You are on page 1of 10

C H A P T E R

72
The Role of 5-HT1A Receptor, and
Nausea and Vomiting Relief by
Cannabidiol (CBD), Cannabidiolic Acid
(CBDA), and Cannabigerol (CBG)
E.M. Rock, L.A. Parker
Department of Psychology and Collaborative Neuroscience Program,
University of Guelph, Guelph, ON, Canada

SUMMARY POINTS KE Y FA CTS O F CA NNA B I D O L A N D


• Cannabidiol and cannabidiolic acid reduce acute CA NNA B I D I O L I C A CI D ’ S E F F E C T S
vomiting, acute nausea, and anticipatory nausea I N A NI M A L M O D E L S O F NAUS E A
in animal models. A ND E M E S I S
• Cannabidiol and cannabidiolic acid’s effects • Cannabidiol and cannabidiolic acid reduce acute
are mediated by indirect agonism of the 5-HT1A vomiting, acute nausea, and anticipatory nausea in
receptor. animal models.
• Cannabidiolic acid is much more potent than
• Cannabigerol acts as a 5-HT1A receptor
cannabidiol in these animal models.
antagonist, as it blocks the antinausea and
• These effects are mediated via the
antiemetic effects of cannabidiol
5-hydroxytryptamine 1A (5-HT1A) receptor, as
• Cannabidiol and/or cannabidiolic acid may be administration of a 5-HT1A antagonist blocks these
potential treatments for chemotherapy-induced effects.
nausea and vomiting. • As these phytocannabinoids do not exert their actions
• Human clinical trials need to evaluate the efficacy via the cannabinoid 1 receptor, these compounds are
of cannabidiolic acid to reduce chemotherapy- nonpsychoactive.
induced nausea and vomiting, especially for • Cannabidiol and/or cannabidiolic acid may be
the reduction of anticipatory nausea, because promising therapeutics for cancer patients undergoing
there is currently no selective treatment for this chemotherapy.
symptom.

Handbook of Cannabis and Related Pathologies. http://dx.doi.org/10.1016/B978-0-12-800756-3.00083-1


Copyright © 2017 Elsevier Inc. All rights reserved.
703
704 72.  CBD, CBDA, AND CBG

potential treatments, and the involvement of the 5-HT1A


KEY FA C T S OF CANNAB IGEROL’ S receptors in nausea and vomiting.
EFFECTS I N ANIMAL MODELS O F
N AU SEA A ND EMESIS
• Cannabigerol blocks cannabidiol’s antiemetic effects, NAUSEA AND VOMITING
suggesting that cannabigerol acts as a 5-HT1A receptor
antagonist. Emesis (vomiting), the act of eliminating poisonous
• Cannabigerol also blocks cannabidiol’s antinausea or toxic contents via expulsion through the mouth, can
effects, suggesting that cannabigerol acts as a 5-HT1A be observed and quantified objectively. In contrast, nau-
receptor antagonist. sea, a subjective experience, is less easily defined. It has
• These two phytocannabinoids may be acting in been characterized as “an unpleasant, but not painful,
opposition at the 5-HT1A receptor to modulate nausea sensation associated with a heightened awareness of the
and vomiting upper gut, cold sweating and the expectation that vomit-
• This finding has important implications for the ing is imminent” (Andrews, Rapeport, & Sanger, 1988).
potential therapeutic effects of various Cannabis sativa Nausea and vomiting are distressing symptoms associ-
strains. ated with pharmacological treatments, such as with an-
• A C. sativa strain with higher cannabidiol relative to ticancer drug therapies.
cannabigerol may be more effective in reducing nausea
and vomiting in human patients.
Chemotherapy-Induced Nausea and Vomiting
Cancer patients undergoing chemotherapy treatment
often experience the distressing side effects of acute nau-
LIST OF ABBREVIATIONS sea and vomiting, as well as delayed nausea and vomit-
ing (Fiore & Gralla,  1984). If posttreatment nausea and
AN Anticipatory nausea vomiting are not properly managed, anticipatory nausea
CB1 Cannabinoid receptor 1 (AN) and vomiting—a conditioned response elicited by
CB2 Cannabinoid receptor 2 reexposure to the illness-paired context of the clinic—
CBD Cannabidiol can result (Morrow,  1982). Please refer to Table  72.1
CBDA Cannabidiolic acid for a description of the three phases of chemotherapy-
CBG Cannabigerol induced nausea and vomiting. Although acute vomiting
DRN Dorsal raphe nucleus has become relatively well managed since the advent of
5-HIAA 5-Hydroxyindoleacetic acid the 5-hydroxytryptamine 3 (5-HT3) receptor antagonists
5-HT Serotonin such as ondansetron, and the neurokinin-1 (NK-1) recep-
5-HT1A 5-Hydroxytryptamine 1A tor antagonists, such as aprepitant (Hickok et al., 2003),
5-HT3 5-Hydroxytryptamine 3 delayed nausea and AN are still unmanaged (Hickok
LiCl Lithium chloride et al., 2003). Nausea is often identified as one of the most
MRN Median raphe nucleus distressing symptoms experienced by cancer patients un-
NK-1 Neurokinin-1 dergoing chemotherapy treatment (Aapro, Molassiotis,
8-OH-DPAT 8-Hydroxy-2-(di-n-propylamino) tetralin
THC ∆9-Tetrahydrocannabinol
TABLE 72.1 Three Phases of Chemotherapy-Induced Nausea
and Vomiting
INTRODUCTION Phase Description

Acute Occurring within 24 h after treatment (day 1 of


Cannabis sativa has been used for medicinal purposes the chemotherapy cycle)
for more than 5000 years. Although the psychoactive can-
nabinoid ∆9-tetrahydrocannabinol (THC) was isolated in Delayed Occurring 24 h to 7 days after treatment (days
2–7 of the chemotherapy cycle)
1964, and extensively studied thereafter, only recently
have other cannabinoid compounds been investigated. Anticipatory A conditioned response, whereby patients feel
This chapter reviews recent findings on the potential of ill upon simply returning (or preparing to
return) to the clinic where they have received
the phytocannabinoids cannabidiol, cannabidiolic acid, chemotherapy
and cannabigerol, to modulate nausea and vomiting via Occurring when acute and delayed phases are not
5-hydroxytryptamine1A (5-HT1A) receptors. First, we de- properly managed
scribe chemotherapy-induced nausea and vomiting, the This table describes the three phases of chemotherapy-induced nausea and
preclinical animal models that are used to evaluate these vomiting experienced by cancer patients, as discussed by Bender et al. (2002).

