You are on page 1of 12

B RA I N RE SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6

a v a i l a b l e a t w w w. s c i e n c e d i r e c t . c o m

w w w. e l s e v i e r. c o m / l o c a t e / b r a i n r e s r e v

Review

Targeting CB2 receptors and the endocannabinoid system for


the treatment of pain

Praveen Anand a,⁎, Garth Whiteside b , Christopher J. Fowler c , Andrea G. Hohmann d


a
Imperial College London, UK
b
Wyeth Research, Princeton, NJ, USA
c
Department of Pharmacology and Clinical Neuroscience, Umeå University, SE-901 87 Umeå, Sweden
d
University of Georgia, Athens, GA, USA

A R T I C LE I N FO AB S T R A C T

Article history: The endocannabinoid system consists of the cannabinoid (CB) receptors, CB1 and CB2, the
Accepted 29 December 2008 endogenous ligands anandamide (AEA, arachidonoylethanolamide) and 2-
Available online 25 December 2008 arachidonoylglycerol (2-AG), and their synthetic and metabolic machinery. The use of
cannabis has been described in classical and recent literature for the treatment of pain, but
Keywords: the potential for psychotropic effects as a result of the activation of central CB1 receptors
Cannabinoid places a limitation upon its use. There are, however, a number of modern approaches
Anandamide being undertaken to circumvent this problem, and this review represents a concise
CB2 receptor summary of these approaches, with a particular emphasis upon CB2 receptor agonists.
Fatty acid amide hydrolase Selective CB2 agonists and peripherally restricted CB1 or CB1/CB2 dual agonists are being
Neuropathic pain developed for the treatment of inflammatory and neuropathic pain, as they demonstrate
Inflammatory pain efficacy in a range of pain models. CB2 receptors were originally described as being
restricted to cells of immune origin, but there is evidence for their expression in human
primary sensory neurons, and increased levels of CB2 receptors reported in human
peripheral nerves have been seen after injury, particularly in painful neuromas. CB2
receptor agonists produce antinociceptive effects in models of inflammatory and
nociceptive pain, and in some cases these effects involve activation of the opioid
system. In addition, CB receptor agonists enhance the effect of μ-opioid receptor
agonists in a variety of models of analgesia, and combinations of cannabinoids and
opioids may produce synergistic effects. Antinociceptive effects of compounds blocking the
metabolism of anandamide have been reported, particularly in models of inflammatory
pain. There is also evidence that such compounds increase the analgesic effect of non-
steroidal anti-inflammatory drugs (NSAIDs), raising the possibility that a combination of
suitable agents could, by reducing the NSAID dose needed, provide an efficacious
treatment strategy, while minimizing the potential for NSAID-induced gastrointestinal
and cardiovascular disturbances. Other potential “partners” for endocannabinoid
modulatory agents include α2-adrenoceptor modulators, peroxisome proliferator-
activated receptor α agonists and TRPV1 antagonists. An extension of the
polypharmacological approach is to combine the desired pharmacological properties of
the treatment within a single molecule. Hopefully, these approaches will yield novel

⁎ Corresponding author. Fax: +44 20 8383 3363.


E-mail address: p.anand@imperial.ac.uk (P. Anand).

0165-0173/$ – see front matter © 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.brainresrev.2008.12.003
256 B RA I N R E SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6

analgesics that do not produce the psychotropic effects that limit the medicinal use of
cannabis.
© 2009 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 256
2. The endocannabinoid system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 256
3. Targeting the endocannabinoid system for the treatment of pain . . . . . . . . . . . . . . . . . . . . . . . . . . . . 259
3.1. Peripherally-restricted cannabinoid agonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 259
3.2. CB2 receptor-selective agonists (see Fig. 2). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 259
3.3. Inhibitors of endocannabinoid metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
4. Endocannabinoid modulating agents as a component of new pharmacotherapies for pain . . . . . . . . . . . . . . . 262
5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 262
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 262

1. Introduction Herkenham 1999, Price et al. 2003), but there is also evidence
for the expression of CB1 receptors in non-neural tissue (see
In 1993 it was reported in Nature that a carbonized material e.g. Tokanovic et al., 2007). Similarly, while the CB2 receptor
recovered from the abdomen of a young girl who had died in was originally considered to be expressed primarily in
childbirth in the fourth century AD contained traces of a stable lymphoid tissues in the periphery (Munro et al. 1993, Di
constituent of cannabis (Zias et al., 1993). Although this Marzo et al. 2004), recent evidence has pointed to a neuronal
represents a documented early use of cannabis for the localization in some regions of the rodent brain (Onaivi et al.
presumed treatment of pain, the use of cannabis extracts for 2006), and CB2 immunoreactivity has been shown in activated
this and other medical indications, as well as for recreational microglia in affected regions of multiple sclerosis and
uses, has been described in both the ancient world and in amyotrophic lateral sclerosis post mortem human spinal cord
more modern times (review, see Mechoulam, 1986). Currently, (Yiangou et al. 2006). A recent study has also presented
Sativex™, a buccal formulation of cannabis extract with evidence for the localization of CB2 receptor-like immunor-
defined ratios of Δ9-tetrahydroxannabinol (THC, the main eactivity in human DRG sensory neurons in vitro (see Fig. 1), in
psychoactive ingredient of cannabis) and cannabidiol, is injured nerves including neuromas, and in nerve fibers in
licensed in Canada for the treatment of pain in multiple human synovium and digit skin (Anand et al. 2008).
sclerosis patients (Perez and Ribera, 2008). A major issue, Both AEA and 2-AG are synthesized upon demand rather
however, for all treatments based on cannabis is the potential than being pre-stored, and have relatively short durations of
for psychotropic effects and concerns about the long-term use
of such medications. These concerns place a limitation upon
the dosages that can be given and hence the potential level of
pain relief. There are, however, a number of approaches that
can be taken to circumvent this problem.

2. The endocannabinoid system

The endocannabinoid system consists of the G-protein coupled


cannabinoid (CB) receptors, CB1 and CB2, the endogenous ligands
anandamide (AEA, arachidonoylethanolamide) and 2-arachido-
noylglycerol (2-AG), and their synthetic and metabolic machinery.
The role of these endocannabinoids and other putative endo-
cannabinoids in pain modulation and pathways for their synth-
esis and degradation has recently been reviewed (Hohmann and
Suplita 2006). CB1 receptors are primarily neuronal (although they
have been found in non-neuronal tissue) and mediate the
psychotropic actions of cannabis (see Monory et al. (2007) for a
genetic dissection of the neuronal populations involved in the
different behavior al and autonomic effects of THC). Fig. 1 – Membrane bound CB2 receptor (red) and cytoplasmic
CB1 receptors are expressed in neurons of the CNS Gap43 (green) immunostaining in a human DRG small
(Matsuda et al. 1990), and in DRG neurons (Hohmann and neuron in vitro. Bar = 50 μm.
B RA I N RE SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6 257

action, due to effective metabolic pathways. In the case of sion to as yet unidentified designated transporter proteins
AEA, this is brought about by a process of cellular having been postulated (see Fowler, 2008). In addition to the
accumulation followed primarily by hydrolysis to arachido- formal components of the endocannabinoid system sum-
nic acid, catalyzed by the enzyme fatty acid amide hydrolase marized above, there are additional candidate cannabinoid
(FAAH) (although it can also act as a substrate for both receptors and endogenous ligands that have been described
cyclooxygenase-2 and lipoxygenases). In the case of 2-AG, a in the literature (reviews see Bradshaw and Walker, 2005;
similar pattern is seen, although in the brain the enzyme Brown, 2007). Therefore, endocannabinoid system can and
monoacylglycerol lipase is more important. At the outset, it should be regarded as a “work in progress” rather than an
should be pointed out that the exact process whereby cells absolutely defined entity. The recent report that mice
accumulate AEA is not known and is a current area of lacking GPR55 receptors (a putative CB receptor) do not
controversy; mechanisms ranging from FAAH-gated diffu- show mechanical hyperalgesia following either complete

