Skip to main content
Philosophical Transactions of the Royal Society B: Biological Sciences logoLink to Philosophical Transactions of the Royal Society B: Biological Sciences
. 2012 Dec 5;367(1607):3353–3363. doi: 10.1098/rstb.2011.0381

Targeting the endocannabinoid system with cannabinoid receptor agonists: pharmacological strategies and therapeutic possibilities

Roger G Pertwee 1,*
PMCID: PMC3481523  PMID: 23108552

Abstract

Human tissues express cannabinoid CB1 and CB2 receptors that can be activated by endogenously released ‘endocannabinoids’ or exogenously administered compounds in a manner that reduces the symptoms or opposes the underlying causes of several disorders in need of effective therapy. Three medicines that activate cannabinoid CB1/CB2 receptors are now in the clinic: Cesamet (nabilone), Marinol (dronabinol; Δ9-tetrahydrocannabinol (Δ9-THC)) and Sativex (Δ9-THC with cannabidiol). These can be prescribed for the amelioration of chemotherapy-induced nausea and vomiting (Cesamet and Marinol), stimulation of appetite (Marinol) and symptomatic relief of cancer pain and/or management of neuropathic pain and spasticity in adults with multiple sclerosis (Sativex). This review mentions several possible additional therapeutic targets for cannabinoid receptor agonists. These include other kinds of pain, epilepsy, anxiety, depression, Parkinson's and Huntington's diseases, amyotrophic lateral sclerosis, stroke, cancer, drug dependence, glaucoma, autoimmune uveitis, osteoporosis, sepsis, and hepatic, renal, intestinal and cardiovascular disorders. It also describes potential strategies for improving the efficacy and/or benefit-to-risk ratio of these agonists in the clinic. These are strategies that involve (i) targeting cannabinoid receptors located outside the blood-brain barrier, (ii) targeting cannabinoid receptors expressed by a particular tissue, (iii) targeting upregulated cannabinoid receptors, (iv) selectively targeting cannabinoid CB2 receptors, and/or (v) adjunctive ‘multi-targeting’.

Keywords: Δ9-tetrahydrocannabinol, cannabinoid CB1 and CB2 receptors, cannabinoid receptor agonists, therapeutic applications and strategies, blood-brain barrier

1. Introduction

The endocannabinoid system consists of at least two types of G-protein-coupled receptor, cannabinoid CB1 and CB2 receptors, of endogenous agonists for these receptors that are known as ‘endocannabinoids’ and include anandamide and 2-arachidonoyl glycerol, and of the processes responsible for endocannabinoid biosynthesis, cellular uptake and degradative metabolism [1]. Importantly, there is convincing evidence that there are some disorders in which the endocannabinoid system upregulates in a manner that induces or exacerbates certain disorders, including obesity [1]. The discovery of the link between obesity and the endocannabinoid system prompted the development of the CB1 receptor antagonist/inverse agonist, rimonabant (SR141716A; Acomplia) as an anti-obesity agent. This drug entered European clinics in 2006 for the management of obesity, but was withdrawn in 2008 because of safety concerns about its adverse effects, particularly an increased incidence of depression, anxiety and suicidality [2]. As a result, major pharmaceutical companies appear to have lost interest entirely in all drugs that block CB1 receptors. This has prompted a need for a strategy that would significantly improve the benefit-to-risk ratios of rimonabant-like drugs, just one possibility being to develop a medicine from a CB1 receptor antagonist or antagonist/inverse agonist that does not readily cross the blood-brain barrier [2,3].

There is also convincing evidence, however, that there are a number of serious disorders that are ameliorated by ‘autoprotective’ increases in the release of endocannabinoids onto subpopulations of their receptors and/or in the expression or coupling efficiency of cannabinoid receptors in certain locations. Such increases have, for example, been observed in human cancer and in animal models of neuropathic and inflammatory pain, multiple sclerosis, intestinal disorders, post-traumatic stress disorder, traumatic brain injury, haemorrhagic, septic and cardiogenic shock, hypertension, atherosclerosis and Parkinson's disease [1].

