Gotfried et al Gastroenterology Vol. 159, No. 1

rat model of induced colonic hypersensitivity.65 Microbes stimulation, typically at higher doses.74,75 Although cannabis
might therefore potentiate or modify visceral pain pathways use benefit these patients, its effects vary, due to unreliable
PERSPECTIVES
REVIEWS AND

through the ECS and be included in treatment strategies for dosing and pharmacokinetics.76
functional gastrointestinal disorders. In obese individuals, CB1 antagonists have been found to
Studies of mice have shown that the gut microbiota af- promote weight loss, but there were negative side effects. A
fects metabolism through effects on intestinal ECS tone. meta-analysis of randomized trials of rimonabant showed
Dysbiosis resulting from high-fat diet can increase ECS tone, that patients lost an average 4.7 kg compared with placebo
modulating gut permeability and subsequently increasing at 1 year (95% confidence interval [CI], 4.1–5.3 kg; P <
plasma level of LPS, which promotes metabolic disorders .0001).77 However, patients receiving rimonabant had high
and inflammation.66 The proposed endocannabinoid–LPS rates of depression and anxiety and, more concerning, a 1.4-
regulatory loop is likely to be affected by genetic and fold increase in risk of serious adverse events (P ¼ .03;
environmental factors, such as diet; the ECS might link number needed to harm ¼ 59), which included suicidal
dysbiosis with obesity.67 This theory is supported by the thoughts, leading to its withdrawal. Taranabant had similar
increased ratio of Firmicutes to Bacteroidetes observed in effects on weight loss; the highest dose (2 mg, once daily)
mice with diet-induced obesity given THC. These findings resulted in loss of 6.7 kg after 52 weeks. However, similar
indicate that THC might affect the intestinal microbiome and concerns about side effects lead to discontinuation.78,79 To
obesity, but further research is needed.62 avoid side effects, researchers evaluated peripherally
restricted CB1 antagonists.67,80 Compared with rimonabant,
the second-generation CB1 antagonist TM38837 had
Endocannabinoid System in Obesity
decreased CNS penetration, but also lower peripheral ac-
The ECS regulates energy intake and appetite through
tivity.81 Other methods, such as targeting non-CB1 path-
central and peripheral metabolic pathways. ECS activation
ways, are being evaluated, most recently in preclinical and
seems to favor net-positive anabolic processes and energy
phase 2 trials. The peripherally restricted agent tetrahy-
storage. Within the CNS, tight control of metabolism occurs
drocannabivarin significantly decreased fasting plasma
through on demand endocannabinoid production based on
glucose and improved pancreatic b-cell function in subjects
energy needs, increasing and decreasing endocannabinoid
with noninsulin-treated type 2 diabetes and was well tol-
levels in fasting and fed states, respectively.68 The effects of
erated.82–84
the ECS on metabolism might be regulated by retrograde
neuromodulation of presynaptic CB1 in excitatory and
inhibitory pathways, depending on energy needs.69 ECS af-
fects homeostatic pathways in the hypothalamus and Cannabinoid Receptor 2 and Energy Metabolism
brainstem through modification of anorexigenic (such as CB2 is expressed in tissues with high levels of meta-
leptin) and orexigenic hormones (such as ghrelin). Obese bolism, such as liver, pancreas, and adipose tissue, so it
individuals have impaired production of leptin, resulting in might be involved in development of obesity or metabolic
diminished inhibition of endocannabinoid levels, which disorders, although it is less well-characterized than CB1.
might also contribute to insulin resistance.1 The CB2 agonist JWH-133 increases the activity of UCP1, a
The CNS also affects energy intake occurs through mitochondrial protein involved in energy balance.85 CB2
behavior–reward pathways of the mesolimbic system.70 agonist stimulation of UCP1 promotes white adipose tissue
Levels of endocannabinoid increase after ingestion of (WAT) browning, converting WAT to beige adipose (an
palatable food. Consequently, the ECS inhibits GABAergic inducible form of WAT that functions similar to brown ad-
neurons, leading to disinhibition of dopamine and activation ipose tissue). WAT browning stimulates thermogenesis and
of the hedonic drive toward further food consumption.70 caloric expenditure during rest and exercise, indicating that
Concomitant orosensory input activates CB1R enhancing cannabinoids might modulate energy utilization (Figure 3).
olfaction and taste circuits, thereby increasing food The effects are reversed after administration of CB2 antag-
intake—especially sweet food.71 onists, leading to increased food intake.85 These and other
The ECS also modulates peripheral metabolism and in- studies showed that CB2 is involved in energy
sulin sensitivity through actions in digestive organs and homeostasis.86
skeletal muscle. ECS stimulation increases insulin resis-
tance, dyslipidemia, and increased adiposity.1,5 Additional
Cannabis Use in Obese Patients
ECS activation occurs in obese patients from aberrant
Epidemiology studies have reported decreased rates of
plasma and intestinal endocannabinoid signaling leading to
obesity among chronic cannabis users.87,88 Lower expres-
inhibition of gut–brain satiation signaling, ultimately
sion of CB1, either from long-term cannabis use or different
contributing to hyperphagia and weight gain.72,73
phenotypic expression across populations, might account
for this observation.87,89,90 Alternatively, although there is
Cannabinoid Therapy for Obesity overwhelming evidence that CB1 promotes energy meta-
CB1 might be targeted for treatment of weight-related bolism, other ECS components, such as CB2, could have
disorders. Dronabinol increases body mass index values in under-recognized effects on metabolic processes, leading to
patients with cancer-related cachexia or advanced acquired net weight loss. Studies of this pathway are needed and
immunodeficiency syndrome, probably through appetite might lead to therapies (Table 2).
July 2020 Role of Cannabis and Its Derivatives in Gastrointestinal and Hepatic Disease 69

PERSPECTIVES
REVIEWS AND
Figure 3. Effects of cannabinoids on fat metabolism. Activation of CB2 or blockade of CB1 might shift white adipose tissue
(principally for fat storage and hormone secretion) to transitional adipocytes (beige) that, when stimulated, can lead to
development of brown adipose tissue, involved in energy expenditure and thermogenesis.