Fig. 2 – Activation of cannabinoid CB2 receptors suppresses the development and maintenance of inflammatory nociception in
behavioral, electrophysiological and neurochemical studies. (a) AM1241 (33 μg/kg i.p.), administered locally in the inflamed
paw, suppresses established carrageenan-evoked mechanical allodynia. Effects were blocked by the CB2 antagonist SR144528
(33 μg/kg i.p.), but not by SR141716A (33 μg/kg i.p.). (b) AM1241 (330 μg/kg i.p.) induces a CB2-mediated suppression of the
development of carrageenan-evoked (b) mechanical allodynia and (c) spinal Fos protein expression. (d) AM1241 (330 μg/kg, i.v.)
suppressed total C-fiber-mediated neuronal excitability in spinal wide dynamic range neurons through a CB2-specific
mechanism. AM1241 (330 μg/kg i.p.) also suppresses capsaicin-evoked (e) thermal hyperalgesia and (f) nocifensive behavior.
(b–f) Effects were completely blocked by SR144528 (1 mg/kg), but not by SR141716A (1 mg/kg). Data (Mean + SEM). Sources: (a)
Gutierrez et al. (2007); (b–c) Nackley et al. (2003a), (d) Nackley et al. (2004); (e–f) Hohmann et al. (2004).
258 B RA I N R E SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6

Table 1 – The pharmacology of the endocannabinoid system—selected compounds


Compound Mechanism of action Effect in pain

THC Primarily activation of CB receptors, although has Inflammatory: +


off-target actions (e.g. Barann et al., 2002) Neuropathic: +

AM1241 CB2-receptor selective ligand; acts as a “protean” Inflammatory: +


agonist in vitro (Yao et al., 2006) and CB2 agonist in vivo Neuropathic: +

GW405833 CB2-receptor selective ligand (efficacy dependent Inflammatory: +


upon assay used, see Yao et al., 2008) Neuropathic: +

LY2318912 Blocks the accumulation and metabolism of AEA Inflammatory: +


(Moore et al., 2005). Acts primarily as a potent FAAH
inhibitor, but with many off-target actions
(Alexander and Cravatt, 2006)

URB597 Selective FAAH inhibitor (Kathuria et al., 2003). Inflammatory: +


Some off-target actions have been reported, but Visceral: +
their importance is unclear Neuropathic: +/−

ibu-am5 Dual COX- and FAAH-inhibitory compound Visceral: +


(Holt et al., 2007).

N-arachidonoylserotonin Dual TRPV1 antagonist/FAAH-inhibitory Inflammatory: +


compound (Maione et al., 2007). Neuropathic: +
B RA I N RE SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6 259

Table 1 (continued)
Compound Mechanism of action Effect in pain

Pravadoline Dual CB agonist/COX-inhibitory compound Inflammatory: +


(D'Ambra et al., 1992). Visceral: +

The compounds shown in this table have been selected since they are those principally discussed in this review. “+” indicates efficacy in a
preclinical model of this pain type (for details, see text). For a more complete list, see Fowler (2008).

Freund's adjuvant treatment or partial nerve ligation (Staton treatment of pain. A good example of this is the study of
et al., 2008) underlines the fact that the detailed character- Dziadulewicz et al. (2007). These authors reported that
ization of the endocannabinoid system is an important naphthalen-1-yl-(4-pentyloxynaphthalen-1-yl)methanone
research priority. Notwithstanding, a number of compounds bound as an agonist to human CB1 and CB2 receptors with IC50
are available that target different components of the values of 15 and 98 nM, respectively, and produced a good
currently accepted endocannabinoid system, compounds separation of effects upon neuropathic pain and catalepsy,
that have been found to have therapeutic potential in consistent with a limited penetration of the compound into
models of inflammatory and in some cases neuropathic the brain.
pain (see Table 1 for a description of the main compounds
forming the core of this review). 3.2. CB2 receptor-selective agonists (see Fig. 2)

Expression of CB2 receptors in the peripheral and central


3. Targeting the endocannabinoid system for nervous systems is modulated in a number of rodent pain
the treatment of pain models. No increase in CB2 receptor mRNA was apparent in the
Freund's Complete Adjuvant (FCA) model of inflammatory pain,
3.1. Peripherally-restricted cannabinoid agonists while upregulation of mRNA and protein were seen in the
ipsilateral dorsal horn of the lumbar spinal cord using two
It has been well established that the ability of cannabinoids to models of neuropathic pain, the chronic constriction injury (CCI)
affect pain perception has supra-spinal, spinal and peripheral (Bennett and Xie, 1988) and spinal nerve ligation (SNL) (Kim and
components (for review, see Hohmann, 2002; Walker and Chung, 1992) models (Zhang et al., 2003). The authors suggest
Hohmann, 2005). With respect to the peripheral component, that the increased expression was in microglia as it colocalized
local administration of both synthetic cannabinoids and with OX-42, a marker of microglia. The upregulation of CB2
exogenous anandamide and 2-AG produce antinociceptive receptor mRNA in spinal cord was confirmed in a subsequent
effects in the formalin model of inflammatory pain (see e.g. study using the SNL model (Beltramo et al., 2006). Here, the cell
Calignano et al., 1998; Guindon et al., 2007). Recently, Agarwal type was ambiguous, though in a separate set of experiments
et al. (2007) reported the effects in pain models of the the authors did provide evidence for upregulation of CB2
conditional deletion of CB1 receptors in nociceptive neurons receptor mRNA in cultured rat spinal microglia. Expression of
of the dorsal root ganglia of the mouse. These mice were more CB2 receptors on microglia is supported by additional studies
sensitive to the effects of intraplantar administration of showing interferon γ (IFNγ) induces upregulation (Carlisle et al.,
capsaicin and formalin, whereas their motor performance on 2002) with a distinct subcellular localization at the surface of
a rotorod test was not affected. The hyperalgesic response to activated microglia in culture (Walter and Stella, 2004). Upregu-
administration of complete Freund's adjuvant was also lation of CB2 receptors in spinal cord was also demonstrated
greater than for wild-type mice, and the ability of a synthetic immunohistochemically following either SNL or axotomy
cannabinoid to alleviate this response was also greatly (Wotherspoon et al., 2005). However, in contrast to the earlier
reduced (Agarwal et al., 2007). These data would suggest that studies, colocalization with GAP-43 and galanin in the super-
peripherally restricted cannabinoids may have utility in ficial lamina suggested expression on primary afferent (C fiber)
inflammatory pain. In this respect, Fride et al. (2004) have terminals (Wotherspoon et al., 2005). This upregulation was
identified analog s of the (+)-enantiomer of cannabidiol that absent in CB2 receptor knockout mice. Supporting neuronal
are active towards CB receptors and reduce the pain response localization, increased CB2 receptor immunoreactivity was also
in the formalin model of inflammatory pain, without produ- seen in nerve sections proximal, but not distal, to the site of
cing overt signs of central CB1 receptor activation. Other ligation (Wotherspoon et al., 2005) (Fig. 2).
peripherally restricted CB1 or CB1/CB2 receptor agonists are In addition to studies demonstrating the presence of CB2
currently being investigated as potential approaches to the receptor in components of the pain pathway and alterations in
260 B RA I N R E SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6