Licensed medicines that exploit beneficial effects of direct cannabinoid receptor activation have already been developed [3,4]. Two of these, the CB1/CB2 receptor agonist, Δ9-tetrahydrocannabinol (Δ9-THC; dronabinol; Marinol) and its synthetic analogue, Nabilone (Cesamet), were approved over 25 years ago as medicines for suppressing nausea and vomiting produced by chemotherapy. Subsequently, the use of dronabinol as an appetite stimulant, for example in AIDS patients experiencing excessive loss of body weight, was also approved. One other medicine that contains Δ9-THC, in this case together with the non-psychoactive plant cannabinoid, cannabidiol, is Sativex. This was licensed in Canada in 2005 for the symptomatic relief of neuropathic pain in multiple sclerosis and as an adjunctive analgesic treatment for adult patients with advanced cancer. In 2010, it was also licensed in the UK and Canada for the treatment of spasticity due to multiple sclerosis and has more recently become an approved medicine in several other countries. Although these medicines do of course all display a favourable benefit-to-risk ratio, they can give rise to unwanted side effects [35].

There is currently a lot of interest in the possibility of developing medicines from compounds that inhibit the cellular uptake and/or metabolism of endocannabinoids when these are being released in an autoprotective manner [1,6]. However, also attracting considerable interest is the idea of exploiting one or other of a wide range of pharmacological strategies expected to maximize the beneficial therapeutic effects and/or minimize the unwanted effects of drugs that activate cannabinoid receptors directly. It is these strategies that form the subject of this review.

2. Direct activation of cannabinoid receptors located outside the blood-brain barrier

It is now generally accepted, first, that many of the unwanted effects of cannabinoid receptor agonists are caused by their activation of CB1 receptors located within the brain and, second, that beneficial effects such as pain relief, amelioration of certain intestinal and cardiovascular disorders, and inhibition of cancer cell proliferation and spread can be induced by selectively activating CB1 and/or CB2 receptors expressed outside the central nervous system [1]. This raises the possibility of developing a peripherally restricted medicine that selectively activates cannabinoid receptors located outside the blood-brain barrier. Attention is focused particularly on the possibility of developing such medicines for pain relief.

One peripherally restricted cannabinoid receptor agonist that possesses antinociceptive activity is naphthalen-1-yl-(4-pentyloxynaphthalen-1-yl)methanone. This is a potent, high-efficacy, orally bioavailable CB1/CB2 receptor agonist that displays significant antihyperalgesic activity in a rat sciatic nerve partial ligation model of neuropathic pain and that appears to act by targeting peripheral CB1 receptors [7]. Thus, its antihyperalgesic effect can be attenuated by a CB1-selective antagonist (SR141716A), but not by a CB2-selective antagonist (SR144528); it can produce this antinociceptive effect without inducing a behavioural effect thought to be mediated by central CB1 receptors (catalepsy), and it does not readily enter the brain. The peripherally restricted potent, orally active CB1/CB2 receptor agonist, ‘compound A’, has also been reported to display anti-hyperalgesic activity at sub-cataleptic doses in a rat spinal nerve ligation model of neuropathic pain and to produce signs of anti-hyperalgesia in the mouse formalin paw model of inflammatory pain [8]. Three other such compounds are AZD1940, AZD1704 and AZ11713908, each of which seems to produce signs of analgesia in rodent models of acute, inflammatory and/or neuropathic pain through the activation of peripheral cannabinoid CB1 receptors when administered orally [9,10]. It is also noteworthy that AZ11713908 generates fewer signs of CNS side effects in a rat Irwin test than the CB1/CB2 receptor agonist, R-(+)-WIN55212. There is evidence too that a synthetic analogue of Δ8-THC, ajulemic acid (CT-3), may ameliorate neuropathic pain mainly by targeting cannabinoid receptors located outside the blood-brain barrier [3]. Five further examples of peripherally restricted cannabinoids that can induce antinociception in animal models are the cannabilactone, AM1710 [11]; the 1-(4-(pyridin-2-yl)benzyl)imidazolidine-2,4-dione derivative, compound 44 [12]; the 5-sulphonyl-benzimidazole derivative, compound 49 [13]; the γ-carboline, compound 29 [14]; and the thiadiazole, compound LBP1 [15]. AM1710 reduces signs of pain elicited by thermal (but not mechanical) stimulation of the rat hind paw at doses that do not produce signs of unwanted CNS side-effects [11]. Similar results were obtained with LBP1 in a rat model of neuropathic pain [15]. AM1710 and compounds 44 and 49 are CB2-selective cannabinoid receptor agonists, whereas compounds 29 and LBP1 are dual CB1/CB2 receptor ligands.