VI.  Effects of specific natural and synthetic cannabinoids


Serotonin1A (5-hydroxytryptamine; 5-HT1A) receptors and nausea and vomiting 705
& Olver, 2005). With some 76% of patients reporting nau- Conditioned Gaping to a Nausea-Paired
sea, and up to 43% of patients reporting vomiting after Context (Anticipatory Nausea)
their first treatment cycle (Morrow et al., 1998), the need Interestingly, in addition to displaying conditioned
for effective pharmacotherapies is apparent. gaping reactions to a LiCl-paired flavor, rats also display
conditioned gaping reactions to a LiCl-paired context
(Limebeer et al., 2008; Rock, Limebeer, Mechoulam, Pio-
ANIMAL MODELS OF NAUSEA melli, & Parker, 2008). Like the chemotherapy patient who
feels nauseous upon returning to the clinic (where they
In order to assess the efficacy of novel pharmaco- have previously experienced nausea-inducing chemother-
therapies, reliable preclinical animal models are neces- apy treatments), these contextually elicited conditioned
sary. While the neural mechanisms of the emetic reflex gaping reactions serve as a rodent model of AN. Indeed,
have been well established using animal models, such once AN develops in human chemotherapy patients, it is
as the ferret and the shrew (Hornby,  2001), the central refractive to classic antiemetic treatments such as ondan-
mechanisms regulating nausea are not well understood setron (Hickok et al., 2003). Similarly, pretreatment with
(Andrews & Horn, 2006). In contrast to vomiting, nausea ondansetron is ineffective in suppressing conditioned
is subjective, making development of an animal model gaping responses elicited by a LiCl-paired context, in rats
difficult. Further, we summarize the current selective (Limebeer, Hall, & Parker, 2006; Rock et al., 2014).
models (for a comprehensive review refer to Sharkey, The conditioned gaping model is a useful tool for as-
Darmani, & Parker, 2014) used to determine the nauseat- sessing the therapeutic potential of a compound, as a re-
ing potential of compounds, and to determine the poten- duction in acute nausea (conditioned gaping to a LiCl-
tial of antinausea agents that can block or reduce nausea. paired flavor) or AN (conditioned gaping to a LiCl-paired
context) can be assessed. The work reviewed here em-
Conditioned Gaping ploys the conditioned gaping model to assess the antin-
ausea properties of the phytocannabinoids that exert their
Conditioned Gaping to a Nausea-Paired action via 5-hydroxytryptamine 1A (5-HT1A) receptors.
Flavor (Acute Nausea)
Recent evidence suggests that conditioned disgust
(gaping) reactions elicited by exposure to a flavor that has SEROTONIN1A
been previously paired with an illness-inducing agent, (5-HYDROXYTRYPTAMINE; 5-HT1A)
such as lithium chloride (LiCl), is a sensitive and selective RECEPTORS AND NAUSEA AND
model of nausea-induced behavior in rats (Parker, Rana, VOMITING
& Limebeer,  2008). Conditioned gaping reactions (see
Fig. 72.1) are only produced by drugs capable of produc- 5-HT1A Receptors
ing vomiting (in emetic species), and antiemetic drugs
suppress conditioned gaping (see Parker et  al.,  2008). 5-HT1A receptors, coupled to Gi/o, are distributed
Using this model, if a rat is pretreated with a compound, throughout the central nervous system (see Hannon
resulting in a reduction of nausea-induced conditioned & Hoyer,  2008), with high levels expressed in limbic
gaping reactions (relative to vehicle-pretreated controls), areas such as the raphe nuclei of the brainstem (Verge
then this compound may be a potential therapeutic for et al., 1986). 5-HT1A autoreceptors, found on the cell bod-
reducing acute nausea. ies and dendrites of serotonin (5-HT) containing neurons
(Riad et al., 2000) in the rat median raphe nucleus (MRN)
and dorsal raphe nucleus (DRN), regulate 5-HT release
(Verge et al., 1986) to terminal forebrain regions by de-
creasing the rate of neuronal firing (see Blier, Pineyro,
el Mansari, Bergeron, & de Montigny, 1998). Activation
of these somatodendritic 5-HT1A autoreceptors leads to a
reduction in neuronal firing (Blier & de Montigny, 1987).
Local 5-HT release in the raphe nuclei reduces neuronal
firing, producing negative feedback regulation of neu-
rotransmitter release (Adell & Artigas,  1991). Indeed,
5-HT receptor agonists inhibited release of 5-HT from in
vitro preparations of rodent brain tissue (Chase, Katz, &
FIGURE 72.1  Conditioned gaping. Conditioned gaping in rats, de- Kopin, 1969). In addition, in vivo, 5-HT receptor agonists
scribed as a wide, triangular opening of the mouth and jaw, exposing reduced the electrical activity of serotonergic neurons in
the incisors, is the most sensitive and reliable rodent model of nausea. the DRN (Aghajanian,  1972). Therefore, stimulation of