Nonalcoholic Fatty Liver Disease increases in steatosis after administration of CB1 and CB2
CB1 signaling might affect lipid metabolism, insulin agonists.99 Moreover, CB2 agonists increased expression of
sensitivity, and development of hepatic steatosis.91 In mice, CB1, increasing lipid accumulation in hepatocytes.
activation of CB1 in hepatocytes increases de novo fatty acid Interestingly, exocannabinoids in cannabis have anti-
synthesis and increases expression of lipogenic enzymes, inflammatory effects and prevent NAFLD, by inhibiting cy-
such as fatty acid synthase, leading to lipid accumulation tokines.100 This likely occurs via the antagonist effects of
and steatosis.92 This was confirmed in CB1-knockout mice, CBD on CB2R.98 However, studies of the effects of rimona-
which did not develop hepatic steatosis after placement on a bant in patients with nonalcoholic steatohepatitis hepatic
high-fat diet.93 Studies of patients indicted a role for CB1 in inflammation were discontinued due to safety concerns.101
development of NAFLD. In a randomized trial, patients given Namacizumab, a negative allosteric antibody against CB1
rimonabant for 48 weeks had decreased hepatic steatosis, that is intended to down-regulate the receptor is the first
measured by the ratio of liver to spleen attenuation.94 peripherally restricted biologic agent designed to target the
However, rimonabant was withdrawn due to psychotropic ECS , and currently evaluated in treatment of nonalcoholic
side effects. steatohepatitis.68
Interestingly, chronic use of cannabis can lower body
weight and decrease hepatic steatosis.95 In a post-hoc
analysis of heavy users of cannabis treated for depen- Effects of Endocannabinoid System on Liver
dence, subjects were found to have normal levels of liver Fibrosis in Patients With Chronic Liver Disease
enzymes, which did not correlate with levels of THC or THC The ECS is up-regulated in patients with advanced liver
metabolites.96 In a cross-sectional, population-based study, disease, cirrhosis, nonalcoholic steatohepatitis, alcoholic liver
a lower prevalence of NAFLD was found in cannabis users disease, autoimmune hepatitis, and viral hepatitis.102,103 The
compared with nonusers (adjusted odds ratio, 0.82; 95% CI, ECS might affect progression from fibrosis to cirrhosis in
0.76–0.88; P < .0001).95 Among chronic cannabis users, patients with these conditions. Additionally, CB1 might in-
NAFLD prevalence was 43% lower in dependent users fluence the circulatory sequelae observed in patients with
compared with nondependent users, defined as moderate to chronic liver disease, including portal hypertension.102
severe consumption through ICD-10 coding (adjusted odds Liver biopsies from patients with acute and chronic liver
ratio, 0.57; 95% CI, 0.42–0.77; P < .0001). These findings injury have increased expression of CB1 and CB2.104–106
seem to contradict the physiologic effects of endocannabi- CB1 and CB2 were expressed in liver tissues from all pa-
noids on cannabinoids receptors. One potential reason, tients with hepatitis examined. Furthermore, a significant
proposed by Dibba et al,97 is that THC tolerance, with re- increase in cells positive for both CB1 and CB2 was detected
petitive use, leads to decreased CB1 density and subsequent in stage 3 and stage 4 disease compared to stage 1 and stage
lower CB1 activity overall. Another potential mechanism 2 disease. There was a strong positive association between
includes the entourage effect, whereby other constituents in CB1 expression and a-SMA expression. Moreover, double
cannabis, such as CBD and tetrahydrocannabivarin, immunofluorescence staining for CB1 and a-SMA demon-
decrease CB1 activation, causing decreased hepatic steatosis strated that activated hepatic stellate cells express CB1.107
and inflammation. This theory is supported by the finding However, the exact effects of cannabis and its derivates
that CBD and tetrahydrocannabivarin (at high doses) are are not clear in patients with chronic liver diseases,
antagonists of CB1 and CB2.98 including alcoholic and viral hepatitis, and little is known
The role of CB2 in hepatic steatosis and its relationship about their effects on fibrogenesis. There are conflicting
with CB1 are complex and not fully understood. Human data from studies of animal models as to whether cannabis
hepatocytes incubated with oleic acid had dose-dependent contributes to or protects against fibrosis.102,108 In
PERSPECTIVES
REVIEWS AND

Table 2.Studies of Nonalcoholic Fatty Liver Disease and Obesity

70
Patients

Gotfried et al
Study, first
Study design Study duration n Age and sex Interventions Key results author

Double-blind, randomized, placebo- 104 wk 2,502 Approximately 48 y, Measured body weight, Mean changes in body weight for Aronne78
controlled study 44% men waist circumference, taranabant 2 mg, 4 mg, and 6
Body mass index, 27–43 kg/m2, 51% lipid, and glycemic end mg group were –2.6 kg, –6.6
with metabolic syndrome points kg, –8.1 kg, respectively, at 52
Taranabant 2 mg (n ¼ 414), 4 mg (down- wk
dosed for year 2 to 2 mg) (n ¼ 415), or Mean changes in body weight for
6 mg (down-dosed during year 1 to 2 taranabant 2 mg, 4 mg (down-
mg) (n ¼ 1256) or placebo dosed to 2 mg during year 2),
(n ¼ 417) daily and 6 mg (down-dosed to 2
mg during year 1) were –8.1 kg
at 52 wk with, respectively,
–1.4 kg, –8.1 kg at 52 wk with,
respectively, 6.4 kg, –8.1 kg at
52 wk with, respectively, 7.6
kg at 104 wk, respectively
Multicenter, double-blind randomized 2-wk lifestyle 832 Approximately 49 y, Measured change in body Mean changes in body weight for Proietto79
placebo-controlled study interventions, 66% men weight, waist taranabant 0.5 mg, 1 mg, and
Body mass index, 27–43 kg/m2 followed by 52-wk circumference, lipid end 2 mg, and placebo were 5.4
Taranabant 0.5 mg (n ¼ 207), 1 mg treatment points, and glycemic kg, 5.3 kg, 6.7 kg, and
(n ¼ 208), or 2 mg (n ¼ 417) daily 4-wk follow-up end points 1.7 kg, respectively (P < .001
vs placebo)
Increased gastrointestinal
(nausea, diarrhea) and
psychiatric-related (anger,
irritability) in taranabant group
Meta-analysis of 4 randomized 52 wk 4105 44–55 y Effects on weight after 1 y Significant weight loss 4.7 kg Christensen77
controlled trials, rimonabant 20 mg, 27% men and 73% compared with placebo Increased adverse events (odds
or placebo daily men Evaluation of side effects ratio, 1.4; number needed to
harm, 25); 2.5-fold more likely
to have depressive mood
disorders and anxiety
Evaluation of metabolic parameters 13 wk 62 Approximately 59 y, Measured absolute weight No significant effects on any Jadoon82
(includes NAFLD and weight) 68% men loss primary end points between

Gastroenterology Vol. 159, No. 1


Randomized, double-blind placebo- Measure of liver treatment groups
controlled study triglycerides by Overall safe and tolerated without
Nonpsychotropic cannabinoids: magnetic resonance significant side effects
5 groups: imaging or magnetic Potential influence of THCV on
CBD 100 mg twice daily, resonance glycemic control in patients
THCV 5 mg twice daily, spectroscopy with type 2 diabetes
CBD and THCV 5 mg each twice daily,
CBD 100 mg and 5 mg twice daily,
or placebo
July 2020 Role of Cannabis and Its Derivatives in Gastrointestinal and Hepatic Disease 71

population-level studies, cannabis has been shown to


Study, first possibly be protective against fibrosis progression, likely

PERSPECTIVES
REVIEWS AND
author

Bergholm94
through anti-inflammatory pathways, via CB2, or other
pathways.109 Further research is needed in this topic.
Cannabis and its derivates have been investigated in
other liver disorders, including cholestatic liver disease and
pruritus, autoimmune hepatitis, and hepatic encephalopa-
Weight loss averaged 8.5 ± 1.4 kg

thy—for reviews, see references.110–112

rimonabant and hypocaloric


hypocaloric diet group (P <

placebo; nonsignificant for


rimonabant vs hypocaloric
placebo, and 7.5 ± 0.2 kg
rimonabant, 1.7 ± 1.0 kg

.001 for rimonabant vs

Overall decrease liver fat


comparable between
Key results

Cannabis and Pancreatic Diseases


CB1 and CB2 are expressed in the pancreas and there
has been increased attention to cannabis’ role in acute
diet group pancreatitis (AP) and chronic pancreatitis (CP). AP is char-
acterized by inflammation, which might be affected by
diet)

cannabis use. However, it is not clear whether cannabis


contributes to or attenuates episodes of AP. A recent sys-
tematic review of case reports of AP found cannabis to be a
causative agent for previously classified idiopathic pancre-
atitis.113 A cohort study of 460 patients with a first episode
assessed by magnetic
Absolute weight loss with

of AP found a high prevalence of cannabis use for all etiol-


Interventions

ogies of AP (10%), including in cases labeled idiopathic.114


Change in liver fat

spectroscopy

The authors suggested cannabis may be the inciting


rimonabant

resonance

source in previously labeled idiopathic AP and additional


investigation into its role in AP is warranted. In mice with
cerulein-induced AP, infusion of the CB1 agonist ananda-
mide increased the severity of pancreatitis. CB1 might
activate an inflammatory response in the pancreas by
increasing production of TNF, unlike its anti-TNF effects
Approximately 57 y,

elsewhere in the gastrointestinal tract.115 Conversely, find-


Age and sex

ings from other studies showed that cannabis might protect


32% female

against AP.116 In the largest database study to date, which


Patients

evaluated 10 years of inpatient hospital data, patients with


cannabis exposure (defined using ICD-9-CM) had lower age-
NAFLD, nonalcoholic fatty liver disease; TCHV, tetrahydrocannabivarin.