Finally, although orally administered AZD1940 displays antinociceptive activity in rat models of acute and neuropathic pain [9], results obtained in single-dose phase-II studies indicated that it was ineffective against acute pain induced in human subjects by capsaicin or by molar tooth extraction [16].

3. Direct activation of cannabinoid receptors expressed by a particular tissue

There is a strong possibility that the benefit-to-risk ratio of a cannabinoid CB1 or CB1/CB2 receptor agonist could be markedly increased by restricting the distribution of active concentrations of this agonist to a tissue that expresses cannabinoid receptors, which, when activated, would mediate relief from the unwanted effects of one or more particular disorders. Two such tissues may be skin and spinal cord, there being good evidence that these both contain cells that express CB1 and CB2 receptors, the activation of which can produce signs of analgesia or anti-hyperalgesia in animal models of acute, inflammatory or neuropathic pain [3]. There is evidence too that when administered intrathecally, the CB1/CB2 receptor agonist, R-(+)-WIN55212, can induce spinal CB1 and CB2 receptor-mediated signs of relief from bone-tumour-related pain [17], and also antinociception in a rat formalin paw model of inflammatory pain, although not in the rat hot plate model of acute pain [18]. In addition, results obtained from experiments with mice indicate that R-(+)-WIN55212 can act through CB1 and CB2 receptors to reduce signs of hyperalgesia without also inducing catalepsy when it is injected into tumour-bearing hind paws [19], and that the CB2-selective agonist, JWH-015, can induce a CB2-receptor-mediated reduction in bone-cancer-related pain caused by implantation of NCTC2472 fibrosarcoma cells into the femur [20]. It has been found too that intraplantar injection of 2-arachidonoyl glycerol can induce CB2 receptor-mediated relief from hyperalgesia in a murine model of human metastatic bone cancer pain in which fibrosarcoma cells are injected into and around the calcaneus bone of the left hind paw of each animal, and in which CB2 receptor expression increases in non-neuronal cells in the plantar skin of the tumour-bearing paw [21]. Other cannabinoid receptor agonists that have been found to reduce signs of hyperalgesia in this experimental model include AM1241, which is CB2-selective and was antagonized by the CB2-selective antagonist, AM630, but not by the CB1-selective antagonist, AM281, and arachidonylcyclopropylamide, which is CB1-selective and was antagonized by AM281, but not by AM630 [22]. Experiments with mice have also shown that R-(+)-WIN55212 can reduce nociception in the radiant heat tail-flick test when it is applied topically to the tail at a dose that did not impair rotarod performance [23,24]. It could well be, therefore, that by applying a cannabinoid receptor agonist directly to the skin, it would be possible to relieve pain that is restricted to one or more specific regions of the body surface without also provoking major off-target cannabinoid receptor-mediated effects. Further support for this possibility comes from experiments performed with human volunteers, which showed that hyperalgesia induced by capsaicin application to the skin, and the perception of itch induced by cutaneous administration of histamine, could both be decreased by pretreatment with the CB1/CB2 receptor agonist, HU-210, when this was administered by skin patch or dermal microdialysis at a dose that did not produce psychological side effects [25,26]. Also meriting further investigation is the possibility that topical application of a cannabinoid CB1 receptor agonist to one or more areas of the skin might be an effective way of treating (or even preventing) melanoma induced by ultraviolet irradiation [27].