expression level in pain models, agonists both selective and Ki value of ∼ 23 nM at CB2 receptors (versus a Ki value that is
non-selective have been used to investigate the role of CB2 in greater than 10 mM for the CB1 receptor) (Hanuš et al., 1999).
nociception. The non-selective endogenous agonists, AEA and When administered intraperitoneally, it reduced both the
PEA, have been combined with the CB1 receptor-selective inflammation associated with arachidonic acid-induced ear
antagonists, AM251, AM281 and SR141716A and the CB2 swelling and the late phase of formalin-induced pain behavior
receptor-selective antagonists, AM630 and SR144528. Admin- in mice, in an SR144528-sensitive manner (Hanuš et al., 1999).
istration of AEA locally into the rat hindpaw reverses pain due In addition, in the plantar incision model of post-surgical pain,
to both intraplantar carrageenan and formalin (Sokal et al., it has been shown to reduce allodynia. This effect was blocked
2003; Guindon et al., 2006a). While the AEA-induce analgesia by the CB2 receptor-selective antagonist SR144528 (LaBuda et
following formalin was blocked by the CB1 receptor antagonist al., 2005). HU-308 appears to have minimal off target effects as
AM251 (Guindon et al., 2006a), both the CB2 and CB1 receptor- it did not affect acute nociception as measured on a 55 °C hot
selective antagonists, SR144528 and SR141716A, respectively, plate, did not cause catalepsy and did not inhibit ambulation
blocked the effect of AEA on carrageenan-induced hypersen- or rearing in the open field (Hanuš et al., 1999).
sitivity of spinal neurons (Sokal et al., 2003). These results The small molecule CB2 receptor-selective compound that
imply a contribution of both peripherally expressed CB1 and has most substantially penetrated the scientific literature is
CB2 receptors to the antinociceptive response. Importantly in AM1241, that is reported to have an affinity for CB2 receptors of
the study by Sokal et al. (2003), local administration had no 2 nM with a high degree of selectivity over CB1 receptors (95-
effect in non-inflamed tissue. Likewise, systemic administra- 340 fold) (Malan et al., 2001a). This high degree of selectivity is
tion has been shown to reverse CFA-induced mechanical maintained in membranes prepared from mouse spleen and
hyperalgesia in a non-CB1 receptor sensitive manner (Smith brain, for CB2 and CB1 receptors, respectively (Ibrahim et al.,
et al., 1998). 2003). AM1241 is a racemic mix; considering the individual
The non-selective synthetic cannabinoid agonists HU-210, enantiomers R,S-AM1241 act as an agonist at human CB2, but
nabilone, CP55,940 and WIN55,212-2, have also been combined an inverse agonist at rat and mouse CB2 receptors. R-AM1241
with CB1 and CB2 receptor-selective antagonists to confirm the binds with more than 40-fold higher affinity than S-AM1241,
role of CB2 receptors in pain transmission. HU-210 reversed to human rat and mouse CB2 receptors and displays a
carrageenan-induced edema through the CB2 receptor, as the functional profile similar to that of the racemate. In contrast,
CB2 receptor-selective antagonist SR144528 blocked this effect S-AM1241 is an agonist at human, rat and mouse CB2
(Clayton et al., 2002). A similar result was found when nabilone receptors. AM1241 has been described as a protean agonist
was combined with SR144528 (Conti et al., 2002). Studies have implying the state of constitutive receptor activity can
also been conducted with CP55,940 and WIN55,212-2. Both determine the functional effect of a ligand-receptor interac-
compounds are analgesic in assays of acute pain; the effect of tion (Yao et al., 2006). Behaviorally, AM1241 did not elicit
CP55,940 is only partially blocked by CB1 receptor-selective deficits in the rotarod or catalepsy assays (Malan et al., 2001a).
antagonists (Scott et al., 2004) while that of WIN55,212-2 is AM1241 did however show antinociceptive effects towards an
reduced in CB2 receptor knockout animals compared to wild acute thermal stimulus (Hargreaves apparatus) when admi-
type, but remains in CB1 receptor knockout animals (Ibrahim nistered either systemically or locally; this effect could be
et al., 2006). CP55,940 administered either systemically or reversed with the CB2 receptor-selective antagonist, AM630
intrathecally has also proven efficacious against neuropathic (Malan et al., 2001a). AM1241 has also been shown to be anti-
pain in rats. Interestingly, only the antihyperalgesic effect of inflammatory and efficacious against inflammatory, neuro-
systemically administered CP55,940 could be partially blocked pathic and post-surgical pain, in addition to chemical-
by SR144528 (Scott et al., 2004). This result could be due to (capsaicin and formalin) induced pain and substance P-
insufficient CNS penetraton of the antagonist, or may imply induced plasma extravasation, when administered either
that the effects in the spinal cord are due to CB1 receptor locally or systemically (Malan et al., 2001b; Nackley et al.,
activation alone. In the same study, the dose-dependent 2003a; Quartilho et al., 2003; Hohmann et al., 2004; LaBuda
catalepsy that was observed was not blocked by a CB2 receptor et al., 2005). The effects of AM1241 are reversible with selective
antagonist, reinforcing the notion that side-effects track with CB2 receptor antagonists (AM630 and SR144528), were not
activation of CB1 receptors. Finally, WIN55,212-2 has been reversible with selective CB1 receptor antagonists (AM251 and
shown to inhibit carrageenan-induced allodynia and mechan- SR141716A), and remained in animals that were null mutants
ical hyperalgesia and burn-induced thermal and mechanical for the CB1 receptor (Ibrahim et al., 2003).
hyperalgesia after local administration, as well as bone cancer GW405833 (L-768,242) is a CB2 receptor agonist with a
pain and inflammatory muscle pain after systemic adminis- reported in vitro Ki value of 4–12 nM at recombinant human
tration (Nackley et al., 2003b; Kehl et al., 2003; Johanek and CB2 receptors (Gallant et al., 1996; Green et al., 1999;
Simone, 2004). All effects, except for muscle pain, were shown Valenzano et al., 2005). At recombinant human CB1 receptors,
to be at least partially blocked by CB2 receptor-selective GW405833 has reported Ki values ranging from 1900 to
antagonists. 4800 nM, leading to a 160–1200-fold selectivity (Gallant et al.,
Concerning studies investigating selective CB2 agonists, a 1996; Valenzano et al., 2005). GW405833 has shown agonist
relatively large number of the pharmacological reports that properties in both direct cAMP accumulation assays (Valen-
have been published on the role of CB2 receptors in pain have zano et al., 2005) and a cAMP reporter system. Against native
been generated using a limited number of selective agonists, rat CB2 receptors the Ki value is comparable to human, but the
namely HU-308, AM1241, GW405833, JWH 015 and JWH 133. compound is more potent at CB1 receptors compared to the
HU-308 is a THC derivative that is reported to have an in vitro human ortholog leading to reduced selectivity (80-fold)
B RA I N RE SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6 261