adjusted disease severity and mortality and fewer compli-


cations (such as acute respiratory distress syndrome and
acute kidney injury) compared to non–cannabis-exposed
n

37

group.117 Cannabis was also reported to reduce risk of


alcohol-induced CP and AP.118 There might be altered CB
Study duration

expression in inflamed tissues, leading to a net anti-


inflammatory effect of cannabis.95 In mice, CB2 agonists
had anti-inflammatory effects, via cytokine inhibition and
decreased oxidative stress.119,120
There is limited evidence of cannabis’ analgesic prop-
48 wk

erties in patients with CP-associated pain. In a randomized


trial of patients with chronic pancreatitis, 1 dose of THC did
not significantly alter pain compared with placebo.121
NAFLD or hepatic steatosis randomized,

rimonabant 20 mg or placebo daily

However, longer-term use might improve pain control. In


double-blind, placebo-controlled,

a small, retrospective cohort study, cannabis use for 34–297


weeks (based on state registry data) decreased levels of
opiate requirement for CP pain, indicating a role for pain
control.122 The visceral inflammatory process in pancreatitis
Table 2. Continued

is likely associated with activation of the ECS. There is a


basis for testing the therapeutic value of cannabinoids as
Study design

supplements to conventional analgesics, as well as their


anti-inflammatory effects. Conflicting findings might be the
results of differences in dosing or delivery methods; further
research is needed to better understand cannabis’ effects on
pancreatitis.
72 Gotfried et al Gastroenterology Vol. 159, No. 1
PERSPECTIVES
REVIEWS AND

Figure 4. The ECS in


Crohn’s disease and ul-
cerative colitis.
Figure shows receptors
and synthesizing and
degrading enzymes and
relative levels reported
from studies of IBD. AEA,
arachidonoylethanolamide;
2AG, 2-arachidonoylglycerol;
DAGL, diacylglycerol
lipase; MGLL, mono-
acylglycerol lipase;
NAPE-PLD, N-arach-
idonoylphosphatidyletha-
nolamine phospholipase D;
PEA, palmitoylethanolamide.

There have been only 2 studies of the effects of more likely to develop severe disease, associated with fis-
cannabidiol in patients with gastrointestinal disorders tulas and extra-intestinal manifestations, and patients with
(IBD and NAFLD)—each produced equivocal re- ulcerative colitis homozygous for that mutation have earlier
sults.123,124 CBD affects enzymes involved in drug meta- onset of disease.128
bolism, including CYP450. Many CBD products are not There have been many studies of the anti-inflammatory
supervised by the US Food and Drug Administration and effects of cannabinoids in various tissues—for reviews, see
little is known about interactions of other drugs with Ambrose et al.129 Synthetic and endogenous cannabinoids
CBD, so caution is advised for patients who consume inhibit LPS-induced release of TNF from rat microglial
these products. cells.131 Exposure of the mast cell line RBL2H3 to cannabi-
noid altered transcription of many genes; CB2 agonists
activated extracellular signal-regulated kinase, AKT, and a
Inflammatory Bowel Disease subset of AKT targets.130
The ECS can modulate IBD pathogenesis, based on cor- Mice with knockout of CB1 and/or CB2 developed more
relations between cannabinoid receptor genotypes and IBD severe colitis after administration of dinitrobenzene sulfonic
characteristics (Figure 4). The CB2 complementary DNA acid or dextran sulfate sodium than wild-type mice.131 In
188–189 GG/GG polymorphism is associated with a 2-fold mice with dinitrobenzene sulfonic acid–induced induced
reduction in endocannabinoid-induced inhibition of T-cell colitis, CBD reduced colon injury, expression of inducible
proliferation.125 The CB2 R63 variant was significantly nitric oxide synthase (but not cyclooxygenase-2), and
associated with IBD, particularly with Crohn’s disease.126 changes in interleukin (IL)1beta, IL10, and endocannabi-
The CB1 p.Thr453Thr polymorphism appears to modulate noids associated with 2,4,6-dinitrobenzene sulfonic acid
susceptibility to ulcerative colitis and Crohn’s disease administration. So CBD reduces colitis in mice.132 In rats,
phenotype.127 Patients with Crohn’s disease who are ho- THC and CBD each reduced myeloperoxidase activity,
mozygous for the FAAH p.Pro129Thr polymorphism were reduced colonic motility (measured in vitro), and protected
Table 3.Studies of Inflammatory Bowel Disease

July 2020
Patients
Study, first
Study design Study duration n Age and sex Interventions Key results author

Crohn’s disease double-blind, 8 wk treatment, 2 wk 21 Approximately 40 y Assessed patients with Primary end point of Naftali142
randomized, placebo-controlled after therapy and 62% men CDAI scores >200 induction of remission
smoked cannabis flowers without response to was not achieved
11.5 mg or placebo, twice daily steroids, 10 of 11 had significant
immunomodulators, or clinical and steroid free
anti-TNF agents benefit
Decrease in CDAI score by
more than 100 patients,
P ¼ .028
Crohn’s disease; double-blind, 8 wk treatment and 2 22 Approximately 39 y Assessed patients with No significant reduction in Naftali123
randomized, placebo-controlled; wk after therapy and 50% men moderately active CD CDAI concluded that
CBD oil (10 mg) vs placebo twice and assessed CBD was safe but not
daily nonresponders to beneficial for patients
steroids, immune- with Crohn’s disease

Role of Cannabis and Its Derivatives in Gastrointestinal and Hepatic Disease


modulators, or anti-TNF
agents
Ulcerative colitis; double-blind, 10 wk 60 (39 Approximately 45 y Assessed remission (by Primary end point not Irving143
randomized, placebo-controlled, completed and 79% men Mayo score) after 10 wk achieved, no significant
parallel group trial; the study) effect on rate of
CBD-rich botanical extract (50–250 remission, according to
mg) and 0.7% THC or placebo, per-protocol analysis of
twice daily total or partial Mayo
scores indicated effects
of CBD-THC group (P ¼
.068 vs P ¼ .038 in
placebo group)
Ulcerative colitis; randomized, 8 wk 28 33 y and 52% men Assessed response to THC THC-rich cannabis is safe Naftali146
placebo-controlled trial; inhaled in patients with and led to clinical and
cannabis (cigarettes) moderately severe endoscopic
0.5 g cannabis (11.5 mg THC) vs ulcerative colitis by improvement
placebo (extracted THC), 2 disease activity index Decrease in CDAI scores of
cigarettes daily and Mayo score approximately 6
patients and decrease
in Mayo score of
approximately 1 patient

73
REVIEWS AND
PERSPECTIVES
74 Gotfried et al Gastroenterology Vol. 159, No. 1

cholinergic nerves. The effects of these 2 phytocannabinoids reviews of randomized trials of patients with Crohn’s dis-
were additive.133 In mice with dinitrobenzene sulfonic acid– ease (3 studies)140 and ulcerative colitis (2 studies).141 In
PERSPECTIVES
REVIEWS AND