4. Activating upregulated cannabinoid receptors

Some disorders seem to trigger a ‘protective’ upregulation of certain cannabinoid CB1 or CB2 receptors that, when activated, can slow the progression of these disorders or ameliorate their symptoms [1,3]. As discussed in greater detail elsewhere [3], the occurrence of such protective upregulation raises the possibility that for the treatment of at least some disorders, a partial cannabinoid receptor agonist—for example, Δ9-THC or cannabinol—might display a greater benefit-to-risk ratio than a higher efficacy agonist such as CP55940. This is because the extent to which the size the maximal effect of an agonist increases in response to any upregulation of its receptors is inversely related to the efficacy of that agonist.

5. Activating cannabinoid CB2 receptors

Significant attention is currently being directed at the possibility of developing medicines from compounds that can activate CB2 receptors at doses that induce little or no CB1 receptor activation. This has been triggered by the evidence that many of the adverse effects induced by mixed CB1/CB2 receptor agonists result from CB1 rather than from CB2 receptor activation, and that CB2-selective agonists have a number of important potential therapeutic applications. These include the relief of various kinds of pain and the treatment of pruritus, of certain types of cancer, of cough and of some neurodegenerative, immunological, inflammatory, cardiovascular, hepatic, renal and bone disorders (table 1). There is also evidence, first, that CB2 receptor activation can ameliorate neuroinflammation by protecting the blood-brain and blood-spinal cord barriers [67,68]), and second that activation of these receptors can reduce inflammation following spinal cord injury by lowering the expression of toll-like receptors [68]).

Table 1.

Examples of potential therapeutic targets for selective CB2 receptor agonists.

disorder or symptom references
acute or post-operative pain [28,29]a
persistent inflammatory pain [28a,29a,30]
neuropathic pain [12,13,28a,29a,3032]
cancer pain including bone cancer pain [20,22,28a,33a,34a]
pruritus [35]
Parkinson's disease [36,37]a
Huntington's disease [37]a
amyotrophic lateral sclerosis [38,39]
multiple sclerosis [4a,13,40a]
autoimmune uveitis [41]a
HIV-1 brain infection [42]a
alcohol-induced neuroinflammation/neurodegeneration [42]a
anxiety-related disorders [43]
impulsivity: e.g. in bipolar disorder, personality disorders, attention-deficit hyperactivity disorder and substance use disorders [44]
cocaine dependenceb [45]
traumatic brain injury [46]
stroke [47,48a]
atherosclerosis [49,50]
systemic sclerosis [41]a
inflammatory bowel disease [41a,51a,52a,53]
chronic liver diseases; alcoholic liver disease [52a,5457a,58]
diabetic nephropathy [59]
osteoporosis [60]a
cough [61]a
breast, prostate, skin, pancreatic, colorectal, hepatocellular and bone cancer; lymphoma/leukaemia and gliomas [34a,51a,52a,62a,63a,64,65]

aReview article.

bNicotine self-administration and reinstatement of nicotine-seeking behaviour have been found to be unaffected by selective CB2 receptor agonism or antagonism in rats [66].