(Valenzano et al., 2005). In vivo, GW405833 reduces edema antagonist, GW818646, and exogenous 8-bromo cAMP, but
and inhibits hypersensitivity associated with intraplantar was unaffected in the presence of the opioid blocker,
injection of carrageenan (Clayton et al., 2002). These effects naloxone, or the CB1 receptor antagonist SR141716A. Studies
were inhibited by SR144528, providing evidence that the effect demonstrating the inhibition of sensory nerve activity by CB2
of GW405833 is mediated by CB2 receptors (Clayton et al., receptor activation in guinea pigs (Patel et al. 2003), and rat
2002). The compound (up to 30 mg/kg) elicits potent and models of acute and chronic pain (Ross et al. 2001, Nackley et
efficacious antihyperalgesic effects in the rat SNL and PSN al. 2004, Elmes et al. 2004, Sagar et al. 2005) also suggest direct
(Seltzer et al., 1990) models of neuropathic pain, as well as action on sensory neurons. In addition, there is evidence for
incisional, chemical-induced (formalin) and inflammatory the expression of CB2 receptors in keratinocytes, which in the
(CFA) pain (Valenzano et al., 2005; LaBuda et al., 2005; presence of CB2 agonists, release endogenous β-endorphins to
Beltramo et al., 2006). Analgesia, sedation and catalepsy activate opioid receptors in the peripheral nerve terminals of
were not seen in this dose range, but were apparent at sensory neurons (Ibrahim et al. 2005).
100 mg/kg (Valenzano et al., 2005). Comparable effects were Taken together these studies summarized above imply
seen in mouse models of inflammatory and neuropathic pain both a peripheral and spinal site of actions at different cell
but were absent in CB2 receptor knockout mice. The side types. Regarding clinical trials, GlaxoSmithKline has under-
effects and frank analgesia remained in these knockout taken a CB2 agonist Phase II trial for inflammatory pain.
animals suggesting a CB1 receptor-related mechanism of Pharmos is still considering clinical trials for their CB2-
action at these high doses (Whiteside et al., 2005). selective agonist PRS-639058 (structure not disclosed) as is
JWH 015 is a synthetic cannabinoid agonist with literature Glenmark with their most advanced candidate GRC 10693
Ki values of 14–54 nM at recombinant human CB2 receptors (structure not disclosed).
and 380 nM at recombinant human CB1 receptors (Showalter
et al., 1996; Aung et al., 2000; Mukherjee et al., 2004). At 3.3. Inhibitors of endocannabinoid metabolism
recombinant rat CB2 receptors, the compound is reported to be
less potent, with a Ki value of 150 nM (Mukherjee et al., 2004). The finding that formalin administration to the hindpaw of
Additionally, JWH 015 decreases FSK-mediated cAMP accu- rats produced a release of AEA into the periaqueductal gray
mulation in vitro (Mukherjee et al., 2004). It is perhaps due to (Walker et al., 1999) led those authors to suggest that
this limited selectivity for CB2 over CB1 receptors that this compounds preventing the metabolism of this endocannabi-
pharmacological tool has not been as extensively profiled in noid may be useful agents for the treatment of pain. This has
vivo compared to those compounds discussed earlier. Impor- subsequently been shown to be the case, in particular for
tantly however, when administered systemically it can reduce inflammatory pain. Thus, selective inhibitors of fatty-acid
microglial activation following infection with TMEV (Theiler's amide hydrolase such as URB597 (structure see Table 1) and
murine encephalomyelitis virus) in mice (Arévalo-Martin OL-135 produce beneficial effects in a variety of models of
et al., 2003), an effect that may indicate the mechanism of inflammatory pain, whereas their effects upon neuropathic
action of CB2 receptor agonists in neuropathic pain. Lastly, pain are less clear (Lichtman et al., 2004; Jayamanne et al.,
JWH 133 is a published CB2 receptor-selective compound with 2006; Chang et al., 2006; Russo et al., 2007a,b). Inhibitors of
a reported Ki value against recombinant human CB2 receptors FAAH and MGL also enhance endocannabinoid-mediated
of ∼ 3 nM and against native rat CB1 receptors of ∼ 670 nM analgesia that is induced by exposure to environmental
(Huffman et al., 1999). No in vitro functional data have been stressors and increase the bioavailability of anandamide
published for JWH 133. When administered systemically, JWH and/or 2-AG (Hohmann et al. 2005; Suplita et al., 2005).
133 can inhibit citric acid-induced cough in conscious guinea Importantly, FAAH inhibitors do not produce behaviors
pigs, possibly indicating a neuronal mechanism of action associated with a central activation of CB1 receptors (Kathuria
(Patel et al., 2003). In support of this conclusion JWH 133 has et al., 2003; Jayamanne et al., 2006) and do not appear to
been shown to decrease capsaicin-induced depolarizations of behave like THC in drug-discrimination tests (Gobbi et al.,
guinea pig and human vagus nerve (Patel et al., 2003). In 2005), although there may be some issues with respect to
addition, it has been shown to reverse hyperalgesia due to alcohol consumption (Vinod et al., 2008; but see Cippitelli et
intraplantar injection of carrageenan with a concurrent al., 2008).
reduction of edema (Elmes et al., 2005). Importantly, JWH 133 It should be emphasized that the notion that FAAH
has been shown to reduce the responses of wide dynamic inhibitors produce all their effects via a potentiation of
range dorsal horn neurons to both innocuous and noxious endocannabinoid signaling may be an oversimplification.
intensities of mechanical stimuli. This effect was seen in FAAH inhibitors also increase the bioavailability of endogen-
models of inflammatory and neuropathic pain, in addition to ous fatty-acid amides that are biologically active, but that do
sham operated animals and was partially blocked by the CB2 not bind to cannabinoid receptors, such as palmitoylethano-
receptor antagonist SR144528 (Elmes et al., 2004). A compar- lamide (PEA). PEA blocks both phases of the formalin response
able effect was observed following direct spinal application of in rats and mice, in addition to carrageenan-induced hyper-
JWH 133 (Sagar et al., 2005). algesia in rats, when applied locally or systemically
Functional studies in cultured human sensory neurons (Calignano et al., 1998, 2001; Jaggar et al., 1998; Conti et al.,
demonstrated CB2 agonists, GW842166 and GW833972, pro- 2002); an effect that was blocked by SR144528 (Calignano et al.,
duced inhibition of capsaicin-induced Ca2+ influx. Further 1998). PEA has also been shown to block acetic acid-induced
mechanistic studies showed CB2 agonist inhibition of capsai- visceral pain (Calignano et al., 2001), nerve growth factor
cin responses was reversed in the presence of the CB2 (NGF)-induced hyperalgesia, and accumulation of immune
262 B RA I N R E SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6

cells at the site of NGF injection; CB2 receptor-, but not CB1 could, by reducing the NSAID dose needed, provide an
receptor-selective antagonists blocked both effects (Farquhar- efficacious treatment strategy while minimizing the poten-
Smith and Rice, 2003). Given that PEA lacks direct effects upon tial for NSAID-induced gastrointestinal and cardiovascular
CB1 and CB2 receptors, it is possible that the effects of the disturbances. Other potential “partners” for endocannabinoid
compound are either indirect (via entourage effects, that modulatory agents include α2-adrenoceptor, peroxisome
increase the bioavailability of anandamide by competing with proliferator-activated receptor α agonists and TRPV1 antago-
the same enzymes for hydrolysis), or via CB2-like receptor nists (see e.g. Yoon and Choi, 2003; Tham et al., 2005; Russo
(review, see Lambert et al., 2002). However, recent data has et al., 2007a,b).
suggested that peroxisome-proliferator activated receptor α An extension of the polypharmacological approach above
(PPARα) may mediate at least some of the effects of PEA, and is to combine the desired pharmacological properties of the
that the ability of SR144528 to block PEA may be related to an treatment within a single molecule. Although this lacks the
off-target action of this compound upon PPARα (LoVerme et flexibility of dosing that is possible when separate drugs are
al., 2006). In this respect, Sagar et al. (in press) recently given, a major advantage is that the potential for variability of
reported that the responsiveness of wide dynamic range response due to inter-individual variations in the metabolism
neurons following carrageenan-injection into the hind paws of the two components relative to each other is eliminated
is attenuated by local injection of URB597 in a manner blocked (for a review of the concept of “designed multiple ligands”
by concomitant local injection of a PPAR-α, but not a CB1 see Morphy and Rankovic, 2005). Such compounds are
receptor antagonist. Indeed, the observation that the effects beginning to appear in the literature. For example, N-
of URB597 in the CFA model are partially blocked by arachidonoylserotonin is a TRPV1 (transient receptor poten-
rimonabant and SR144528 (the combination producing a tial vanilloid type 1) antagonist with FAAH inhibitory proper-
complete block) (Jayamanne et al., 2006) may reflect involve- ties that is active in a number of models of inflammatory and
ment of PPAR-α rather than CB2 receptors in addition to CB1 neuropathic pain (Maione et al., 2007). The ibuprofen analog
receptors. N-(3-methylpyridin-2-yl)-2-(4′-isobutylphenyl)propionamide,
Inhibitors of endocannabinoid uptake are also effective in a compound active in models of visceral pain (Cocco et al.,
models of inflammatory and neuropathic pain (see Maione 2003), is 2-3 orders of magnitude more potent an inhibitor of
et al., in press, for a recent article investigating the efficacy and FAAH than ibuprofen, while retaining its cyclooxygenase-
degree of CB-receptor involvement in a series of compounds inhibitory potency (Holt et al., 2007) and may be useful as a
with different relative potencies towards FAAH and AEA template for the design of potent FAAH/cyclooxygenase-2
uptake). The most potent compound so far described is inhibitors. Pravadoline, which has both cyclooxygenase
LY2318912, which produces good effects in the formalin inhibitory and cannabinoid receptor agonist properties
model (Moore et al., 2005). This compound, however, is a (D'Ambra et al., 1992), has greater analgesic efficacy than
potent FAAH inhibitor (Alexander and Cravatt, 2006), and so it NSAIDs such as zomepirac in several different tests for
is unclear as to whether its effects in the formalin test are due analgesia (Haubrich et al., 1990).
to blockade of AEA accumulation or its subsequent metabo-
lism. Indeed, the uncertainty concerning the mechanism(s) by
which AEA is accumulated into cells remains an important 5. Conclusions
issue that in the worst case could prevent development of a
potentially useful class of drugs. The aim of this review has been to highlight different
approaches whereby the endocannabinoid system can be
harnessed to produce novel analgesic drugs that lack the
4. Endocannabinoid modulating agents as a psychotropic effects that place a limit upon the usefulness of
component of new pharmacotherapies for pain THC. Lead discovery and lead optimization activities at
numerous industrial and academic pharmacology/chemistry
The approaches outlined in section 3 have all been laboratories has led to the identification of a number of novel,
considered per se, but a useful approach may be the selective and peripherally restricted modulators of the endo-
combination of these actions with other drugs, with the cannabinoid system highlighted here. The preclinical profile
aim of either improving efficacy or providing a better safety of a number of these proof-of-concept compounds highlighted
profile than that seen with currently available analgesics. An here is extremely encouraging, and it is hoped that these
obvious combination is that of cannabinoids and opioids, potent, selective and efficacious compounds will translate to
given that some of the antinociceptive effects of cannabi- the clinic.
noids involve activation of the opioid system and vice versa
(Ibrahim et al., 2005; da Fonseca Pacheco et al., 2008). REFERENCES
Moreover, CB receptor agonists enhance the effect of μ-
opioid receptor agonists in a variety of models of analgesia
(Reche et al., 1996; Yesilyurt et al., 2003; Finn et al., 2004; Agarwal, N., Pacher, P., Tegeder, I., Amaya, F., Constantin, C.E.,
Brenner, G.J., Rubino, T., Michalski, C.W., Marsicano, G.,
Tham et al., 2005; Cox et al., 2007). Activation of CB receptors
Monory, K., Mackie, K., Marian, C., Batkai, S., Parolaro, D.,
either directly or indirectly also increases the analgesic effect
Fischer, M.J., Reeh, P., Kunos, G., Kress, M., Lutz, B., Woolf, C.J.,
of non-steroidal anti-inflammatory drugs (NSAIDs) (Guindon Kuner, R., 2007. Cannabinoids mediate analgesia largely via
et al., 2006a,b; Ulugöl et al., 2006; Naidu and Lichtman, 2007), peripheral type 1 cannabinoid receptors in nociceptors. Nat.
raising the possibility that a combination of suitable agents Neurosci. 10, 870–879.
B RA I N RE SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6 263