induced colitis, cannabigerol, a non-psychotropic cannabis- the first trial, 21 patients with Crohn’s Disease Activity In-
derived cannabinoid, reduced colon weight to length ratios, dex (CDAI) scores >200 were randomly assigned to groups
myeloperoxidase activity, and levels of inducible nitric oxide given either cannabis flowers containing THC or placebo.
synthase; increased superoxide dismutase activity; and Complete remission (CDAI score <150) was achieved by 5
normalized levels of IL1B, IL10, and interferon gamma.134 of 11 subjects in the cannabis group (45%) and 1 of 10
For reviews of these studies, see Couch et al.135 subjects in the placebo group (10%; P ¼ .43), not achieving
the primary end point of the study. However, clinical re-
sponses (decrease in CDAI score by >100 points) were
Cannabis Use by Patients With observed in 10 of 11 subjects in the cannabis group (90%;
Inflammatory Bowel Disease from a mean 330 ± 105 to 152 ± 109) and 4 of 10 in the
Despite improvements in IBD therapy, many patients do placebo group (40%; from a mean 373 ± 94 to 306 ± 143;
not respond to treatment and some have turned to alter- P ¼ .028). Patients reported improved appetite and sleep
native therapies, including cannabis. Data from a nationwide without significant adverse events.142
health survey support this, showing increased use of Due to the lack of central effect and the anti-
cannabis among patients with IBD compared to participants inflammatory activity observed in animal models, CBD is
without IBD.136 an attractive treatment option. A randomized trial evaluated
Several observational studies have shown improvement the effects of low-dose CBD in 19 patients with CDAI scores
in both symptoms and clinical outcomes with cannabis use. >200 who were randomly assigned to groups given 10 mg
In a retrospective, observational study of the effects of oral CBD or twice daily placebo for 8 weeks. Mean CDAI
cannabis on disease activity, use of medication, need for scores before CBD consumption were 337 ± 108 and 308 ±
surgery, and hospitalization were examined in 30 patients 96 in the CBD and placebo groups, respectively. After 8
with Crohn’s disease. A significant improvement was weeks of treatment, CDAI scores were 220 ± 122 and 216 ±
observed in 21 of 30 patients, with reduction in the average 121 in the CBD and placebo groups, respectively (not
Harvey Bradshaw index from 14 ± 6.7 to 7.0 ± 4.7 (P < significantly different). The negative result of this study
.001). The need for other medication was reduced signifi- could be attributed to inefficacy of CBD in patients with
cantly.137 Observational data from a national inpatient Crohn’s disease or inadequate dosing.123
sample of patients with Crohn’s disease–related complica- A randomized trial of 50 patients with active Crohn’s
tions compared 615 cannabis users and nonusers hospital- disease compared the effects of cannabis oil (15% CBD and
ized from 2012 through 2014. Cannabis users were 4% THC) vs placebo for 8 weeks. At study completion, the
significantly less likely to have had active fistulizing disease mean CDAI scores were 118.6 in the cannabis oil group vs
and intra-abdominal abscess, blood product transfusion, 212.6 in the placebo group (P < .05). Quality of life scores
colectomy, and parenteral nutrition requirement.138 Lastly, were 96.3 in the cannabis oil group vs 79.9 in the placebo
a survey of 313 patients with IBD investigating motives, group (P < .05).140
patterns of use, and subjective beneficial and adverse effects In a study of patients with ulcerative colitis, participants
of cannabis use found that cannabis had been used by were randomly assigned to groups that received CBD-rich
17.6% of respondents, who reported reduced abdominal botanical extract (also containing THC) for 10 weeks or pla-
pain (83.9%), abdominal cramping (76.8%), joint pain cebo capsules. At the end of the study, the proportion of
(48.2%), and diarrhea (28.6%). Interestingly, despite patients in remission did not differ significantly between the
improvement in these subject symptoms, patients with CBD (28%) and placebo (26%) groups. However, many pa-
Crohn’s disease, cannabis use for longer than 6 months for tients were intolerant to the CBD capsules, so 40% of patients
symptoms was associated with requirement for surgery. It in this group had a protocol deviation. Per-protocol analysis
is not clear whether cannabis use increases risk for surgery revealed that patients in the CBD group had significant re-
or if cannabis is used more frequently in patients with se- ductions in total and partial Mayo scores (P ¼ .068 and P ¼
vere, perhaps fibrostenotic, disease.139 .038, respectively), indicating clinical and endoscopic
Although there are data to support cannabis use for improvement. Quality of life also improved in patients given
subjective symptoms in patients with IBD, these were from CBD. Although this study did not reach its primary end point,
observational studies, and results should be interpreted the findings indicate some beneficial effects of cannabis for
with caution. Although patients might have subjective patients with ulcerative colitis.143 A separate study compared
improvement in their symptoms, there is no evidence for the effects of cannabis cigarettes (23 mg THC/day) vs pla-
reductions in disease progression or severity. cebo in 30 patients with active ulcerative colitis. This study
found that after 8 weeks, mean disease activity index scores
in patients taking cannabis was 4 compared with 8 for pa-
Trials of Cannabis Use for Inflammatory tients in the placebo group (P ¼ .001).140
Bowel Disease Olorinab (APD371), a peripherally restricted, selective
Despite abundant preclinical evidence that cannabinoid agonist of CB2, reduced pain scores in patients with Crohn’s
drugs can reduce intestinal inflammation, few clinical disease and has a favorable safety profile, specifically
studies have been completed. There were 2 Cochrane without CNS effects.144 Studies are planned to evaluate its
July 2020 Role of Cannabis and Its Derivatives in Gastrointestinal and Hepatic Disease 75

effects on chronic abdominal pain in patients with Crohn’s 9. Chanda D, Neumann D, Glatz JFC. The endocannabi-
disease. There have been no studies of the ability of noid system: overview of an emerging multi-faceted