Importantly, none of the CB2-selective agonists that have been developed to-date are completely CB2-specific. As a result, they are expected to display CB2-selectivity only within a finite dose range and to target CB1 receptors as well when administered at a dose that lies above this range. Indeed, there is evidence from experiments with CB1 wild-type and knockout mice that although some CB2-selective agonists can reduce spasticity in an autoimmune encephalomyelitis model of multiple sclerosis, this depends on their ability to activate CB1 receptors at doses above those at which they activate CB2 receptors [69]. Evidence has also been obtained first, that cannabinoid receptor-dependent alleviation of mechanical allodynia that is induced in mice by brachial plexus avulsion appears to be mainly CB2-mediated in the initial phase but both CB1- and CB2-mediated in the late phase [32], and second, that in a mouse collagen-induced arthritis model, although signs of arthritis are reduced by prolonged CB2 but not prolonged CB1 receptor activation, thermal hyperalgesia is reduced by acute CB1 but not by acute CB2 receptor activation [70]. In addition, it is likely that pharmacological targets other than CB2 or CB1 receptors contribute to sought-after or unwanted effects of CB2-selective agonists, there being evidence, for example, that some but not all such agonists can activate GPR55 and/or modulate activation of this deorphanized receptor by l-α-lysophosphatidylinositol [71]. It is noteworthy too that CB2 receptors seem to increase survival rate in a model of mild sepsis but to reduce survival rate in a model of more severe sepsis, that CB2 receptor activation appears both to exaggerate and to block inflammatory responses in a model of allergic contact dermatitis, and that some inflammatory responses that seem to be aggravated by CB2 receptor agonists are alleviated by CB2 receptor inverse agonists [41].

6. Potential adjunctive strategies for cannabinoid receptor activation

There is good evidence that it may be possible to improve the benefit-to-risk ratio of a cannabinoid receptor agonist such as Δ9-THC, CP55940, R-(+)-WIN55212 or HU-210 for the management of pain by administering it together with a second drug. Thus, for example, additive or synergistic interactions resulting in antinociception have been reported to occur in the rat formalin paw model of inflammatory pain between

  • — intraperitoneal Δ9-THC and morphine [72];

  • — intrathecal R-(+)-WIN55212 and an intrathecally administered α2-adrenoceptor agonist (clonidine), cholinesterase inhibitor (neostigmine) or local anaesthetic (bupivicaine) [73,74];

  • — anandamide and the cyclooxygenase inhibitor, ibuprofen, administered by intraplantar injection [75]; and

  • — HU-210 and the non-steroidal anti-inflammatory drug, acetylsalicylic acid, co-administered systemically [76].

Additive or synergistic interactions resulting in antinociception have also been found to occur between

  • — low-dose R-(+)-WIN55212 and a cyclooxygenase-2 inhibitor, NS-398, co-administered intracisternally, for the attenuation of nociceptive scratching behaviour induced in rats by formalin injection into the temporomandibular joint of the jaw [77];

  • — R-(+)-WIN55212 and the non-steroidal anti-inflammatory drug, ketorolac, co-administered systemically, for the attenuation of nociception in a mouse model of inflammatory visceral pain, although not in the mouse tail flick model of acute pain [78];

  • — HU-210 and the non-steroidal anti-inflammatory drug, acetylsalicylic acid, co-administered systemically, in the rat hot plate model of acute pain [76];

  • — Δ9-THC and an opioid such as morphine, codeine or fentanyl in mouse, rat, guinea pig and monkey models of acute or arthritic pain [7989];

  • — CP55940 and the α2-adrenoceptor agonist, dexmedetomidine, in the mouse hot plate and tail flick models of acute pain [83];

  • — CP55940 and the N-methyl-d-aspartate (NMDA) receptor antagonist, (–)-6-phosphonomethyl-deca-hydroisoquinoline-3-carboxylic acid (LY235959), in the mouse hot plate test [90]; and

  • — R-(+)-WIN55212, given by intracerebroventricular or intraplantar injection, and a selective agonist for the neuropeptide FF1 or FF2 receptor, injected intracerebroventricularly, in mouse models of acute pain [91].