Alexander, J.P., Cravatt, B.F., 2006. The putative endocannabinoid D Ambra, T.E., Estep, K.G., Bell, M.R., Eissenstat, M.A., Josef, K.A.,
transport blocker LY2183240 is a potent inhibitor of FAAH and Ward, S.J., Haycock, D.A., Baizman, E.R., Casiano, F.M., Beglin,
several other brain serine hydrolases. J. Am. Chem. Soc. 128, N.C., Chippari, S.M., Grego, J.D., Kullnig, R.K., Daley, G.T., 1992.
9699–9704. Conformationally restrained analogues of pravadoline:
Anand, U., Otto, W.R., Sanchez-Herrera, D., Facer, P., Yiangou, Y., nanomolar potent, enantioselective, (aminoalkyl) indole
Korchev, Y., Birch, R., Benham, C., Bountra, C., Chessell, I.P., agonists of the cannabinoid receptor. J. Med. Chem. 35, 124–135.
Anand, P., 2008. Cannabinoid receptor CB2 localisation and da Fonseca Pacheco, D., Klein, A., de Castro Perez, A., da Fonseca
agonist-mediated inhibition of capsaicin responses in human Pacheco, C.M., de Francischi, J.N., Duarte, I.D., 2008. The
sensory neurons. Pain 138, 667–680. 1-opioid receptor agonist morphine, but not agonists at
Arévalo-Martin, N., Vela, J.M., Molina-Holgado, E., Borrell, J., Guaza, d- or j-opioid receptors, induces peripheral antinociception
C., 2003. Therapeutic action of cannabinoids in a murine model mediated by cannabinoid receptors. Br. J. Pharmacol. 154,
of multiple sclerosis. J. Neurosci. 23, 2511–2516. 1143–1149.
Aung, M.M., Griffin, G., Huffman, J.W., Wu, M.J., Keel, C., Yang, B., Di Marzo, V., Bifulco, M., De Petrocellis, L., 2004. The
Showalter, V.M., Abood, M.E., Martin, B.R., 2000. Influence endocannabinoid system and its therapeutic exploitation.
of the N-1 alkyl chain length of cannabimimetic indoles upon Nat. Rev. Drug Discov. 3 (9), 771–784.
CB1 and CB2 receptor binding. Drug Alcohol Depend. 60, Dziadulewicz, E.K., Bevan, S.J., Brain, C.T., Coote, P.R., Culshaw,
133–140. A.J., Davis, A.J., Edwards, L.J., Fisher, A.J., Fox, A.J., Gentry, C.,
Barann, M., Molderings, G., Brüss, M., Bönisch, H., Urban, B.W., Groarke, A., Hart, T.W., Huber, W., James, I.F., Kesingland, A., La
Göthert, M., 2002. Direct inhibition by cannabinoids of human Vecchia, L., Loong, Y., Lyothier, I., McNair, K., O, Farrell, C.,
5-HT3A receptors: probable involvement of an allosteric Peacock, M., Portmann, R., Schopfer, U., Yaqoob, M., Zadrobilek,
modulatory site. Br. J. Pharmacol. 137, 589–596. J., 2007. Naphthalen-1-yl-(4-pentyloxynaphthalen-1-yl)
Beltramo, M., Bernardini, N., Bertorelli, R., Campanella, M., methanone: a potent, orally bioavailable human CB1/CB2 dual
Nicolussi, E., Fredduzzi, S., Reggiani, A., 2006. CB2 agonist with antihyperalgesic properties and restricted central
receptor-mediated antihyperalgesia: possible direct nervous system penetration. J. Med. Chem. 50, 3851–3856.
involvement of neural mechanisms. Eur. J. Neurosci. 23, Elmes, S.J.R., Jhaveri, M.D., Smart, D., Kendall, D.A., Chapman, V.,
1530–1538. 2004. Cannabinoid CB2 receptor activation inhibits
Bennett, G.J., Xie, Y.K., 1988. A peripheral mononeuropathy in rat mechanically evoked responses of wide dynamic range dorsal
that produces disorders of pain sensation like those seen in horn neurons in naïve rats and in rat models of inflammatory
man. Pain 33, 87–107. and neuropathic pain. Eur. J. Neurosci. 20, 2311–2320.
Bradshaw, H.B., Walker, J.M., 2005. The expanding field of Elmes, S.J., Winyard, L.A., Medhurst, S.J., Clayton, N.M., Wilson, A.W.,
cannabimimetic and related lipid mediators. Br. J. Pharmacol. Kendall, D.A., Chapman, V., 2005. Activation of CB1 and CB2
144, 459–465. receptors attenuates the induction and maintenance of
Brown, A.J., 2007. Novel cannabinoid receptors. Br. J. Pharmacol. inflammatory pain in the rat. Pain 118 (3), 327–335.
152, 567–575. Farquhar-Smith, W.P., Rice, A.S., 2003. A novel neuroimmune
Calignano, A., La Rana, G., Giuffrida, A., Piomelli, D., 1998. Control mechanism in cannabinoid-mediated attenuation of nerve
of pain initiation by endogenous cannabinoids. Nature 394, growth factor-induced hyperalgesia. Anesthesiology 99 (6),
277–281. 1391–1401.
Calignano, A., La Rana, G., Piomelli, D., 2001. Antinociceptive Finn, D.P., Beckett, S.R., Roe, C.H., Madjd, A., Fone, K.C., Kendall, D.A.,
activity of the endogenous fatty acid amide, Marsden, C.A., Chapman, V., 2004. Effects of coadministration of
palmitylethanolamide. Eur. J. Pharmacol. 419, 191–198. cannabinoids and morphine on nociceptive behaviour, brain
Carlisle, S.J., Marciano-Cabral, F., Staab, A., Ludwick, C., Cabral, monoamines and HPA axis activity in a rat model of persistent
G.A., 2002. Differential expression of the CB2 cannabinoid pain. Eur. J. Neurosci. 19, 678–686.
receptor by rodent macrophages and macrophage-like Fowler, C.J., 2008. “The tools of the trade”—an overview of the
cells in relation to cell activation. Int. Immunopharmacol. 2, pharmacology of the endocannabinoid system. Curr. Pharm.
69–82. Des. 14, 2254–2265.
Chang, L., Luo, L., Palmer, J.A., Sutton, S., Wilson, S.J., Barbier, A.J., Fride, E., Feigin, C., Ponde, D.E., Breuer, A., Hanus, L., Arshavsky, N.,
Breitenbucher, J.G., Chaplan, S.R., Webb, M., 2006. Inhibition of Mechoulam, R., 2004. (+)-Cannabidiol analogues which bind
fatty acid amide hydrolase produces analgesia by multiple cannabinoid receptors but exert peripheral activity only. Eur. J.
mechanisms. Br. J. Pharmacol. 148, 102–113. Pharmacol. 506, 179–188.
Cippitelli, A., Cannella, N., Braconi, S., Duranti, A., Tontini, A., Gallant, M., Dufresne, C., Gareau, Y., Guay, D., Leblanc, Y., Prasit, P.,
Bilbao, A., Defonseca, F.R., Piomelli, D., Ciccocioppo, R., 2008. Rochette, C., Sawyer, N., Slipetz, D.M., Tremblay, N., Metters, K.
Increase of brain endocannabinoid anandamide levels by M., Labelle, M., 1996. New class of potent ligands for the human
FAAH inhibition and alcohol abuse behaviours in the rat. peripheral cannabinoid receptor. Bioorg. Med. Chem. Lett. 6,
Psychopharmacology 198, 449–460 (Berl.). 2263–2268.
Clayton, N., Marshall, F.H., Bountra, C., O, Shaughnessy, C.T., 2002. Gobbi, G., Bambico, F.R., Mangieri, R., Bortolato, M., Campolongo,
CB1 and CB2 cannabinoid receptors are implicated in P., Solinas, M., Cassano, T., Morgese, M.G., Debonnel, G.,
inflammatory pain. Pain 96, 253–260. Duranti, A., Tontini, A., Tarzia, G., Mor, M., Trezza, V., Goldberg,
Cocco, M.T., Congiu, C., Onnis, V., Morelli, M., Cauli, O., 2003. S.R., Cuomo, V., Piomelli, D., 2005. Antidepressant-like activity
Synthesis of ibuprofen heterocyclic amides and investigation and modulation of brain monoaminergic transmission by
of their analgesic and toxicological properties. Eur. J. Med. blockade of anandamide hydrolysis. Proc. Natl. Acad. Sci.
Chem. 38, 513–518. U. S. A. 102, 18620–18625.
Conti, S., Costa, B., Colleoni, M., Parolaro, D., Giagnoni, G., 2002. Green, A., O'Shaughnessy, C., Disney, G., Marshall, F., 1999.
Antiinflammatory action of endocannabinoid Reporter assays for human cannabinoid CB1 and CB2 receptors
palmitoylethanolamide and the synthetic cannabinoid for the identification of novel agonists. Br. J. Pharmacol. 126
nabilone in a model of acute inflammation in the rat. Br. J. Proc. Suppl. p. 112P.
Pharmacol. 135, 181–187. Guindon, J., De Léan, A., Beaulieu, P., 2006a. Local interactions
Cox, M.L., Haller, V.L., Welch, S.P., 2007. Synergy between between anandamide, an endocannabinoid, and ibuprofen, a
Δ9-tetrahydrocannabinol and morphine in the arthritic rat. nonsteroidal anti-inflammatory drug, in acute and
Eur. J. Pharmacol. 567, 125–130. inflammatory pain. Pain 121, 85–93.
264 B RA I N R E SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6