PERSPECTIVES
REVIEWS AND
cannabis to maintain remission in patients with Crohn’s therapeutic target. Prostaglandins Leukot Essent Fat
disease or ulcerative colitis (Table 3). Acids 2019;140:51–56.
10. Gubatan J, Staller K, Barshop K, et al. Cannabis abuse is
increasing and associated with increased emergency
Future Directions department utilization in gastroenterology patients. Dig
The ECS helps maintain gastrointestinal homeostasis and Dis Sci 2016;61:1844–1852.
might be manipulated therapeutically. There is abundant 11. Izzo AA, Mascolo N, Pinto L, et al. The role of cannabi-
evidence for the anti-inflammatory and anti-nociceptive ef- noid receptors in intestinal motility, defaecation and
fects of cannabinoids. Many patients with gastrointestinal diarrhoea in rats. Eur J Pharmacol 1999;384:37–42.
conditions could benefit from cannabis or cannabinoids. 12. Yuece B, Sibaev A, Broedl UC, et al. Cannabinoid type 1
Findings from several small studies support use of cannabis receptor modulates intestinal propulsion by an attenua-
or cannabinoids in patients with IBD. Interestingly, results tion of intestinal motor responses within the myenteric
from epidemiologic studies contradict results from animal part of the peristaltic reflex. Neurogastroenterol Motil
2007;19:744–753.
and human studies of cannabis and cannabinoids—especially
their effects on obesity and fatty liver, relief from symptoms 13. Sibaev A, Yüce B, Kemmer M, et al. Cannabinoid-1
(CB1) receptors regulate colonic propulsion by acting at
of gastroparesis, and IBS. These discordant results indicate
motor neurons within the ascending motor pathways in
the complicated pathways and interactions of the ECS with
mouse colon. Am J Physiol Liver Physiol 2009;
other organ systems. The ECS is far more complex than we
296:G119–G128.
appreciate, and most studies have evaluated CB1 and CB2
14. Beaumont H, Jensen J, Carlsson A, et al. Effect of
only. Targeting individual receptors is beneficial, but future
D9-tetrahydrocannabinol, a cannabinoid receptor
studies, such as those evaluating synthesizing and degrading agonist, on the triggering of transient lower oesophageal
enzymes and their effects on endocannabinoids, as well as sphincter relaxations in dogs and humans. Br J Phar-
new, peripherally restricted agents, could lead to new ther- macol 2009;156:153–162.
apeutic avenues for cannabis and its derivatives in treatment 15. Scarpellini E, Blondeau K, Boecxstaens V, et al. Effect of
of gastrointestinal disease. Importantly, cannabinoids are far rimonabant on oesophageal motor function in man.
from the panacea that some claim—additional investigation Aliment Pharmacol Ther 2011;33:730–737.
is needed to further our understanding of their sometimes 16. Parikh M, Sookal S, Ahmad A. Cannabis use in patients
detrimental effects. presenting to a gastroenterology clinic: associations with
symptoms, endoscopy findings, and esophageal
References manometry. Gastrointest Disord 2019;1:301–307.
1. Mechoulam R, Hanus LO, Pertwee R, etal. Early phyto- 17. Malik Z, Bayman L, Valestin J, et al. Dronabinol
cannabinoid chemistry to endocannabinoids and increases pain threshold in patients with functional chest
beyond. Nat Rev Neurosci 2014;15:757–764. pain: a pilot double-blind placebo-controlled trial. Dis
2. Hourani W, Alexander SPH. Cannabinoid ligands, re- Esophagus 2017;30:1–8.
ceptors and enzymes: pharmacological tools and ther- 18. Izzo AA, Pinto L, Borrelli F, et al. Central and peripheral
apeutic potential. Brain Neurosci Adv 2018;2: cannabinoid modulation of gastrointestinal transit in
2398212818783908. physiological states or during the diarrhoea induced by
3. Izzo AA, Sharkey KA. Cannabinoids and the gut: new croton oil. Br J Pharmacol 2000;129:1627–1632.
developments and emerging concepts. Pharmacol Ther 19. McCallum RW, Soykan I, Sridhar KR, et al. Delta-9-
2010;126:21–38. tetrahydrocannabinol delays the gastric emptying of
4. Hillard CJ. Circulating endocannabinoids: from whence solid food in humans: a double-blind, randomized study.
do they come and where are they going? Neuro- Aliment Pharmacol Ther 1999;13:77–80.
psychopharmacology 2018;43:155. 20. Esfandyari T, Camilleri M, Ferber I, et al. Effect of a
5. Aviello G, Romano B, Izzo AA. Cannabinoids and cannabinoid agonist on gastrointestinal transit and
gastrointestinal motility: animal and human studies. Eur postprandial satiation in healthy human subjects: a
Rev Med Pharmacol Sci 2008;12(Suppl 1):81–93. randomized, placebo-controlled study. Neuro-
6. Wright KL, Duncan M, Sharkey KA. Cannabinoid CB2 gastroenterol Motil 2006;18:831–838.
receptors in the gastrointestinal tract: a regulatory sys- 21. Jehangir A, Parkman HP. Cannabinoid use in patients
tem in states of inflammation. Br J Pharmacol 2008; with gastroparesis and related disorders: prevalence and
153:263–270. benefit. Am J Gastroenterol 2019;114:945–953.
7. Acharya N, Penukonda S, Shcheglova T, et al. Endo- 22. McCallum RW, Bashashati M. Cannabis for gastro-
cannabinoid system acts as a regulator of immune paresis: hype or hope? Am J Gastroenterol 2019;
homeostasis in the gut. Proc Natl Acad Sci 2017;114:5005– 114:865–866.
5010. 23. Smid SD, Bjorklund CK, Svensson KM, et al. The
8. Di Marzo V, Piscitelli F. Gut feelings about the endo- endocannabinoids anandamide and
cannabinoid system. Neurogastroenterol Motil 2011; 2-arachidonoylglycerol inhibit cholinergic contractility in
23:391–398. the human colon. Eur J Pharmacol 2007;575:168–176.
76 Gotfried et al Gastroenterology Vol. 159, No. 1

24. Pellesi L, Verga MC, De Maria N, et al. Nabilone syndrome-diarrhea. Neurogastroenterol Motil 2012;24.
administration in refractory chronic diarrhea: a case se- 358–e169.
PERSPECTIVES
REVIEWS AND

ries. BMC Gastroenterol 2019;19:105. 38. Esfandyari T, Camilleri M, Busciglio I, et al. Effects of a
25. Fabisiak A, Wlodarczyk M, Fabisiak N, et al. Gastroin- cannabinoid receptor agonist on colonic motor and
testinal adverse events of cannabinoid 1 receptor in- sensory functions in hu4mans: a randomized, placebo-
verse agonists suggest their potential use in irritable controlled study. Am J Physiol Liver Physiol 2007;
bowel syndrome with constipation: a systematic review 293:G137–G145.
and meta-analysis. J Gastrointestin Liver Dis 2019; 39. Cremon C, Stanghellini V, Barbaro MR, et al. Rando-
28:473–481. mised clinical trial: the analgesic properties of dietary
26. Zhang S, Wang W, Su P, et al. Decreased enteric fatty supplementation with palmitoylethanolamide and poly-
acid amide hydrolase activity is associated with colonic datin in irritable bowel syndrome. Aliment Pharmacol
inertia in slow transit constipation. J Gastroenterol Ther 2017;45:909–922.
Hepatol 2014;29:276–283. 40. Dothel G, Chang L, Shih W, et al. m-opioid receptor,
27. Wasilewski A, Misicka A, Sacharczuk M, et al. Modulation b-endorphin, and cannabinoid receptor-2 are increased
of the endocannabinoid system by the fatty acid amide in the colonic mucosa of irritable bowel syndrome pa-
hydrolase, monoacylglycerol and diacylglycerol lipase tients. Neurogastroenterol Motil 2019e13688.
inhibitors as an attractive target for secretory diarrhoea 41. Sharkey KA, Darmani NA, Parker LA. Regulation of
therapy. J Physiol Pharmacol 2017;68:591–596. nausea and vomiting by cannabinoids and the endo-
28. Adejumo AC, Flanagan R, Kuo B, et al. Relationship cannabinoid system. Eur J Pharmacol 2014;722:134–146.
between recreational marijuana use and bowel function 42. Darmani NA, Belkacemi L, Zhong W. D9-THC and
in a nationwide cohort study. Am J Gastroenterol 2019; related cannabinoids suppress substance P-induced
114:1894–1903. neurokinin NK1-receptor-mediated vomiting via activa-
29. Storr MA, Yüce B, Andrews CN, Sharkey KA. The role of tion of cannabinoid CB1 receptor. Eur J Pharmacol
the endocannabinoid system in the pathophysiology and 2019:172806.
treatment of irritable bowel syndrome. Neuro- 43. Whiting PF, Wolff RF, Deshpande S, et al. Cannabinoids
gastroenterol Motil 2008;20:857–868. for medical use: a systematic review and meta-analysis.
30. Kikuchi A, Ohashi K, Sugie Y, et al. Pharmacological Jama 2015;313:2456–2473.
evaluation of a novel cannabinoid 2 (CB2) ligand, PF- 44. Badowski ME. A review of oral cannabinoids and
03550096, in vitro and in vivo by using a rat model of medical marijuana for the treatment of chemotherapy-
visceral hypersensitivity. J Pharmacol Sci 2008;106:219– induced nausea and vomiting: a focus on pharmacoki-
224. netic variability and pharmacodynamics. Cancer
31. Fioramonti J, Bueno L. Role of cannabinoid receptors Chemother Pharmacol 2017;80:441–449.
in the control of gastrointestinal motility and percep- 45. Mersiades A, Tognela A, Haber PS, et al. 1805TiP Pilot
tion. Expert Rev Gastroenterol Hepatol 2008;2:385– and definitive randomised double-blind placebo-
397. controlled trials evaluating an oral cannabinoid-rich
32. Hong S, Zheng G, Wu X, et al. Corticosterone mediates THC/CBD cannabis extract for secondary prevention of
reciprocal changes in CB 1 and TRPV1 receptors in chemotherapy-induced nausea and vomiting (CINV).
primary sensory neurons in the chronically stressed rat. Ann Oncol 2018;29(Suppl 8).
Gastroenterology 2011;140:627–637. 46. Roila F, Molassiotis A, Herrstedt J, et al. 2016 MASCC
33. Hong S, Fan J, Kemmerer ES, et al. Reciprocal changes and ESMO guideline update for the prevention of
in vanilloid (TRPV1) and endocannabinoid (CB1) re- chemotherapy-and radiotherapy-induced nausea and
ceptors contribute to visceral hyperalgesia in the water vomiting and of nausea and vomiting in advanced can-
avoidance stressed rat. Gut 2009;58:202–210. cer patients. Ann Oncol 2016;27(Suppl 5):v119–v133.
34. Sakin YS, Dogrul A, Ilkaya F, et al. The effect of FAAH, 47. Hesketh PJ, Kris MG, Basch E, et al. Antiemetics:
MAGL, and dual FAAH/MAGL inhibition on inflamma- American Society of Clinical Oncology clinical practice
tory and colorectal distension-induced visceral pain guideline update. J Clin Oncol 2017;35:3240–3261.
models in Rodents. Neurogastroenterol Motil 2015; 48. Committee on Obstetric Practice. Committee opinion
27:936–944. no. 722: marijuana use during pregnancy and lactation.
35. Camilleri M, Kolar GJ, Vazquez-Roque MI, et al. Obstet Gynecol 2017;130(4):e205.
Cannabinoid receptor 1 gene and irritable bowel syn- 49. Dickson B, Mansfield C, Guiahi M, et al. Recommenda-
drome: phenotype and quantitative traits. Am J Physiol tions from cannabis dispensaries about first-trimester
Liver Physiol 2013;304:G553–G560. cannabis use. Obstet Gynecol 2018;131:1031–1038.
36. Wong BS, Camilleri M, Busciglio I, et al. Pharmacogenetic 50. Kim HS, Monte AA. Colorado cannabis legalization and
trial of a cannabinoid agonist shows reduced fasting its effect on emergency care. Ann Emerg Med 2016;
colonic motility in patients with nonconstipated irritable 68:71.
bowel syndrome. Gastroenterology 2011;141:1638–1647. 51. Venkatesan T, Levinthal DJ, Li BUK, et al. Role of chronic
37. Wong BS, Camilleri M, Eckert D, et al. Randomized cannabis use: Cyclic vomiting syndrome vs cannabinoid
pharmacodynamic and pharmacogenetic trial of dro- hyperemesis syndrome. Neurogastroenterol Motil 2019;
nabinol effects on colon transit in irritable bowel 31:e13606.
July 2020 Role of Cannabis and Its Derivatives in Gastrointestinal and Hepatic Disease 77