Importantly, evidence has been obtained through the construction of isobolograms that of the above interactions, those between R-(+)-WIN55212 and clonidine, neostigmine or bupivicaine [73,74] as well as those between anandamide and ibuprofen [75], Δ9-THC and an opioid [81,83,86], CP55940 and dexmedetomidine [83] and CP55940 and LY235959 [90], are all synergistic rather than just additive in nature. A synergistic antinociceptive interaction has also been reported to occur between the CB1-selective agonist, arachidonylcyclopropylamide, and the CB2-selective agonist, AM1241, in a mouse model of cancer pain following intraplantar coadministration of these two compounds [22]. This is of interest since it raises the possibility that, for at least some kinds of pain, a mixed CB1/CB2 agonist may be more effective as an analgesic medicine than a CB1- or CB2-selective agonist.

Results from clinical studies with patients experiencing chronic non-cancer pain have also provided evidence that cannabinoid receptor agonists can enhance opioid-induced analgesia [92,93] and that inhaled vapourized cannabis can augment the analgesic effect of the opioids, morphine and oxycodone in patients experiencing various kinds of chronic pain without inducing any unacceptable adverse events [94]. In contrast, no synergistic or additive antinociceptive interaction has been detected between Δ9-THC and the µ-opioid receptor agonist, piritramide, in patients suffering from acute post-operative pain [95] or between Δ9-THC and morphine in human volunteers subjected to noxious electrical or thermal stimulation of the skin or to painful digital pressure [96,97]. However, together (but not separately), these drugs did reduce the affective response to cutaneous thermal stimulation [97].

Evidence that certain potentially beneficial effects of a cannabinoid receptor agonist other than pain relief can be enhanced by administering it together with one or other of a set of non-cannabinoid receptor ligands has also emerged from in vivo animal experiments (table 2). It should be noted that the anticonvulsant interactions between R-(+)-WIN55212 and ethosuximide or valproate that are referred to in table 2 were probably at least partly pharmacokinetic in nature [104], whereas those between R-(+)-WIN55212 or arachidonyl-2′-chloroethylamidamide and phenobarbital were most likely pharmacodynamic in nature [104,106]. It is noteworthy too that additive or synergistic interactions have also been observed to occur in vitro between cannabinoid receptor agonists and anti-cancer drugs for the production of apoptosis or anti-proliferative effects in certain cancer cell lines [110,112].

Table 2.

Examples of additive or synergistic interactions observed in vivo between cannabinoid receptor agonists and non-cannabinoid receptor ligands in animal models of certain disorders.a ACEA, arachidonyl-2′-chloroethylamide; 8-OH-DPAT, 8-hydroxy-2-(di-n-propylamino) tetralin hydrobromide; i.p., intraperitoneal; i.v., intravenous; s.c., subcutaneous.

disorder and measured effect cannabinoid receptor agonist co-administered compound reference
anxiety or depression
anxiolytic effects in mouse elevated plus-mazec and mouse hole-board test R-(+)-WIN55212 (i.p.) diazepam (i.p.) [98]
anxiolytic effects in mouse light-dark box, open-field test and elevated plus-maze low-dose Δ9-THC (i.p.) low-dose nicotine (s.c.) [99,100]
anxiolytic effect in rat elevated plus-maze low-dose Δ9-THC (i.p.) low dose of the 5-HT1A receptor-selective agonist, 8-OH-DPAT (i.p.) [101]
antidepressant effect in rat forced swim test low-dose CP55940 (i.p.) low-dose imipramine (i.p.) [102]
epilepsy
anticonvulsant effect on mouse pentylenetetrazole-induced clonic or tonic-clonic seizures low-dose of the CB1-selective agonist, ACEA (i.p.) low-dose naltrexone (i.p.) [103]
anticonvulsant effect on mouse pentylenetetrazole-induced clonic seizures low-dose R-(+)-WIN55212 (i.p.) ethosuximide, phenobarbital or valproate (i.p.) [104]
anticonvulsant effect on mouse maximal electroshock-induced seizures low-dose R-(+)-WIN55212 (i.p.) carbamazepine, phenytoin, phenobarbital or valproate (i.p.) [105]
anticonvulsant effect on mouse maximal electroshock-induced seizures low-dose of the CB1-selective agonist, ACEA (i.p.) phenobarbital (i.p.) [106]
anticonvulsant effect on mouse electroshock-induced seizures low-dose R-(+)-WIN55212 (i.p.) diazepam (i.p.) [107]
haemorrhagic shock or glaucoma
increased survival time in a rat model of haemorrhagic shock Δ8-THC (i.v.) cyclooxygenase-2 inhibitor, NS-398 (i.v.) [108]
reduction of rat intraocular pressure low-dose R-(+)-WIN55212 (i.p.) low-dose abnormal-cannabidiol or cannabigerol-dimethyl heptyl (topical) [109]
cancer or chemotherapy-induced vomiting
reduction of glioma xenograft growth in nude mice low-dose Δ9-THC (peritumorally) low-dose temozolomide (peritumorally)b [110]
inhibition of vomiting and retching induced by cisplatin in house musk shrews low-dose Δ9-THC (i.p.) low dose of the 5-HT3 receptor antagonist, ondansetron (i.p.) [111]