VI.  Effects of specific natural and synthetic cannabinoids


706 72.  CBD, CBDA, AND CBG

somatodendritic 5-HT1A autoreceptors results in a de- such as pigeons (Wolff & Leander, 1994, 1995, 1997), Sun-
crease of available 5-HT. cus murinus (house musk shrew; Javid & Naylor,  2006;
Okada, Torii, Saito, & Matsuki,  1994), cats (Lucot &
Crampton,  1987a,b,  1989; Lucot,  1990a,b) and dogs
Classic 5-HT1A Receptor (Gupta & Sharma, 2002). In the rat acute nausea model,
Agonists and Antagonists 8-OH-DPAT reduces conditioned gaping (Limebeer
8-hydroxy-2-(di-n-propylamino) tetralin (8-OH-DPAT) & Parker,  2003; Limebeer, Litt, & Parker,  2009). Please
is a highly selective and centrally active potent ligand refer to Table 72.2 for a summary of the involvement of
(Arvidsson et  al.,  1981) preferentially binding to the so- 5-HT1A receptor ligands in nausea and vomiting using
matodendritic (Hjorth & Magnusson, 1988) 5-HT1A auto- animal models. This evidence indicates that agonism of
receptor (Middlemiss & Fozard,  1983). 8-OH-DPAT has the 5-HT1A receptor via classic 5-HT1A ligands reduces
traditionally been utilized as the standard receptor ago- toxin-induced nausea and vomiting. The remainder of
nist to evaluate behavioral effects of the 5-HT1A receptor. this chapter will focus on the constituents of the canna-
Indeed, systemically administered 8-OH-DPAT dose- bis plant (phytocannabinoids) that exert their antinau-
dependently reduces the 5-HT synthesis rate, decreases sea and antiemetic effects via activation of the 5-HT1A
5-hydroxyindoleacetic acid (5-HIAA), the main metabo- receptor.
lite of 5-HT (Sharp, Bramwell, & Grahame-Smith, 1989),
and inhibits 5-HT neuronal firing in the DRN (Blier,
Steinberg, Chaput, & de Montigny,  1989), ultimately PHYTOCANNABINOIDS AND
reducing release of 5-HT in the brain (Hjorth et al., 1982). NAUSEA AND VOMITING
WAY100135, a 5-HT1A receptor antagonist, has been
used to investigate this receptor, however, WAY100135 C. sativa has been used for centuries as a therapeu-
has since been shown to act at both presynaptic and tic for many ailments (Mechoulam,  2005), such as the
postsynaptic 5-HT1A receptors (Fletcher et  al.,  1993). alleviation of nausea and vomiting. In fact, in the 19th
More recently, the highly selective and potent 5-HT1A century, C. sativa was used in British medical practice to
receptor antagonist WAY100635 has replaced the use of treat nausea and vomiting (O’Shaughnessy, 1843). In the
WAY100135 (Forster et al., 1995). These 5-HT1A receptor 1970s, young chemotherapy patients anecdotally report-
antagonists effectively block the effects of 8-OH-DPAT ed experiencing less (or none) of their nausea and vom-
on extracellular levels of 5-HT in the rat hippocampus iting when they were “high” from smoking cannabis
(Assie & Koek, 1996) and, when given alone, WAY100135 (Sweet, Miller, Weddington, Senay, & Sushelsky,  1981),
but not WAY100635 decreases 5-HT levels, indicating prompting early clinical trials demonstrating the effi-
possible partial agonist properties of WAY100135 at so- cacy of THC, the psychoactive component in cannabis,
matodendritic 5-HT1A receptors (Assie & Koek, 1996). to reduce chemotherapy-induced nausea and vomiting
(Sweet et  al.,  1981). Due to the unsuccessful control of
chemotherapy-induced nausea and vomiting, oncolo-
Classic 5-HT1A Receptors, and
gists began to investigate cannabinoids for their thera-
Nausea and Vomiting in Humans peutic potential. Recently, the mechanisms by which
With the advent of the antiemetic 5-hydroxytryptamine cannabinoids reduce chemotherapy-induced nausea and
3 (5-HT3) receptor antagonists, most nausea and vomiting vomiting have been investigated (see Sharkey et al., 2014
research has focused on the involvement of this recep- for review) using animal models.
tor, rather than the 5-HT1A receptor, in mediating these
effects. Although a great deal of animal literature sup-
ports the involvement of the somatodendritic 5-HT1A
Cannabidiol
autoreceptors in the regulation of emesis and nausea, A major nonpsychoactive component in cannabis,
little human literature exists. Chial et al. (2003) reported cannabidiol (CBD), was isolated from Lebanese hash-
that administration of the 5-HT1A receptor partial ago- ish in 1963 by Mechoulam and coworkers (Mechoulam
nist buspirone selectively suppressed self-report nausea & Hanus, 2002). CBD has a very low affinity (in the mi-
scores in the nutrient drink test (an assessment of gastric cromolar range) for the cannabinoid 1 (CB1) and can-
functioning). nabinoid 2 (CB2) receptors (Pertwee,  2008). Although
CBD exerts its therapeutic effects via a number of
Classic 5-HT1A Receptors and mechanisms, CBD’s antiemetic and antinausea effects
seem to be mediated by action at the 5-HT1A receptor.
Nausea and Vomiting in Animals
For a summary of the antinausea and antiemetic ef-
5-HT1A receptor agonists such as 8-OH-DPAT, buspi- fects of cannabidiol in animal models, please refer to
rone, and LY228729 suppress vomiting in emetic species, Table 72.3.

VI.  Effects of specific natural and synthetic cannabinoids


Phytocannabinoids and nausea and vomiting 707
TABLE 72.2 Summary of Classic 5-HT1A Receptor Ligand Modulation of Nausea and Vomiting in Animal Models
Compound Species Effect References

8-OH-DPAT Pigeon Eliminated ditolyguanidine-induced emesis Wolff and Leander (1994)

Suncus murinus Eliminated motion-induced emesis Brame and Lucot (2011); Javid and Naylor
(2006); Okada et al. (1994)

Eliminated nicotine-, veratrine-, cisplatin-, or copper Brame and Lucot (2011); Okada et al. (1994)
sulfate-induced emesis

Cat Reduced motion-, cisplatin- or xylazine-induced emesis Lucot and Crampton (1989)

Eliminated RU 24969-induced emesis Lucot (1990b)

Rat Reduced acute nausea Limebeer and Parker (2003); Limebeer et al.
(2009); Rock et al. (2011, 2012)

Buspirone S. murinus Eliminated motion-induced emesis Okada et al. (1994)

Cat Reduced motion-, xylazine-induced emesis Lucot (1990a); Lucot and Crampton (1987b)

Eliminated cisplatin-induced emesis Lucot and Crampton (1987a)

Dog Reduced cisplatin-induced emesis Gupta and Sharma (2002)

Eliminated apomorphine-induced emesis Gupta and Sharma (2002)

LY228729 Pigeon Eliminated ditolyguanidine-induced emesis Wolff and Leander (1994)

Eliminated cisplatin-, ipecac-, emetine- and Wolff and Leander (1995)


m-(chlorophenyl)-biguanide-induced emesis

Eliminated cyclophosphamide-induced emesis Wolff and Leander (1997)

SUN8399 S. murinus Eliminated motion-induced emesis Okada et al. (1994)


Eliminated nicotine-, veratrine-, cisplatin-, copper
sulfate-induced emesis

Ipsapirone S. murinus Eliminated motion-induced emesis Okada et al. (1994)

Tandospirone S. murinus Eliminated motion-induced emesis Okada et al. (1994)

Gepirone S. murinus Eliminated motion-induced emesis Okada et al. (1994)

WAY100635 S. murinus No effect alone on motion-induced emesis Javid and Naylor (2006)
Unable to block 8-OH-DPAT-induced attenuation of
motion-induced emesis
This table summarizes the published effects of classic 5-HT1A receptor ligands in animal models of nausea and emesis.

CBD Reduces Nausea and Vomiting induced vomiting has been suppressed by low doses (5–
in Human Patients 10  mg/kg) of CBD, while higher doses (20–40  mg/kg)
In a phase II clinical trial, when added to the standard of CBD were found to facilitate (rather than reduce) Li-
therapy, the oromucosally administered cannabis-based Cl-induced vomiting (Parker et  al.,  2004). Interestingly,
medicine Sativex (containing approximately 50/50 THC CBD is ineffective in reducing motion-induced vomiting
and CBD) was effective (57% of Sativex patients vs 22% of (Cluny et al., 2008). Consistent with the low binding af-
placebo patients reporting no delayed emesis, and 71% of finity of CBD to the CB1 receptor (Pertwee, 2008), the CB1
Sativex patients vs 22% of placebo patients reporting no receptor antagonist SR141716 did not block the CBD-
delayed emesis) in suppressing delayed nausea and vom- induced suppression of vomiting (Parker et  al.,  2004).
iting (Duran et al., 2010). To date, there are no published WAY100135 effectively blocked the CBD- (5 mg/kg) in-
human clinical trials evaluating the ability of CBD alone duced suppression of acute nicotine-, LiCl- and cisplatin-
to reduce chemotherapy-induced nausea and vomiting. (20  mg/kg, but not 40  mg/kg) induced vomiting in
S. murinus, indicating a 5-HT1A receptor mediated anti-
CBD Reduces Vomiting in Animal Models emetic effect (Rock et al., 2012).
In S. murinus, CBD produces a biphasic effect, at
low doses (5  mg/kg) suppressing, and at higher doses CBD Reduces Acute Nausea in Animal Models
(40 mg/kg) potentiating acute cisplatin-induced vomit- In rats, CBD (5  mg/kg) has been shown to reduce
ing (Kwiatkowska et al., 2004). In addition, acute LiCl- conditioned gaping to a LiCl-paired flavor, when