Guindon, J., LoVerme, J., De Léan, A., Piomelli, D., Beaulieu, P., the putative CB2 receptor agonist palmitoylethanolamide
2006b. Synergistic antinociceptive effects of anandamide, an in visceral and somatic inflammatory pain. Pain 76, 189–199.
endocannabinoid, and nonsteroidal anti-inflammatory drugs Jayamanne, A., Greenwood, R., Mitchell, V.A., Aslan, S., Piomelli,
in peripheral tissue: a role for endogenous fatty-acid D., Vaughan, C.W., 2006. Actions of the FAAH inhibitor URB597
ethanolamides? Eur. J. Pharmacol. 550, 68–77. in neuropathic and inflammatory chronic pain models. Br. J.
Guindon, J., Desroches, J., Beaulieu, P., 2007. The antinociceptive Pharmacol. 147, 281–288.
effects of intraplantar injections of 2-arachidonoyl glycerol are Johanek, L.M., Simone, D.A., 2004. Activation of peripheral
mediated by cannabinoid CB2 receptors. Br. J. Pharmacol. 150, cannabinoid receptors attenuates cutaneous hyperalgesia
693–701. produced by a heat injury. Pain 109, 432–442.
Gutierrez, T., Farthing, J.N., Zvonok, A.M., Makriyannis, A., Kathuria, S., Gaetani, S., Fegley, D., Valiño, F., Duranti, A., Tontini,
Hohmann, A.G., 2007. Activation of peripheral cannabinoid CB1 A., Mor, M., Tarzia, G., La Rana, G., Calignano, A., Giustino, A.,
and CB2 receptors suppresses the maintenance of Tattoli, M., Palmery, M., Cuomo, V., Piomelli, D., 2003.
inflammatory nociception: a comparative analysis. Br. J. Modulation of anxiety through blockade of anandamide
Pharmacol. 150, 153–163. hydrolysis. Nat. Med. 9, 76–81.
Hanuš, L., Breuer, A., Tchilibon, S., Shiloah, S., Goldenberg, D., Kehl, L.J., Hamamoto, D.T., Wacnik, P.W., Croft, D.L., Norsted, B.D.,
Horowitz, M., Pertwee, R.G., Ross, R.A., Mechoulam, R., Fride, E., Wilcox, G.L., Simone, D.A., 2003. A cannabinoid agonist
1999. HU-308: a specific agonist for CB2, a peripheral differentially attenuates deep tissue hyperalgesia in animal
cannabinoid receptor. Proc. Natl. Acad. Sci. U. S. A. 96, models of cancer and inflammatory muscle pain. Pain 103,
14228–14233. 175–186.
Haubrich, D.R., Ward, S.J., Baizman, E., Bell, M.R., Bradford, J., Kim, S.H., Chung, J.M., 1992. An experimental model for peripheral
Ferrari, R., Miller, M., Perrone, M., Pierson, A.K., Saelens, J.K., neuropathy produced by segmental spinal nerve ligation in the
Luttinger, D., 1990. Pharmacology of pravadoline: a new rat. Pain 50, 355–363.
analgesic agent. J. Pharmacol. Exp. Ther. 255, 511–522. LaBuda, C.J., Koblish, M., Little, P.J., 2005. Cannabinoid CB2 receptor
Hohmann, A.G., 2002. Spinal and peripheral mechanisms of agonist activity in the hindpaw incision model of postoperative
cannabinoid antinociception: behavioral, neurophysiological pain. Eur. J. Pharmacol. 527, 172–174.
and neuroanatomical perspectives. Chem. Phys. Lipids 121, Lambert, D.M., Vandevoorde, S., Jonsson, K.-O., Fowler, C.J., 2002.
173–190. The palmitoylethanolamide family: a new class of
Hohmann, A.G., Herkenham, M., 1999. Localization of central anti-inflammatory agents? Curr. Med. Chem. 9, 663–674.
cannabinoid CB1 receptor messenger RNA in neuronal Lichtman, A.H., Leung, D., Shelton, C.C., Saghatelian, A., Hardouin,
subpopulations of rat dorsal root ganglia: a double-label in situ C., Boger, D.L., Cravatt, B.F., 2004. Reversible inhibitors of fatty
hybridization study. Neuroscience 90 (3), 923–931. acid amide hydrolase that promote analgesia: evidence for an
Hohmann, A.G., Suplita, R.L., Bolton, N.M., Neely, M.H., Fegley, D., unprecedented combination of potency and selectivity.
Mangieri, R., Krey, J.F., Walker, J.M., Holmes, P.V., Crystal, J.D., J. Pharmacol. Exp. Ther. 311, 441–448.
Duranti, A., Tontini, A., Mor, M., Tarzia, G., Piomelli, D., 2005. An LoVerme, J., Russo, R., La Rana, G., Fu, J., Farthing, J., Mattace-Raso,
endocannabinoid mechanism for stress-induced analgesia. G., Meli, R., Hohmann, A., Calignano, A., Piomelli, D., 2006.
Nature 435 (7045), 1108–1112. Rapid broad-spectrum analgesia through activation of
Hohmann, A.G., Farthing, J.N., Zvonok, A.M., Makriyannis, A., 2004. peroxisome proliferator-activated receptor-. J. Pharmacol. Exp.
Selective activation of cannabinoid CB2 receptors suppresses Ther. 319, 1051–1061.
hyperalgesia evoked by intradermal capsaicin. J. Pharmacol. Maione, S., De Petrocellis, L., de Novellis, V., Moriello, A.S.,
Exp. Ther. 308, 446–453. Petrosino, S., Palazzo, E., Rossi, F.S., Woodward, D.F., Di Marzo,
Hohmann, A.G., Suplita, R.L., 2nd., 2006. Endocannabinoid V., 2007. Analgesic actions of N-arachidonoyl-serotonin,
mechanisms of pain modulation. AAPS J. 8 (4), E693–E708. a fatty acid amide hydrolase inhibitor with antagonistic
Holt, S., Paylor, B., Boldrup, L., Alajakku, K., Vandevoorde, S., activity at vanilloid TRPV1 receptors. Br. J. Pharmacol. 150,
Sundström, A., Cocco, M.T., Onnis, V., Fowler, C.J., 2007. 766–781.
Inhibition of fatty acid amide hydrolase, a key Maione, S., Morera, E., Marabese, I., Ligresti, A., Luongo, L., Ortar,
endocannabinoid metabolizing enzyme, by analogues of G., Di Marzo, V., in press. Antinociceptive effects of tetrazole
ibuprofen and indomethacin. Eur. J. Pharmacol. 565, 26–36. inhibitors of endocannabinoid inactivation: cannabinoid
Huffman, J.W., Liddle, J., Yu, S., Aung, M.M., Abood, M.E., Wiley, J.L., and non-cannabinoid receptor-mediated mechanisms.
Martin, B.R., 1999. 3-(1′,1′.Dimethylbutyl)-1-deoxy-Δ8-THC and Br. J. Pharmacol. 155 (5), 775–782.
related compounds: synthesis of selective ligands for the CB2 Malan, T.P., Ibrahim, M.M., Deng, H.F., Liu, Q., Mata, H.P.,
receptor. Bioorg. Med. Chem. 7, 2905–2914. Vanderah, T., Porreca, F., Makriyannis, A., 2001a. CB2
Ibrahim, M.M., Deng, H.F., Zvonok, A., Cockayne, D.A., Kwan, J., cannabinoid receptor-mediated peripheral antinociception.
Mata, H.P., Vanderah, T.W., Lai, J., Porreca, F., Makriyannis, A., Pain 93, 239–245.
Malan, T.P., 2003. Activation of CB2 cannabinoid receptors by Malan, T.P., Ibrahim, M.M., Deng, H., Makriyannis, A., Vanderah, T.
AM1241 inhibits experimental neuropathic pain: pain W., 2001b. Anti-inflammatory effects of the CB2 cannabinoid
inhibition by receptors not present in the CNS. Proc. Natl. Acad. receptor-selective agonist AM1241. Anesthesiology 95, A894.
Sci. U. S. A. 100, 10529–10533. Matsuda, L.A., Lolait, S.J., Brownstein, M.J., Young, A.C., Bonner, T.I.,
Ibrahim, M.M., Porreca, F., Lai, J., Albrecht, P.J., Rice, F.L., 1990. Structure of a cannabinoid receptor and functional
Khodorova, A., Davar, G., Makriyannis, A., Vanderah, T.W., expression of the cloned cDNA. Nature 346 (6284), 561–564.
Mata, H.P., Malan, T.P., 2005. CB2 cannabinoid receptor Mechoulam, R., 1986. In: Mechoulam, R. (Ed.), The Pharmacohis-
activation produces antinociception by stimulating peripheral tory of Cannabis Sativa in Cannabinoids as
release of endogenous opioids. Proc. Natl. Acad. Sci. U. S. A. 102, Theapeutic Agents. CRC, Boca Raton, pp. 1–19.
3093–3098. Monory, K., Blaudzun, H., Massa, F., Kaiser, N., Lemberger, T.,
Ibrahim, M.M., Rude, M.L., Stagg, N.J., Mata, H.P., Lai, J., Vanderah, Schütz, G., Wotjak, C.T., Lutz, B., Marsicano, G., 2007. Genetic
T.W., Porreca, F., Buckley, N.E., Makriyannis, A., Malan, T.P., dissection of behavioural and autonomic effects of
2006. CB2 cannabinoid receptor mediation of nociception. Pain Δ9-tetrahydrocannabinol in mice. PLoS Biol. 5, e269.
122, 36–42. Moore, S.A., Nomikos, G.G., Dickason-Chesterfield, A.K., Schober,
Jaggar, S.I., Hasnie, F.S., Sellaturay, S., Rice, A.S.C., 1998. The D.A., Schaus, J.M., Ying, B.P., Xu, Y.C., Phebus, L., Simmons,
anti-hyperalgesic actions of the cannabinoid anandamide and R.M., Li, D., Iyengar, S., Felder, C.C., 2005. Identification of a
B RA I N RE SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6 265