aSee introduction to this section for antinociceptive interactions.

bLow-dose temozolomide also exerted a strong anti-tumoral effect in combination with a low-dose mixture of Δ9-THC and the non-psychoactive phytocannabinoid, cannabidiol.

cIsobolographic analysis indicated this interaction to be synergistic.

One other adjunctive strategy for a cannabinoid receptor agonist may be to administer it together with a CB1 receptor antagonist/inverse agonist. Thus, for example, it has been found that

  • — an ultra-low dose of SR141716A can prolong R-(+)-WIN55212-induced antinociception in a rat model of acute pain [113];

  • — an ultra-low dose of AM251 can enhance the ability of the CB1-selective agonist, arachidonyl-2′-chloroethylamide, to protect mice from pentylenetetrazole-induced seizures [114]; and

  • — administration of a selective CB1 receptor antagonist/inverse agonist together with a CB2-selective agonist may be particularly effective for the treatment of hepatic ischaemia/reperfusion injury caused by liver transplantation [115] and of disorders, such as Parkinson's disease [116], systemic sclerosis [117], chronic liver diseases, including alcohol-induced liver injury [56] and stroke [118], and perhaps also for the management of cocaine dependence [45,119].

It is possible that the last of these three potential adjunctive strategies could be exploited using Δ9-tetrahydrocannabivarin, because this plant cannabinoid can both block CB1 receptors and activate CB2 receptors [120,121]. Indeed, there is already evidence from experiments using animal models of Parkinson's disease and hepatic ischaemia/reperfusion injury, that Δ9-tetrahydrocannabivarin would display efficacy as a medicine against both of these disorders [115,116].