VI.  Effects of specific natural and synthetic cannabinoids


708 72.  CBD, CBDA, AND CBG

TABLE 72.3 Summary of Cannabidiol’s Effects in Animal Models of Nausea and Vomiting


Phytocannabinoid Animal model Effect Mechanism of action References

Cannabidiol Emesis in Suncus Reduced cisplatin-induced emesis 5-HT1A receptor Kwiatkowska, Parker,
murinus (5 mg/kg) Burton, and Mechoulam (2004);
Rock et al. (2012)

Potentiated cisplatin-induced Not evaluated Kwiatkowska et al. (2004)


emesis (40 mg/kg)

Reduced LiCl-induced emesis 5-HT1A receptor Parker, Kwiatkowska,


(5–10 mg/kg) Burton, and Mechoulam (2004);
Rock et al. (2012)

Potentiated LiCl-induced emesis Not evaluated Parker et al. (2004)


(20–40 mg/kg)

Reduced nicotine-induced emesis 5-HT1A receptor Rock et al. (2012)

Ineffective for motion-induced Not evaluated Cluny, Naylor, Whittle,


emesis and Javid (2008)

Acute nausea Reduced conditioned gaping 5-HT1A receptor Rock et al. (2012)
in rats
Enhanced 8-OH-DPAT-induced Not evaluated Rock et al. (2012)
reduction of conditioned gaping

Anticipatory Reduced conditioned gaping Not evaluated Rock et al. (2008)


nausea in rats
This table summarizes the published effects of cannabidiol (CBD) in animal models of nausea and emesis, as well as whether these effects are mediated by the
5-HT1A receptor.

administered systemically (Rock et al., 2012) or centrally however, CBD’s mechanism of action in AN has not yet
to the DRN (Rock et al., 2012). In addition, at a subthresh- been evaluated. As there are currently no specific treat-
old dose (0.5  mg/kg), CBD systemically enhances the ments for AN, this nonpsychoactive component in can-
antinausea effect of a subthreshold dose (0.005 mg/kg) nabis could be a promising therapeutic option; however,
of 8-OH-DPAT (Rock et al., 2012). its limited effective dose range may prove difficult.
The antinausea effect of CBD has been blocked by
WAY100635, when it is systemically or centrally (intra- Conclusions
DRN) administered (Rock et  al.,  2012). In fact, when It is unfortunate that CBD has not yet been investigat-
WAY100635 was infused outside of the DRN, CBD’s ed in published human clinical trials with chemotherapy
antinausea effects were not blocked, indicating that patients, despite its efficacy in reducing acute vomiting,
the effect was selective to the DRN. Since the DRN is acute nausea, and anticipatory nausea in animal mod-
a site of somatodendritic 5-HT1A autoreceptors, activa- els (via indirect 5-HT1A receptor agonism). Indeed, CBD
tion of which decreases the firing rate of 5-HT afferents has also been shown to inhibit paclitaxel-induced neuro-
projecting to terminal brain regions (Verge et al., 1985), pathic pain (through 5-HT1A receptors), without dimin-
these results suggest that CBD may be exerting its an- ishing chemotherapeutic efficacy (Ward et al., 2014), and
tinausea effects by reducing the firing rate of 5-HT af- has even been shown to have antitumor action itself (De
ferents which project to terminal forebrain areas. CBD Petrocellis et al., 2013).
enhanced the action of 8-OH-DPAT in vitro (% stimu-
lation of [35S]GTPγS binding above basal) and in vivo
Cannabidiolic Acid
(conditioned gaping), without acting as a direct agonist
at the 5-HT1A receptor in brainstem preparations (Rock Cannabidiolic acid (CBDA) is the nonpsychoac-
et al., 2012). Therefore, CBD may be acting as an indi- tive carboxylic acidic precursor of CBD. It is present
rect agonist, thereby augmenting endogenous 5-HT ac- in the fresh cannabis plant, and decarboxylates upon
tivation of the 5-HT1A receptor to reduce toxin-induced heating, such as when cannabis is smoked or dried.
acute nausea. In comparison to CBD, much less work has focused
on this acidic precursor. Studies in our laboratory
CBD Reduces Anticipatory Nausea indicate that it is a highly potent antiemetic and an-
in Animal Models tinausea agent. For a summary of the antinausea and
CBD (1 and 5, but not 10  mg/kg) reduced condi- antiemetic effects of CBDA in animal models, please
tioned gaping to a LiCl-paired context (Rock et al., 2008); refer to Table 72.4.

VI.  Effects of specific natural and synthetic cannabinoids


Phytocannabinoids and nausea and vomiting 709
TABLE 72.4 Summary of Cannabidiolic Acid’s Effects in Animal Models of Nausea and Vomiting
Phytocannabinoid Animal model Effect Mechanism of action References

Cannabidiolic acid Emesis in Suncus Reduced motion-, LiCl-, or Not evaluated Bolognini et al. (2013)
murinus cisplatin-induced emesis

Acute nausea in rats Reduced conditioned gaping 5-HT1A receptor Bolognini et al. (2013)

Enhanced ondansetron- or Not evaluated Rock and Parker (2013a,b)


metoclopramide-induced
suppression of conditioned gaping

Anticipatory nausea Reduced conditioned gaping 5-HT1A receptor Bolognini et al. (2013)
in rats
Enhanced tetrahydrocannabinolic 5-HT1A receptor and Rock et al. (2014)
acid-induced suppression of CB1 receptor
conditioned gaping
This table summarizes the published effects of cannabidiolic acid (CBDA) in animal models of nausea and emesis, as well as whether these effects are mediated by
the 5-HT1A receptor.