52. Galli JA, Andari Sawaya R, Friedenberg FK. Cannabinoid 70. Ceccarini J, Weltens N, Ly HG, et al. Association be-
hyperemesis syndrome. Curr Drug Abuse Rev 2011; tween cerebral cannabinoid 1 receptor availability and

PERSPECTIVES
REVIEWS AND
4:241–249. body mass index in patients with food intake disorders
53. Bick BL, Szostek JH, Mangan TF. Synthetic cannabinoid and healthy subjects: a [18 F] MK-9470 PET study.
leading to cannabinoid hyperemesis syndrome. Mayo Transl Psychiatry 2016;6(7):e853.
Clin Proc 2014;89:1168–1169. 71. Yoshida R, Ohkuri T, Jyotaki M, et al. Endocannabinoids
54. Abell TL, Adams KA, Boles RG, et al. Cyclic vomiting selectively enhance sweet taste. Proc Natl Acad Sci
syndrome in adults. Neurogastroenterol Motil 2008; 2010;107:935–939.
20:269–284. 72. Argueta DA, Perez PA, Makriyannis A, et al. Cannabinoid
55. Richards JR, Gordon BK, Danielson AR, et al. Pharma- CB1 receptors inhibit gut-brain satiation signaling in
cologic treatment of cannabinoid hyperemesis syn- diet-induced obesity. Front Physiol 2019;10:704.
drome: a systematic review. Pharmacother J Hum 73. Argueta DA, DiPatrizio NV. Peripheral endocannabinoid
Pharmacol Drug Ther 2017;37:725–734. signaling controls hyperphagia in western diet-induced
56. Ross RA. Anandamide and vanilloid TRPV1 receptors. Br obesity. Physiol Behav 2017;171:32–39.
J Pharmacol 2003;140:790–801. 74. Badowski ME, Yanful PK. Dronabinol oral solution in the
57. Yang F, Zheng J. Understand spiciness: mechanism of management of anorexia and weight loss in AIDS and
TRPV1 channel activation by capsaicin. Protein Cell cancer. Ther Clin Risk Manag 2018;14:643.
2017;8:169–177. 75. Wang J, Wang Y, Tong M, et al. Medical cannabinoids
58. McConachie SM, Caputo RA, Wilhelm SM, et al. Efficacy for cancer cachexia: a systematic review and meta-
of capsaicin for the treatment of cannabinoid hyper- analysis. Biomed Res Int 2019;2019.
emesis syndrome: a systematic review. Ann Pharmac- 76. Reuter SE, Martin JH. Pharmacokinetics of cannabis in
other 2019:1060028019852601. cancer cachexia-anorexia syndrome. Clin Pharmacoki-
59. Adejumo AC, Bukong TN. Cannabis use and risk of net 2016;55:807–812.
Clostridioides difficile infection: analysis of 59,824 hos- 77. Christensen R, Kristensen PK, Bartels EM, et al. Efficacy
pitalizations. Anaerobe 2019:102095. and safety of the weight-loss drug rimonabant: a meta-
60. Mehrpouya-Bahrami P, Chitrala KN, Ganewatta MS, analysis of randomised trials. Lancet 2007;
et al. Blockade of CB1 cannabinoid receptor alters gut 370(9600):1706–1713.
microbiota and attenuates inflammation and diet- 78. Aronne LJ, Tonstad S, Moreno M, et al. A clinical trial
induced obesity. Sci Rep 2017;7:15645. assessing the safety and efficacy of taranabant, a CB1R
61. Schneeberger M, Everard A, Gómez-Valadés AG, et al. inverse agonist, in obese and overweight patients: a
Akkermansia muciniphila inversely correlates with the high-dose study. Int J Obes 2010;34:919.
onset of inflammation, altered adipose tissue meta- 79. Proietto J, Rissanen A, Harp JB, et al. A clinical trial
bolism and metabolic disorders during obesity in mice. assessing the safety and efficacy of the CB1R inverse
Sci Rep 2015;5:16643. agonist taranabant in obese and overweight patients:
62. Cluny NL, Keenan CM, Reimer RA, et al. Prevention of low-dose study. Int J Obes 2010;34:1243–1254.
diet-induced obesity effects on body weight and 80. Amato G, Khan NS, Maitra R. A patent update on
gut microbiota in mice treated chronically with cannabinoid receptor 1 antagonists (2015-2018). Expert
D9-tetrahydrocannabinol. PLoS One 2015;10(12): Opin Ther Pat 2019;29:261–269.
e0144270. 81. Takano A, Gulyás B, Varnäs K, et al. Low brain CB1
63. Thompson GR, Tuscano JM, Dennis M, et al. receptor occupancy by a second generation CB1 re-
A microbiome assessment of medical marijuana. Clin ceptor antagonist TM38837 in comparison with rimo-
Microbiol Infect 2017;23:269–270. nabant in nonhuman primates: a PET study. Synapse
64. Drossman DA. Functional gastrointestinal disorders: 2014;68:89–97.
history, pathophysiology, clinical features, and Rome IV. 82. Jadoon KA, Ratcliffe SH, Barrett DA, et al. Efficacy and
Gastroenterology 2016;150:1262–1279. safety of cannabidiol and tetrahydrocannabivarin on
65. Rousseaux C, Thuru X, Gelot A, et al. Lactobacillus ac- glycemic and lipid parameters in patients with type 2
idophilus modulates intestinal pain and induces opioid diabetes: a randomized, double-blind, placebo-
and cannabinoid receptors. Nat Med 2007;13:35. controlled, parallel group pilot study. Diabetes Care
66. Muccioli GG, Naslain D, Bäckhed F, et al. The endo- 2016;39:1777–1786.
cannabinoid system links gut microbiota to adipo- 83. Silvestri C, Paris D, Martella A, et al. Two non-
genesis. Mol Syst Biol 2010;6(1). psychoactive cannabinoids reduce intracellular lipid
67. Di Marzo V. New approaches and challenges to targeting levels and inhibit hepatosteatosis. J Hepatol 2015;
the endocannabinoid system. Nat Rev Drug Discov 62:1382–1390.
2018;17:623–639. 84. Klumpers LE, Fridberg M, de Kam ML, et al. Peripheral
68. Gatta-Cherifi B, Cota D. New insights on the role of the selectivity of the novel cannabinoid receptor antagonist
endocannabinoid system in the regulation of energy TM 38837 in healthy subjects. Br J Clin Pharmacol 2013;
balance. Int J Obes 2016;40:210. 76:846–857.
69. Bellocchio L, Lafenêtre P, Cannich A, et al. Bimodal 85. Verty ANA, Stefanidis A, McAinch AJ, et al. Anti-obesity
control of stimulated food intake by the endocannabi- effect of the CB2 receptor agonist JWH-015 in diet-
noid system. Nat Neurosci 2010;13:281. induced obese mice. PLoS One 2015;10(11):e0140592.
78 Gotfried et al Gastroenterology Vol. 159, No. 1