high-affinity binding site involved in the transport of Russo, R., LoVerme, J., La Rana, G., D, Agostino, G., Sasso, O.,
endocannabinoids. Proc. Natl. Acad. Sci. U. S. A. 102, Calignano, A., Piomelli, D., 2007b. Synergistic antinociception
17852–17857. by the cannabinoid receptor agonist anandamide and the
Morphy, R., Rankovic, Z., 2005. Designed multiple ligands. An PPAR-α receptor agonist GP7647. Eur. J. Pharmacol. 566, 117–119.
emerging drug discovery paradigm. J. Med. Chem. 48, Sagar, D.R., Kelly, S., Millns, P.J., O, Shaughnessey, C.T., Kendall,
6523–6543. D.A., Chapman, V., 2005. Inhibitory effects of CB1 and CB2
Mukherjee, S., Adams, M., Whiteaker, K., Daza, A., Kage, K., Cassar, receptor agonists on responses of DRG neurons and
S., Meyer, M., Yao, B.B., 2004. Species comparison and dorsal horn neurons in neuropathic rats. Eur. J. Neurosci. 22,
pharmacological characterization of rat and human CB2 371–379.
cannabinoid receptors. Eur. J. Pharmacol. 505, 1–9. Sagar, D.R., Kendall, D.A., Chapman, V., 2008. Inhibition of fatty acid
Munro, S., Thomas, K.L., Abu-Shaar, M., 1993. Molecular amide hydrolase produces PPAR-α-mediated analgesia in a rat
characterization of a peripheral receptor for cannabinoids. model of inflammatory pain. Br. J. Pharmacol. 155 (8), 1297–1306.
Nature 365 (6441), 61–65. Scott, D.A., Wright, C.E., Angus, J.A., 2004. Evidence that CB-1 and
Nackley, A.G., Suplita, R.L., Hohmann, A.G., 2003a. A peripheral CB-2 cannabinoid receptors mediate antinociception in
cannabinoid mechanism suppresses spinal Fos protein neuropathic pain in the rat. Pain 109, 124–131.
expression and pain behavior in a rat model of inflammation. Seltzer, Z., Dubner, R., Shir, Y., 1990. A novel behavioral model of
Neuroscience 117, 659–670. neuropathic pain disorders produced in rats by partial sciatic
Nackley, A.G., Makriyannis, A., Hohmann, A.G., 2003b. Selective nerve injury. Pain 43, 205–218.
activation of cannabinoid CB2 receptors suppresses spinal fos Showalter, V.M., Compton, D.R., Martin, B.R., Abood, M.E., 1996.
protein expression and pain behavior in a rat model of Evaluation of binding in a transfected cell line expressing a
inflammation. Neuroscience 119 (3), 747–757. peripheral cannabinoid receptor (CB2): identification of
Nackley, A.G., Zvonok, A.M., Makriyannis, A., Hohmann, A.G., cannabinoid receptor subtype selective ligands. J. Pharmacol.
2004. Activation of cannabinoid CB2 receptors suppresses Exp. Ther. 278, 989–999.
C-fiber responses and windup in spinal wide dynamic range Smith, F.L., Fujimori, K., Lowe, J., Welch, S.P., 1998.
neurons in the absence and presence of inflammation. Characterization of Δ9-tetrahydrocannabinol and anandamide
J. Neurophysiol. 92, 3562–3574. antinociception in nonarthritic and arthritic rats. Pharmacol.
Naidu, P.S., Lichtman, A.H., 2007. Synergistic antinociceptive Biochem. Behav. 60, 183–191.
effects of URB597 and diclofenac in a mouse visceral pain Sokal, D.M., Elmes, S.J.R., Kendall, D.A., Chapman, V., 2003.
model. 17th Annual Symposium on the Cannabinoids, Intraplantar injection of anandamide inhibits
Burlington, Vermont, International Cannabinoid Research mechanically-evoked responses of spinal neurons via
Society, #172. Available online at http://cannabinoidsociety. activation of CB2 receptors in anaesthetised rats.
org/SYMPOSIUM.2007/2007.ICRS.Program.and.Abstracts.pdf Neuropharmacology 45, 404–411.
Onaivi, E.S., Ishiguro, H., Gong, J.P., Patel, S., Perchuk, A., Meozzi, Staton, P.C., Hatcher, J.P., Walker, D.J., Morrison, A.D., Shapland,
P.A., Myers, L., Mora, Z., Tagliaferro, P., Gardner, E., Brusco, A., E.M., Hughes, J.P., Chong, E., Mander, P.K., Green, P.J., Billinton,
Akinshola, B.E., Liu, Q.R., Hope, B., Iwasaki, S., Arinami, T., A., Fulleylove, M., Lancaster, H.C., Smith, J.C., Bailey, L.T., Wise,
Teasenfitz, L., Uhl, G.R., 2006. Discovery of the presence and A., Brown, A.J., Richardson, J.C., Chessell, I.P., 2008. The
functional expression of cannabinoid CB2 receptors in brain. putative cannabinoid receptor GPR55 plays a role in
Ann. N.Y. Acad. Sci. 1074, 514–536. mechanical hyperalgesia associated with inflammatory and
Patel, H.J., Birrell, M.A., Crispino, N., Hele, D.J., Venkatesan, P., neuropathic pain. Pain 139, 225–236.
Barnes, P.J., Yacoub, M.H., Belvisi, M.G., 2003. Inhibition of Suplita, R.L., Farthing 2nd, J.N., Gutierrez, T., Hohmann, A.G., 2005.
guinea-pig and human sensory nerve activity and the cough Inhibition of fatty-acid amide hydrolase enhances cannabinoid
reflex in guinea-pigs by cannabinoid (CB2) receptor activation. stress-induced analgesia: sites of action in the dorsolateral
Br. J. Pharmacol. 140, 261–268. periaqueductal gray and rostral ventromedial medulla.
Perez, J., Ribera, M.V., 2008. Managing neuropathic pain with Neuropharmacology 49 (8), 1201–1209.
SativexR: a review of its pros and cons. Expert Opin. Tham, S.M., Angus, J.A., Tudor, E.M., Wright, C.E., 2005. Synergistic
Pharmacother. 9, 1189–1195. and additive interactions of the cannabinoid agonist CP55,940
Price, T.J., Helesic, G., Parghi, D., Hargreaves, K.M., Flores, C.M., with μ opioid receptor and α2-adrenoceptor agonists in acute
2003. The neuronal distribution of cannabinoid receptor type 1 pain models in mice. Br. J. Pharmacol. 144, 875–884.
in the trigeminal ganglion of the rat. Neuroscience 120 (1), Tokanovic, S., Malone, D.T., Ventura, S., 2007. Stimulation of
155–162. epithelial CB1 receptors inhibits contractions of the rat prostate
Quartilho, A., Mata, H.P., Ibrahim, M.M., Vanderah, T.W., Porreca, gland. Br. J. Pharmacol. 150, 227–234.
F., Makriyannis, A., Malan, T.P., 2003. Inhibition of Ulugöl, A., Ozyigit, F., Yesilyurt, O., Dogrul, A., 2006. The additive
inflammatory hyperalgesia by activation of peripheral CB2 antinociceptive interaction between WIN 55,212-2, a
cannabinoid receptors. Anesthesiology 99, 955–960. cannabinoid agonist, and ketorolac. Anesth. Analg. 102,
Reche, I., Fuentes, J.A., Ruiz-Gayo, M., 1996. Potentiation of 443–447.
Δ9-tetrahydrocannabinol-induced analgesia by morphine in Valenzano, K.J., Tafesse, L., Lee, G., Harrison, J.E., Boulet, J.M.,
mice: involvement of μ- and κ-opioid receptors. Eur. J. Gottshall, S.L., Mark, L., Pearson, M.S., Miller, W., Shan, S.,
Pharmacol. 318, 11–16. Rabadi, L., Rotshteyn, Y., Chaffer, S.M., Turchin, P.I., Elsemore,
Ross, R.A., Coutts, A.A., McFarlane, C.M., Anavi-Goffer, S., Irving, A.J., D.A., Toth, M., Koetzner, L., Whiteside, G.T., 2005.
Pertwee, R.G., MacEwan, D.J., Scott, R.H., 2001. Actions of Pharmacological and pharmakokinetic characterization of the
cannabinoid receptor ligands on rat cultured sensory neurones: cannabinoid receptor 2 agonist, GW405833, utilizing rodent
implications for antinociception. Neuropharmacology 40 (2), models of acute and chronic pain, anxiety, ataxia and
221–232. catalepsy. Neuropharmacology 48, 658–672.
Russo, R., LoVerme, J., La Rana, G., Compton, T.R., Parrott, J., Vinod, K.Y., Sanguino, E., Yalamanchili, R., Manzanares, J.,
Duranti, A., Tontini, A., Mor, M., Tarzia, G., Calignano, A., Hungund, B.L., 2008. Manipulation of fatty acid amide
Piomelli, D., 2007a. The fatty acid amide hydrolase inhibitor hydrolase functional activity alters sensitivity and dependence
URB597 (cyclohexylcarbamic acid 3′-carbamoylbiphenyl-3-yl to ethanol. J. Neurochem. 104, 233–243.
ester) reduces neuropathic pain after oral administration in Walker, J.M., Hohmann, A.G., 2005. Cannabinoid mechanisms of
mice. J. Pharmacol. Exp. Ther. 322, 236–242. pain suppression. Handb. Exp. Pharmacol. (168), 509–554.
266 B RA I N R E SE A R CH RE V I EW S 60 ( 20 0 9 ) 2 5 5–2 6 6