CBDA Reduces Vomiting in Animal Models THCA, indicating the involvement of both receptors in
Recent evidence indicates that CBDA (0.1 and/or this effect (Rock et al., 2014).
0.5  mg/kg, i.p.) potently reduces motion-, LiCl-, and Conclusions
cisplatin-induced vomiting in S. murinus (Bolognini
et  al.,  2013). Unfortunately, the 5-HT1A receptor media- CBDA suppresses nausea and vomiting in rats, in
tion of these effects has not yet been investigated. a 5-HT1A receptor dependent manner, suggesting that
CBDA could be developed as a treatment for nausea and
CBDA Reduces Acute Nausea in Animal Models vomiting because of its potency. CBDA could be a par-
CBDA (0.1  mg/kg) also reduced LiCl-induced acute ticularly interesting candidate treatment for AN, as there
nausea, an effect that was blocked by WAY100635, but not is no current selective therapeutic available, once AN
SR141716 (Bolognini et al., 2013). CBDA also increased the develops. CBDA is also promising for development as a
ability of the 5-HT1A receptor agonist, 8-OH-DPAT, to po- treatment for acute nausea, because, unlike CBD, it does
tently stimulate [35S]GTPγS binding to rat brainstem mem- not potentiate nausea and/or vomiting at high doses. In
brane, without activating CB1 receptors in vitro (Bolognini fact, CBDA is active at much lower doses than CBD, but
et al., 2013). CBDA, like CBD, seems to inhibit nausea in at high doses CBDA is merely ineffective. As well, CBDA
rats by enhancing the activation of 5-HT1A receptors. More does not modify overall activity level of rats (Bologini
recently, CBDA has been shown to reduce acute nausea at et al., 2013; Rock et al., 2014). In addition to its antiemetic
a dose as low as 0.5 µg/kg (Rock & Parker, 2013a). As well, and antinausea properties, CBDA (5, 10, 25  mM) inhib-
a subthreshold dose of CBDA (0.1 µg/kg) enhanced the its highly aggressive human breast cancer cell migration,
ability of a mildly effective dose of ondansetron (1 µg/kg), a factor crucial in the prevention of cancer metastasis
or a subthreshold dose of metoclopramide (0.3 mg/kg, a (Takeda et al., 2012). These results highlight the need for
dopamine antagonist) to reduce LiCl-induced acute nau- clinical trials with cancer patients undergoing chemo-
sea (Rock & Parker, 2013a,b). therapy, perhaps especially in breast cancer patients, in
order to prevent metastasis.
CBDA Reduces Anticipatory Nausea
in Animal Models Cannabigerol
CBDA suppressed contextually elicited conditioned Cannabigerol (CBG), a nonpsychoactive component
gaping (at doses as low as 0.001  mg/kg) in the rodent of cannabis (Gaoni & Mechoulam, 1964) has been shown
model of AN (Rock et  al.,  2014), and this effect was to act as a 5-HT1A receptor antagonist (Cascio, Gauson,
blocked by the 5-HT1A receptor antagonist, WAY100635 Stevenson, Ross, & Pertwee, 2010), and recent work in our
(Bolognini et  al.,  2013). In addition, coadministration laboratory has confirmed a 5-HT1A mediated effect in our
of subthreshold doses of CBDA (0.1  µg/kg) and tetra- animal models. For a summary of CBG’s effects in animal
hydrocannabinolic acid (the acidic precursor of THC, models of nausea and vomiting, please refer to Table 72.5.
5  µg/kg) enhanced the suppression of AN, over that
of either agent alone (Rock et  al.,  2014), indicating CBG Blocks CBD-Induced Antiemetic
that these acidic precursors potently reduce AN. Pre- Effects in Animal Models
treatment with SR141716 (CB1 receptor antagonist) or CBG (5 mg/kg, i.p.) blocked the CBD- (5 mg/kg, i.p.)
WAY100635 blocked the combined effect of CBDA and induced suppression of LiCl-induced acute vomiting in

VI.  Effects of specific natural and synthetic cannabinoids


710 72.  CBD, CBDA, AND CBG

TABLE 72.5 Summary of Cannabigerol’s Effects in Animal Models of Nausea and Vomiting


Phytocannabinoid Animal model Effect Mechanism of action References

Cannabigerol Emesis in Suncus Blocked the CBD-induced reduction of LiCl-induced 5-HT1A receptor Rock et al. (2011)
murinus emesis

Acute nausea in Blocked the CBD- or 8-OH-DPAT-induced reduction 5-HT1A receptor Rock et al. (2011)
rats of conditioned gaping (5 or 10 mg/kg)

Reduced conditioned gaping (1 mg/kg) Not evaluated Rock et al. (2011)


This table summarizes the published effects of cannabigerol (CBG) in animal models of nausea and emesis, as well as whether these effects are mediated by the
5-HT1A receptor.

S. murinus, suggesting that CBG exerts its action at the Anticipatory nausea  A classically conditioned response to the
5-HT1A receptor to block the effect of CBD at this receptor hospital context that becomes associated with the chemotherapy-
induced illness experienced by patients (see Table 72.1). This
(Rock et al., 2011). condition is modeled in rats with conditioned gaping to a lithium
chloride-paired context.
CBG Blocks CBD-Induced Antinausea Cannabidiol (CBD)  A nonpsychoactive phytocannabinoid
Effects in Animal Models exhibiting a low binding affinity for the CB 1 and 2 receptors.
CBG (5 or 10 mg/kg, i.p.) blocked the CBD- (5 mg/kg, Its antinausea and antiemetic effects are mediated by the
5-HT1A receptor, as the CBD-induced effects can be blocked
i.p.) and 8-OH-DPAT- (0.01  mg/kg, s.c.) induced sup- by a 5-HT1A receptor antagonist.
pression of acute nausea (Rock et al., 2011), again suggest- Cannabidiolic acid (CBDA)  The acidic precursor to cannabidiol,
ing that CBG is acting on the 5-HT1A receptor to block the found in the fresh Cannabis sativa plant. CBDA potently reduces
5-HT1A mediated effects of CBD and 8-OH-DPAT. Also nausea and vomiting through a 5-HT1A receptor mediated
of interest is the finding that CBG (1 mg/kg, i.p.) actually mechanism, as the CBDA-induced effects can be blocked by a
5-HT1A receptor antagonist.
suppressed acute nausea on its own (Rock et  al.,  2011). Cannabigerol (CBG)  A nonpsychoactive phytocannabinoid that
This result is in line with the in vitro concentration spe- acts as a 5-HT1A receptor antagonist, in contrast to CBD. In fact,
cific effects of CBG, such that low concentrations of CBG CBG blocks CBD’s antiemetic and antinausea effects.
stimulate GTPγS binding to mouse brain membranes, but Conditioned gaping  Although rats cannot vomit, they exhibit
this effect disappears at higher concentrations where CBG conditioned disgust reactions (gaping), in response to the
delivery into their oral cavity of a flavor that has been previously
acts as a 5-HT1A receptor antagonist (Cascio et al., 2010). paired with illness (see Fig. 72.1). This is considered a rodent
model of acute nausea. Gaping, described as a wide, triangular
Summary opening of the mouth and jaw exposing the incisors, is the most
Interestingly, CBG and CBD, two constituents found sensitive and reliable rodent model of nausea. Indeed, rats also
in the cannabis plant, both target 5-HT1A receptors, al- exhibit gaping in response to a context that has been previously
paired with illness—a rodent model of anticipatory nausea.
though with opposite actions, to modulate the effects of Emesis  Also known as vomiting, it is the body’s means of
these receptors on nausea and vomiting. Therefore, the rejecting noxious or toxic substances via the oral cavity. Therefore,
concentration of these opposing cannabinoids in a spe- antiemetic compounds prevent (or reduce) vomiting.
cific strain of cannabis may be important in its efficacy 5-Hydroxytryptamine 1A (5-HT1A) receptors  A serotonin (5-HT)
to reduce chemotherapy-induced nausea and vomiting. receptor subtype binding the endogenous 5-HT neurotransmitter,
mediating inhibitory neurotransmission. The 5-HT1A receptors
located in the dorsal raphe nucleus are largely somatodendritic
(located on the soma and dendrites of serotonergic neurons).
CONCLUSIONS Phytocannabinoid  Cannabinoids derived from C. sativa.
S. murinus  Also known as the house musk shrew, these
It is only recently that the therapeutic potential of phy- insectivores vomit in response to toxins such as lithium chloride.
tocannabinoids (other than THC) has been investigated. Therefore, S. murinus are an animal model to assess whether
certain compounds can reduce (or eliminate) vomiting, indicating
Here we provide evidence that several nonpsychoac- an antiemetic effect.
tive phytocannabinoids (CBD, CBDA, CBG), which act
at 5-HT1A receptors, are potential treatment options for Acknowledgments
chemotherapy-induced nausea and vomiting.
We would like to thank NSERC (Grant #92057) and GW Pharmaceuti-
cals for their support of this work, and in particular GW Pharmaceu-
ticals for supply of several compounds that we have evaluated in the
MINI-DICTIONARY experiments reviewed here.