86. Rossi F, Bellini G, Luongo L, et al. Cannabinoid receptor 100. Braunersreuther V, Viviani GL, Mach F, et al. Role of
2 as antiobesity target: Inflammation, fat storage, and cytokines and chemokines in non-alcoholic fatty liver
PERSPECTIVES
REVIEWS AND

browning modulation. J Clin Endocrinol Metab 2016; disease. World J Gastroenterol 2012;18:727–735.
101:3469–3478. 101. Chavez-Tapia NC, Tellez-Avila FI, Bedogni G, et al.
87. Clark TM, Jones JM, Hall AG, et al. Theoretical expla- Systematic review and meta-analysis on the adverse
nation for reduced body mass index and obesity rates in events of rimonabant treatment: considerations for its
cannabis users. Cannabis Cannabinoid Res 2018;3:259– potential use in hepatology. BMC Gastroenterol 2009;
271. 9:75.
88. Le Strat Y, Le Foll B. Obesity and cannabis use: results 102. Baldassarre M, Giannone FA, Napoli L, et al. The
from 2 representative national surveys. Am J Epidemiol endocannabinoid system in advanced liver cirrhosis:
2011;174:929–933. pathophysiological implication and future perspectives.
89. D’Souza DC, Cortes-Briones JA, Ranganathan M, et al. Liver Int 2013;33:1298–1308.
Rapid changes in cannabinoid 1 receptor availability in 103. Basu PP, Aloysius MM, Shah NJ, Brown RS. Review
cannabis-dependent male subjects after abstinence article: The endocannabinoid system in liver disease, a
from cannabis. Biol psychiatry Cogn Neurosci neuro- potential therapeutic target. Aliment Pharmacol Ther
imaging 2016;1:60–67. 2014;39:790–801.
90. Benzinou M, Chevre J-C, Ward KJ, et al. Endocannabi- 104. Auguet T, Berlanga A, Guiu-Jurado E, et al. Endo-
noid receptor 1 gene variations increase risk for cannabinoid receptors gene expression in morbidly
obesity and modulate body mass index in European obese women with nonalcoholic fatty liver disease.
populations. Hum Mol Genet 2008;17:1916– Biomed Res Int 2014;2014:502542.
1921. 105. Mendez-Sanchez N, Zamora-Valdes D, Pichardo-
91. Tam J. Role of the endocannabinoid system in hepatic Bahena R, et al. Endocannabinoid receptor CB2 in
lipogenesis. In: Ntambi JM, ed. Hepatic De Novo Lipo- nonalcoholic fatty liver disease. Liver Int 2007;27:215–
genesis and Regulation of Metabolism. Berlin: Springer: 219.
2016:131–142. 106. González-Mariscal I, Krzysik-Walker SM, Doyle ME, et al.
92. Osei-Hyiaman D, DePetrillo M, Pacher P, et al. Human CB1 receptor isoforms, present in hepatocytes
Endocannabinoid activation at hepatic CB 1 receptors and b-cells, are involved in regulating metabolism. Sci
stimulates fatty acid synthesis and contributes to Rep 2016;6:33302.
diet-induced obesity. J Clin Invest 2005;115:1298– 107. Dai E, Zhang L, Ye L, et al. Hepatic expression of
1305. cannabinoid receptors CB1 and CB2 correlate with
93. Osei-Hyiaman D, Liu J, Zhou L, et al. Hepatic CB 1 re- fibrogenesis in patients with chronic hepatitis B. Int J
ceptor is required for development of diet-induced Infect Dis 2017;59:124–130.
steatosis, dyslipidemia, and insulin and leptin resis- 108. Patsenker E, Stoll M, Millonig G, et al. Cannabinoid re-
tance in mice. J Clin Invest 2008;118:3160–3169. ceptor type I modulates alcohol-induced liver fibrosis.
94. Bergholm R, Sevastianova K, Santos A, et al. CB 1 Mol Med 2011;17:1285–1294.
blockade-induced weight loss over 48 weeks decreases 109. Farooqui MT, Khan MA, Cholankeril G, et al. Marijuana is
liver fat in proportion to weight loss in humans. Int J not associated with progression of hepatic fibrosis in
Obes 2013;37:699–703. liver disease: a systematic review and meta-analysis. Eur
95. Adejumo AC, Alliu S, Ajayi TO, et al. Cannabis use is J Gastroenterol Hepatol 2019;31:149.
associated with reduced prevalence of non-alcoholic 110. Avraham Y, Israeli E, Gabbay E, et al. Endocannabinoids
fatty liver disease: A cross-sectional study. PLoS One affect neurological and cognitive function in
2017;12:e0176416. thioacetamide-induced hepatic encephalopathy in mice.
96. Bonnet U, Canbay A, Specka M, et al. Long-term heavy Neurobiol Dis 2006;21:237–245.
recreational cannabis use and serum delta-9- 111. Hegde VL, Hegde S, Cravatt BF, et al. Attenuation of
tetrahydrocannabinol levels are not associated with an experimental autoimmune hepatitis by exogenous and
impaired liver function in cannabis dependents. endogenous cannabinoids: involvement of regulatory T
J Psychoactive Drugs 2018;50:355–360. cells. Mol Pharmacol 2008;74:20–33.
97. Dibba P, Li AA, Cholankeril G, et al. Potential mecha- 112. Neff GW, O’Brien CB, Reddy KR, et al. Preliminary
nisms influencing the inverse relationship observation with dronabinol in patients with intractable
between cannabis and nonalcoholic fatty liver disease: a pruritus secondary to cholestatic liver disease. Am J
commentary. Nutr Metab Insights 2019;12: Gastroenterol 2002;97:2117.
1178638819847480. 113. Barkin JA, Nemeth Z, Saluja AK, et al. Cannabis-induced
98. Pertwee RG. The diverse CB1 and CB2 receptor phar- acute pancreatitis: a systematic review. Pancreas 2017;
macology of three plant cannabinoids: D9-tetrahydro- 46:1035–1038.
cannabinol, cannabidiol and D9-tetrahydrocannabivarin. 114. Simons-Linares CR, Barkin JA, Wang Y, et al. Is there an
Br J Pharmacol 2008;153:199–215. effect of cannabis consumption on acute pancreatitis?
99. De Gottardi A, Spahr L, Ravier-Dall’Antonia F, et al. Dig Dis Sci 2018;63:2786–2791.
Cannabinoid receptor 1 and 2 agonists increase lipid 115. Dembinski A, Warzecha Z, Ceranowicz P, et al. Canna-
accumulation in hepatocytes. Liver Int 2010;30:1482– binoids in acute gastric damage and pancreatitis.
1489. J Physiol Pharmacol 2006;57(Suppl 5):137–154.
July 2020 Role of Cannabis and Its Derivatives in Gastrointestinal and Hepatic Disease 79

116. Michalski CW, Laukert T, Sauliunaite D, et al. Cannabi- potential for inflammatory bowel disease? J Crohn’s
noids ameliorate pain and reduce disease pathology in Colitis 2019;13:525–535.