Walter, L., Stella, N., 2004. Cannabinoids and neuroinflammation. Wensink, E.J., Honore, P., Sullivan, J.P., Dart, M.J., Meyer, M.D.,
Br. J. Pharmacol. 141, 775–785. 2008. In vitro and in vivo characterization of A-796260: a
Walker, J.M., Huang, S.M., Strangman, N.M., Tsou, K., selective cannabinoid CB2 receptor agonist exhibiting
Sañudo-Peña, M.C., 1999. Pain modulation by release of the analgesic activity in rodent pain models. Br. J. Pharmacol.
endogenous cannabinoid anandamide. Proc. Natl. Acad. Sci. 153, 390–401.
U. S. A. 96, 12198–12203. Yesilyurt, O., Dogrul, A., Gul, H., Seyrek, M., Kusmez, O., Ozkan, Y.,
Whiteside, G.T., Gottshall, S.L., Boulet, J.M., Chaffer, S.M., Harrison, Yildiz, O., 2003. Topical cannabinoid enhances topical
J.E., Pearson, M.S., Turchin, P.I., Mark, L., Garrison, A.E., morphine antinociception. Pain 105, 303–308.
Valenzano, K.J., 2005. A role for cannabinoid receptors, Yiangou, Y., Facer, P., Durrenberger, P., Chessell, I.P., Naylor, A.,
but not endogenous opioids, in the antinociceptive activity of Bountra, C., Banati, R., Anand, P., 2006. COX-2, CB2 and
the CB2-selective agonist, GW405833. Eur. J. Pharmacol. 528, P2X7-immunoreactivities are increased in activated microglial
65–72. cells/macrophages of multiple sclerosis and amyotrophic
Wotherspoon, G., Fox, A., McIntyre, P., Colley, S., Bevan, S., Winter, lateral sclerosis spinal cord. BMC Neurol. 6, 12.
J., 2005. Peripheral nerve injury induces cannabinoid receptor Yoon, M.H., Choi, J.I., 2003. Pharmacologic interaction between
2 protein expression in rat sensory neurons. Neuroscience 135, cannabinoid and either clonidine or neostigmine in the rat
235–245. formalin test. Anesthesiology 99, 701–707.
Yao, B.B., Mukherjee, S., Fan, Y., Garrison, T.R., Daza, A.V., Zhang, J., Hoffert, C., Vu, H.K., Groblewski, T., Ahmad, S., O,
Grayson, G.K., Hooker, B.A., Dart, M.J., Sullivan, J.P., Meyer, Donnell, D., 2003. Induction of CB2 receptor expression in the
M.D., 2006. In vitro pharmacological characterization of rat spinal cord of neuropathic but not inflammatory chronic
AM1241: a protean agonist at the cannabinoid CB2 receptor? pain models. Eur. J. Neurosci. 17, 2750–2754.
Br. J. Pharmacol. 149, 145–154. Zias, J., Stark, H., Sellgman, J., Levy, R., Werker, E., Breuer, A.,
Yao, B.B., Hsieh, G.C., Frost, J.M., Fan, Y., Garrison, T.R., Daza, A.V., Mechoulam, R., 1993. Early medical use of cannabis. Nature
Grayson, G.K., Zhu, C.Z., Pai, M., Chandran, P., Salyers, A.K., 363, 215.

You might also like