Acute nausea  Chemotherapy-induced nausea that occurs within


24 h of the initial chemotherapy administration (see Table 72.1).
References
This condition is modeled in rats with conditioned gaping to a Aapro, M. S., Molassiotis, A., & Olver, I. (2005). Anticipatory nausea
lithium chloride-paired flavor. and vomiting. Supportive Care in Cancer, 13(2), 117–121.

VI.  Effects of specific natural and synthetic cannabinoids


REFERENCES 711
Adell, A., & Artigas, F. (1991). Differential effects of clomipramine given Fiore, J. J., & Gralla, R. J. (1984). Pharmacologic treatment of
locally or systemically on extracellular 5-hydroxytryptamine in ra- chemotherapy-induced nausea and vomiting. Cancer Investigation,
phe nuclei and frontal cortex. an in vivo brain microdialysis study. 2(5), 351–361.
Naunyn-Schmiedebergs Archives of Pharmacology, 343(3), 237–244. Fletcher, A., Bill, D. J., Bill, S. J., Cliffe, I. A., Dover, G. M., Forster, E.
Aghajanian, G. K. (1972). Influence of drugs on the firing of serotonin- A.,  et  al. (1993). WAY100135: A novel, selective antagonist at pre-
containing neurons in brain. Federation Proceedings, 31(1), 91–96. synaptic and postsynaptic 5-HT1A receptors. European Journal of
Andrews, P. L., & Horn, C. C. (2006). Signals for nausea and emesis: Pharmacology, 237(2–3), 283–291.
implications for models of upper gastrointestinal diseases. Auto- Forster, E. A., Cliffe, I. A., Bill, D. J., Dover, G. M., Jones, D., Reilly,
nomic Neuroscience: Basic and Clinical, 125(1–2), 100–115. Y.,  et  al. (1995). A pharmacological profile of the selective silent
Andrews, P. L., Rapeport, W. G., & Sanger, G. J. (1988). Neuropharma- 5-HT1A receptor antagonist, WAY-100635. European Journal of
cology of emesis induced by anti-cancer therapy. Trends in Pharma- Pharmacology, 281(1), 81–88.
cological Sciences, 9(9), 334–341. Gaoni, Y., & Mechoulam, R. (1964). The structure and synthesis of
Arvidsson, L. E., Hacksell, U., Nilsson, J. L., Hjorth, S., Carlsson, A., cannabigerol, a new hashish constituent. Proceedings of the Chemical
Lindberg, P., et al. (1981). 8-hydroxy-2-(di-n-propylamino)tetralin, Society, 1964, 82–182.
a new centrally acting 5-hydroxytryptamine receptor agonist. Jour- Gupta, Y. K., & Sharma, S. S. (2002). Involvement of 5-HT1A and
nal of Medicinal Chemistry, 24(8), 921–923. 5-HT2 receptor in cisplatin induced emesis in dogs. Indian Journal of
Assie, M. B., & Koek, W. (1996). Effects of 5-HT1A receptor antagonists Physiology and Pharmacology, 46(4), 463–467.
on hippocampal 5-hydroxytryptamine levels: (S)-WAY100135, but Hannon, J., & Hoyer, D. (2008). Molecular biology of 5-HT receptors.
not WAY100635, has partial agonist properties. European Journal of Behavioural Brain Research, 195(1), 198–213.
Pharmacology, 304(1–3), 15–21. Hickok, J. T., Roscoe, J. A., Morrow, G. R., King, D. K., Atkins, J. N., &
Bender, C. M., McDaniel, R. W., Murphy-Ende, K., Pickett, M., Ritten- Fitch, T. R. (2003). Nausea and emesis remain significant problems
berg, C. N., Rogers, M. P., et al. (2002). Chemotherapy-induced nau- of chemotherapy despite prophylaxis with 5-hydroxytryptamine-3
sea and vomiting. Clinical Journal of Oncology Nursing, 6(2), 94–102. antiemetics: a University of Rochester James P. Wilmot Cancer
Blier, P., & de Montigny, C. (1987). Modification of 5-HT neuron prop- Center community clinical oncology program study of 360 cancer
erties by sustained administration of the 5-HT1A agonist gepirone: patients treated in the community. Cancer, 97(11), 2880–2886.
electrophysiological studies in the rat brain. Synapse (New York, Hjorth, S., Carlsson, A., Lindberg, P., Sanchez, D., Wikström, H.,
NY), 1(5), 470–480. Arvidsson, L., et al. (1982). 8-hydroxy-2-(di-n-propylamino)tetralin,
Blier, P., Pineyro, G., el Mansari, M., Bergeron, R., & de Montigny, C. 8-OH-DPAT, a potent and selective simplified ergot congener with
(1998). Role of somatodendritic 5-HT autoreceptors in modulating central 5-HT-receptor stimulating activity. Journal of Neural Trans-
5-HT neurotransmission. Annals of the New York Academy of Sciences, mission, 55(3), 169–188.
861, 204–216. Hjorth, S., & Magnusson, T. (1988). The 5-HT 1A receptor agonist,
Blier, P., Steinberg, S., Chaput, Y., & de Montigny, C. (1989). Electrophysi- 8-OH-DPAT, preferentially activates cell body 5-HT autoreceptors