PERSPECTIVES
REVIEWS AND
cerulein-induced acute pancreatitis. Gastroenterology 130. Samson M-T, Small-Howard A, Shimoda LMN, et al.
2007;132:1968–1978. Differential roles of CB1 and CB2 cannabinoid
117. Simons-Linares CR, Barkin JA, Jang S, et al. The impact receptors in mast cells. J Immunol 2003;170:4953–
of cannabis consumption on mortality, morbidity, and 4962.
cost in acute pancreatitis patients in the United States: a 131. Engel MA, Kellermann CA, Burnat G, et al. Mice lacking
10-Year analysis of the national inpatient sample. cannabinoid CB1-, CB2-receptors or both receptors
Pancreas 2019;48:850–855. show increased susceptibility to trinitrobenzene sulfonic
118. Adejumo AC, Akanbi O, Adejumo KL, et al. Reduced risk acid (TNBS)-induced colitis. J Physiol Pharmacol 2010;
of alcohol-induced pancreatitis with cannabis use. 61:89–97.
Alcohol Clin Exp Res 2019;43:277–286. 132. Borrelli F, Aviello G, Romano B, et al. Cannabidiol, a safe
119. Michler T, Storr M, Kramer J, et al. Activation of and non-psychotropic ingredient of the marijuana plant
cannabinoid receptor 2 reduces inflammation in acute Cannabis sativa, is protective in a murine model of colitis.
experimental pancreatitis via intra-acinar activation of J Mol Med 2009;87:1111–1121.
p38 and MK2-dependent mechanisms. Am J Physiol 133. Jamontt JM, Molleman A, Pertwee RG, et al. The effects
Liver Physiol 2012;304:G181–G192. of Delta-tetrahydrocannabinol and cannabidiol alone
120. Parlar A, Arslan SO, Dog  an MF, et al. The exogenous and in combination on damage, inflammation and in vitro
administration of CB2 specific agonist, GW405833, in- motility disturbances in rat colitis. Br J Pharmacol 2010;
hibits inflammation by reducing cytokine production 160:712–723.
and oxidative stress. Exp Ther Med 2018;16:4900– 134. Borrelli F, Fasolino I, Romano B, et al. Beneficial effect of
4908. the non-psychotropic plant cannabinoid cannabigerol on
121. de Vries M, Van Rijckevorsel DCM, Vissers KCP, et al. experimental inflammatory bowel disease. Biochem
Single dose delta-9-tetrahydrocannabinol in chronic Pharmacol 2013;85:1306–1316.
pancreatitis patients: analgesic efficacy, pharmacoki- 135. Couch DG, Maudslay H, Doleman B, et al. The use of
netics and tolerability. Br J Clin Pharmacol 2016;81:525– cannabinoids in colitis: a systematic review and meta-
537. analysis. Inflamm Bowel Dis 2018;24:680–697.
122. Barlowe TS, Koliani-Pace JL, Smith KD, et al. Effects of 136. Weiss A, Friedenberg F. Patterns of cannabis use in
medical cannabis on use of opioids and hospital visits by patients with inflammatory bowel disease: a popula-
patients with painful chronic pancreatitis. Clin Gastro- tion based analysis. Drug Alcohol Depend 2015;
enterol Hepatol 2019;17:2608–2609.e1. 156:84–89.
123. Naftali T, Mechulam R, Marii A, et al. Low-dose canna- 137. Naftali T, Lev LB, Yablecovitch D, et al. Treatment of
bidiol is safe but not effective in the treatment for Crohn’s disease with cannabis: an observational study.
Crohn’s disease, a randomized controlled trial. Dig Dis Isr Med Assoc J 2011;13:455–458.
Sci 2017;62:1615–1620. 138. Mbachi C, Attar B, Wang Y, et al. Association Between
124. NCT01284634, Study to Evaluate the Effect of cannabis use and complications related to Crohn’s
GWP42003 on Liver Fat Levels in Participants With Fatty disease: a retrospective cohort study. Dig Dis Sci
Liver Disease. Available at: https://clinicaltrials.gov/ct2/ 2019:1–6.
show/results/NCT01284634?cond¼cannabidiol&rank¼89. 139. Storr M, Devlin S, Kaplan GG, et al. Cannabis use pro-
Accessed July 28, 2019. vides symptom relief in patients with inflammatory bowel
125. Sipe JC. Reduced endocannabinoid immune modulation disease but is associated with worse disease prognosis
by a common cannabinoid 2 (CB2) receptor gene poly- in patients with Crohn’s disease. Inflamm Bowel Dis
morphism: possible risk for autoimmune disorders. 2014;20:472–480.
J Leukoc Biol 2005;78:231–238. 140. Ts K, Tm N, Jk M, et al. Cannabis for the treatment of
126. Strisciuglio C, Bellini G, Miele E, et al. Cannabinoid re- Crohn’s disease. Cochrane Database Syst Rev 2018;
ceptor 2 functional variant contributes to the risk for 11:CD012853.
pediatric inflammatory bowel disease. J Clin Gastro- 141. Kafil TS, Nguyen TM, MacDonald JK, et al. Cannabis for
enterol 2018;52(5):e37–e43. the treatment of ulcerative colitis. Cochrane Database
127. Storr M, Emmerdinger D, Diegelmann J, et al. The Syst Rev 2018; 2018;11:CD012954.
cannabinoid 1 receptor (CNR1) 1359 G/A polymorphism 142. Naftali T, Bar Lev L, Dotan I, et al. Cannabis induces a
modulates susceptibility to ulcerative colitis and the clinical response in patients with Crohn’s disease: a
phenotype in Crohn’s disease. PLoS One 2010;5(2): prospective placebo-controlled study. Clin Gastro-
e9453. enterol Hepatol 2013;11:1276–1280.e1.
128. Storr M, Emmerdinger D, Diegelmann J, et al. The role of 143. Irving PM, Iqbal T, Nwokolo C, et al. A randomized,
fatty acid hydrolase gene variants in inflammatory bowel double-blind, placebo-controlled, parallel-group, pilot
disease. Aliment Pharmacol Ther 2009;29:542–551. study of cannabidiol-rich botanical extract in the symp-
129. Ambrose T, Simmons A. Cannabis, cannabinoids, and tomatic treatment of ulcerative colitis. Inflamm Bowel Dis
the endocannabinoid system-is there therapeutic 2018;19;24:714–724.
80 Gotfried et al Gastroenterology Vol. 159, No. 1

144. Yacyshyn B, Ginsberg DC, Gilder K, et al.


Received August 8, 2019. Accepted March 29, 2020.
Su1930—Safety and efficacy of olorinab, a peripherally
PERSPECTIVES
REVIEWS AND

restricted, highly selective, cannabinoid receptor 2 Correspondence


agonist in a phase 2A study in chronic abdominal pain Address correspondence to: Ron Schey, MD, FACG, Division of
Gastroenterology/Hepatology, Department of Internal Medicine, University of
associated with Crohn’s disease. Gastroenterology Florida College of Medicine, Jacksonville, Florida. e-mail:
2019;156:S-665. Ron.Schey@jax.ufl.edu.
145. Vemuri VK, Makriyannis A. Medicinal chemistry of can-
CRediT Authorship Contributions
nabinoids. Clin Pharmacol Ther 2015;97:553–558. Siyuan Ding, PhD (Writing – original draft: Equal; Writing – review & editing:
146. Naftali T, Bar-lev Schleider L, Sklerovsky Benjaminov F, Equal); T. Jake Liang, M.D. (Writing – original draft: Equal; Writing – review &
et al. Cannabis induces clinical and endoscopic editing: Equal).
improvement in moderately active ulcerative colitis. Conflicts of interest
Gastroenterology 2018;154:S378; Abstract Suppl 1. The authors disclose no conflicts.

You might also like