ological assessment of putative antagonists of 5-hydroxytryptamine in rat brain in vivo. Naunyn-Schmiedebergs Archives of Pharmacology,
receptors: a single-cell study in the rat dorsal raphe nucleus. Cana- 338(5), 463–471.
dian Journal of Physiology and Pharmacology, 67(2), 98–105. Hornby, P. J. (2001). Central neurocircuitry associated with emesis. The
Bolognini, D., Rock, E. M., Cluny, N. L., Cascio, M. G., Limebeer, C. L., American Journal of Medicine, 111(Suppl. 8A), 106S–112S.
Duncan, M., et al. (2013). Cannabidiolic acid prevents vomiting in Javid, F. A., & Naylor, R. J. (2006). The effect of the 5-HT1A receptor
Suncus murinus and nausea-induced behaviour in rats by enhanc- agonist, 8-OH-DPAT, on motion-induced emesis in Suncus murinus.
ing 5-HT1A receptor activation. British Journal of Pharmacology, Pharmacology, Biochemistry and Behavior, 85(4), 820–826.
168(6), 1456–1470. Kwiatkowska, M., Parker, L. A., Burton, P., & Mechoulam, R. (2004).
Brame, R. E., & Lucot, J. B. (2011). Guamanian Suncus murinus respon- A comparative analysis of the potential of cannabinoids and
siveness to emetic stimuli and the antiemetic effects of 8-OH-DPAT. ondansetron to suppress cisplatin-induced emesis in the Suncus
Pharmacology, Biochemistry and Behavior, 99(3), 381–384. murinus (house musk shrew). Psychopharmacology, 174(2), 254–259.
Cascio, M. G., Gauson, L. A., Stevenson, L. A., Ross, R. A., & Pertwee, Limebeer, C. L., Hall, G., & Parker, L. A. (2006). Exposure to a lithium-
R. G. (2010). Evidence that the plant cannabinoid cannabigerol is a paired context elicits gaping in rats: a model of anticipatory nausea.
highly potent alpha2-adrenoceptor agonist and moderately potent Physiology and Behavior, 88(4–5), 398–403.
5HT1A receptor antagonist. British Journal of Pharmacology, 159(1), Limebeer, C. L., Krohn, J. P., Cross-Mellor, S., Litt, D. E., Ossenkopp,
129–141. K. P., & Parker, L. A. (2008). Exposure to a context previously
Chase, T. N., Katz, R. I., & Kopin, I. J. (1969). Release of [3H]serotonin associated with nausea elicits conditioned gaping in rats: a model
from brain slices. Journal of Neurochemistry, 16(4), 607–615. of anticipatory nausea. Behavioural Brain Research, 187(1), 33–40.
Chial, H. J., Camilleri, M., Burton, D., Thomforde, G., Olden, K. W., & Limebeer, C. L., Litt, D. E., & Parker, L. A. (2009). Effect of 5-HT3
Stephens, D. (2003). Selective effects of serotonergic psychoactive antagonists and a 5-HT(1A) agonist on fluoxetine-induced condi-
agents on gastrointestinal functions in health. American Journal of tioned gaping reactions in rats. Psychopharmacology, 203(4), 763–770.
Physiology. Gastrointestinal and Liver Physiology, 284(1), G130–G137. Limebeer, C. L., & Parker, L. A. (2003). The 5-HT1A agonist 8-OH-
Cluny, N. L., Naylor, R. J., Whittle, B. A., & Javid, F. A. (2008). The DPAT dose-dependently interferes with the establishment and the
effects of cannabidiol and tetrahydrocannabinol on motion- expression of lithium-induced conditioned rejection reactions in
induced emesis in Suncus murinus. Basic and Clinical Pharmacology rats. Psychopharmacology, 166(2), 120–126.
and Toxicology, 103(2), 150–156. Lucot, J. B. (1990a). Effects of serotonin antagonists on motion sickness
De Petrocellis, L., Ligresti, A., Schiano Moriello, A., Iappelli, M., Verde, and its suppression by 8-OH-DPAT in cats. Pharmacology, Biochemis-
R., Stott, C. G., et al. (2013). Non-THC cannabinoids inhibit prostate try and Behavior, 37(2), 283–287.
carcinoma growth in vitro and in vivo: pro-apoptotic effects and un- Lucot, J. B. (1990b). RU 24969-induced emesis in the cat: 5-HT1 sites
derlying mechanisms. British Journal of Pharmacology, 168(1), 79–102. other than 5-HT1A, 5-HT1B or 5-HT1C implicated. European Journal
Duran, M., Perez, E., Abanades, S., Vidal, X., Saura, C., Majem, M., of Pharmacology, 180(2–3), 193–199.
et al. (2010). Preliminary efficacy and safety of an oromucosal stan- Lucot, J. B., & Crampton, G. H. (1987a). Buspirone blocks cisplatin-
dardized cannabis extract in chemotherapy-induced nausea and induced emesis in cats. Journal of Clinical Pharmacology, 27(10),
vomiting. British Journal of Clinical Pharmacology, 70(5), 656–663. 817–818.

VI.  Effects of specific natural and synthetic cannabinoids


712 72.  CBD, CBDA, AND CBG

Lucot, J. B., & Crampton, G. H. (1987b). Buspirone blocks motion sick- Rock, E. M., Limebeer, C. L., Mechoulam, R., Piomelli, D., & Parker,
ness and xylazine-induced emesis in the cat. Aviation, Space and En- L. A. (2008). The effect of cannabidiol and URB597 on conditioned
vironmental Medicine, 58(10), 989–991. gaping (a model of nausea) elicited by a lithium-paired context in
Lucot, J. B., & Crampton, G. H. (1989). 8-OH-DPAT suppresses vomit- the rat. Psychopharmacology, 196(3), 389–395.
ing in the cat elicited by motion, cisplatin or xylazine. Pharmacology, Rock, E. M., Limebeer, C. L., Navaratnam, R., Sticht, M. A., Bonner,
Biochemistry and Behavior, 33(3), 627–631. N., Engeland, K., et al. (2014). A comparison of cannabidiolic acid
Mechoulam, R. (2005). Plant cannabinoids: a neglected pharmacologi- with other treatments for anticipatory nausea using a rat model of
cal treasure trove. British Journal of Pharmacology, 146(7), 913–915. contextually elicited conditioned gaping. Psychopharmacology, 231,
Mechoulam, R., & Hanus, L. (2002). Cannabidiol: an overview of some 3207–3215.
chemical and pharmacological aspects. Part I: chemical aspects. Rock, E. M., & Parker, L. A. (2013a). Effect of low doses of cannabi-
Chemistry and Physics of Lipids, 121(1–2), 35–43. diolic acid and ondansetron on LiCl-induced conditioned gaping
Middlemiss, D. N., & Fozard, J. R. (1983). 8-hydroxy-2-(di-n- (a model of nausea-induced behaviour) in rats. British Journal of
propylamino)-tetralin discriminates between subtypes of the 5-HT1 Pharmacology, 169(3), 685–692.
recognition site. European Journal of Pharmacology, 90(1), 151–153. Rock, E. M., & Parker, L. A. (2013b). Suppression of lithium chloride-
Morrow, G. R. (1982). Prevalence and correlates of anticipatory nau- induced conditioned gaping (a model of nausea-induced behav-
sea and vomiting in chemotherapy patients. Journal of the National iour) in rats (using the taste reactivity test) with metoclopramide
Cancer Institute, 68(4), 585–588. is enhanced by cannabidiolic acid. Pharmacology, Biochemistry and
Morrow, G. R., Roscoe, J. A., Hynes, H. E., Flynn, P. J., Pierce, H. I., Behavior, 111, 84–89.
& Burish, T. (1998). Progress in reducing anticipatory nausea and Sharkey, K. A., Darmani, N. A., & Parker, L. A. (2014). Regulation of
vomiting: a study of community practice. Supportive Care in Cancer: nausea and vomiting by cannabinoids and the endocannabinoid
Official Journal of the Multinational Association of Supportive Care in system. European Journal of Pharmacology, 722, 134–146.
Cancer, 6(1), 46–50. Sharp, T., Bramwell, S. R., & Grahame-Smith, D. G. (1989). 5-HT1 ago-
O’Shaughnessy, W. B. (1843). On the preparations of the Indian hemp, nists reduce 5-hydroxytryptamine release in rat hippocampus in
or gunjah: Cannabis indica, their effects on the animal system in vivo as determined by brain microdialysis. British Journal of Phar-
health, and their utility in the treatment of tetanus and other con- macology, 96(2), 283–290.
vulsive diseases. Provincial Medical Journal and Retrospect of the Sweet, D. L., Miller, N. J., Weddington, W., Senay, E., & Sushelsky, L.
Medical Sciences, 5, 363–369. (1981). Delta 9-tetrahydrocannabinol as an antiemetic for patients
Okada, F., Torii, Y., Saito, H., & Matsuki, N. (1994). Antiemetic effects of receiving cancer chemotherapy. A pilot study. Journal of Clinical
serotonergic 5-HT1A-receptor agonists in Suncus murinus. Japanese Pharmacology, 21(8–9 Suppl.), 70S–75S.
Journal of Pharmacology, 64(2), 109–114. Takeda, S., Okajima, S., Miyoshi, H., Yoshida, K., Okamoto, Y., Okada,
Parker, L. A., Kwiatkowska, M., Burton, P., & Mechoulam, R. (2004). T., et  al. (2012). Cannabidiolic acid, a major cannabinoid in fiber-
Effect of cannabinoids on lithium-induced vomiting in the Suncus type cannabis, is an inhibitor of MDA-MB-231 breast cancer cell
murinus (house musk shrew). Psychopharmacology, 171(2), 156–161. migration. Toxicology Letters, 214(3), 314–319.
Parker, L. A., Rana, S. A., & Limebeer, C. L. (2008). Conditioned nau- Verge, D., Daval, G., Marcinkiewicz, M., Patey, A., el Mestikawy,
sea in rats: assessment by conditioned disgust reactions, rather S., Gozlan, H., et  al. (1986). Quantitative autoradiography of
than conditioned taste avoidance. Canadian Journal of Experimental multiple 5-HT1 receptor subtypes in the brain of control or
Psychology, 62(3), 198–209. 5,7-dihydroxytryptamine-treated rats. The Journal of Neuroscience:
Pertwee, R. G. (2008). The diverse CB1 and CB2 receptor pharmacol- The Official Journal of the Society for Neuroscience, 6(12), 3474–3482.
ogy of three plant cannabinoids: Delta9-tetrahydrocannabinol, Verge, D., Daval, G., Patey, A., Gozlan, H., el Mestikawy, S., & Hamon,
cannabidiol and delta9-tetrahydrocannabivarin. British Journal of M. (1985). Presynaptic 5-HT autoreceptors on serotonergic cell bod-
Pharmacology, 153(2), 199–215. ies and/or dendrites but not terminals are of the 5-HT1A subtype.
Riad, M., Garcia, S., Watkins, K. C., Jodoin, N., Doucet, E., Langlois, X., European Journal of Pharmacology, 113(3), 463–464.
et al. (2000). Somatodendritic localization of 5-HT1A and pretermi- Ward, S. J., McAllister, S. D., Kawamura, R., Murase, R., Neelakantan,
nal axonal localization of 5-HT1B serotonin receptors in adult rat H., & Walker, E. A. (2014). Cannabidiol inhibits paclitaxel-induced
brain. Journal of Comparative Neurology, 417(2), 181–194. neuropathic pain through 5-HT(1A) receptors without diminishing
Rock, E. M., Bolognini, D., Limebeer, C. L., Cascio, M. G., Anavi-Goffer, nervous system function or chemotherapy efficacy. British Journal of
S., Fletcher, P. J., et al. (2012). Cannabidiol, a non-psychotropic com- Pharmacology, 171(3), 636–645.
ponent of cannabis, attenuates vomiting and nausea-like behaviour Wolff, M. C., & Leander, J. D. (1994). Antiemetic effects of 5-HT1A ago-
via indirect agonism of 5-HT(1A) somatodendritic autoreceptors nists in the pigeon. Pharmacology, Biochemistry, and Behavior, 49(2),
in the dorsal raphe nucleus. British Journal of Pharmacology, 165(8), 385–391.
2620–2634. Wolff, M. C., & Leander, J. D. (1995). Comparison of the antiemetic
Rock, E. M., Goodwin, J. M., Limebeer, C. L., Breuer, A., Pertwee, R. G., effects of a 5-HT1A agonist, LY228729, and 5-HT3 antagonists in
Mechoulam, R., et al. (2011). Interaction between non-psychotropic the pigeon. Pharmacology, Biochemistry and Behavior, 52(3), 571–575.
cannabinoids in marihuana: effect of cannabigerol (CBG) on the Wolff, M. C., & Leander, J. D. (1997). Effects of a 5-HT1A receptor ago-
anti-nausea or anti-emetic effects of cannabidiol (CBD) in rats and nist on acute and delayed cyclophosphamide-induced vomiting.
shrews. Psychopharmacology, 215(3), 505–512. European Journal of Pharmacology, 340(2–3), 217–220.

VI.  Effects of specific natural and synthetic cannabinoids

You might also like