Ideally, a multi-targeting strategy should of course be one that enhances sought-after effects to a greater extent than unwanted effects. It is noteworthy, therefore, that there is already evidence from experiments performed with mice or rats that the risk of developing dependence to opioids [122,123] and nicotine [99] increases when such a compound is co-administered with a cannabinoid CB1/CB2 receptor agonist. There is evidence too that Δ9-THC can undergo additive or synergistic interactions with a range of non-cannabinoids to disrupt motor function and thermoregulation, as indicated by the production of catalepsy, hypokinesia or hypothermia in mice or rats. These non-cannabinoids include opioids, nicotine, benzodiazepines, prostaglandins, reserpine and ligands that activate or block muscarinic cholinoceptors or some types of dopamine, noradrenaline, 5-hydroxytryptamine or γ-aminobutyric acid receptors [72,99,124,125]. There is also evidence that R-(+)-WIN55212 enhances not only the anticonvulsant effects of carbamazepine, phenytoin, phenobarbital, valproate and ethosuximide in mice (table 2), but also the impairment of skeletal muscle strength by all these compounds, the impairment of motor co-ordination by phenobarbital, valproate and ethosuximide, and the impairment of long-term memory by phenytoin, phenobarbital, valproate and ethosuximide [104,105]. In contrast, however, the CB1-selective agonist, arachidonyl-2′-chloroethylamide, enhanced the anticonvulsant effect of phenobarbital in mice (table 2) without augmenting impairment by this barbiturate of skeletal muscle strength, motor co-ordination or long-term memory [106]. It is also noteworthy that administration of a cannabinoid receptor agonist, together with morphine, seems to oppose the development of tolerance to the antinociceptive effects of these compounds. Thus, for example, chronic systemic administration of a low-dose combination of Δ9-THC and morphine to rats has been reported to induce antinociception without also producing tolerance in a rat paw pressure model of acute pain in which tolerance did develop when morphine or Δ9-THC was administered chronically by itself at a higher dose [87]. Furthermore, it has been found first, that chronic systemic co-administration of CP55940 with morphine can attenuate the tolerance that develops to the antinociceptive effect of morphine in the mouse hot plate test when it is administered repeatedly by itself [90], and second, that an ultra-low dose of SR141716A that prolongs R-(+)-WIN55212-induced antinociception in the rat tail flick test also opposes the development of tolerance to this CB1/CB2 receptor agonist [113]. There is evidence too that the CB2-selective agonist, AM1241, can prevent the neuroinflammatory consequences of sustained morphine treatment [126].

7. Mixing strategies

There may be therapeutic benefits to be gained from combining some of the strategies that have been mentioned in this review. One possibility for pain relief would be to administer a CB2-selective agonist intrathecally instead of orally. Thus, there have been reports that JWH-015 can reduce signs of post-operative pain in rats [127], and that signs of neuropathic pain can be reduced by JWH-133 in mice [128], and by AM1710 in rats [129] when these three CB2-selective agonists are injected intrathecally. There is evidence too that signs of analgesia induced in models of acute pain by transdermal administration of an opioid can be enhanced by transdermal or intrathecal co-administration of a low dose of a CB1/CB2 receptor agonist [24,84]. It is also noteworthy that antinociceptive synergy has been detected in the mouse tail flick test between low-doses of R-(+)-WIN55212 co-administered topically and intrathecally [23].

8. Conclusions and future directions

This review has focused on preclinical findings described in papers published up to April 2012 that together provide an indication of the likely strengths and weaknesses of a number of potential strategies for improving the therapeutic efficacy and/or minimizing the adverse effects of cannabinoid receptor agonists in the clinic.

The available published information about each of these strategies suggests that, for many of them, their strengths significantly outweigh any of their identified weaknesses. This information has, however, come almost entirely from preclinical research. Consequently there is now an urgent need, first, unless they have already been performed, for phase I clinical trials with healthy human subjects that test the safety of each drug that is selected to implement one or other of these potential strategies and, second, for phase II trials with patients. When planning such clinical trials, it will be important to construct a short-list of disorders that are in need of better medicines, and whose signs, symptoms and/or progression are most likely to be managed effectively in the clinic by one or other of the strategies described in this review. For each of these disorders, it will also be important to select the strategy that would be the one most likely to produce the greatest benefit-to-risk ratio in patients.

References

  • 1.Pertwee R. G. 2005. The therapeutic potential of drugs that target cannabinoid receptors or modulate the tissue levels or actions of endocannabinoids. AAPS J. 7, E625–E654 10.1208/aapsj070364 (doi:10.1208/aapsj070364) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Le Foll B., Gorelick D. A., Goldberg S. R. 2009. The future of endocannabinoid-oriented clinical research after CB1 antagonists. Psychopharmacology 205, 171–174 10.1007/s00213-009-1506-7 (doi:10.1007/s00213-009-1506-7) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Pertwee R. G. 2009. Emerging strategies for exploiting cannabinoid receptor agonists as medicines. Br. J. Pharmacol. 156, 397–411 10.1111/j.1476-5381.2008.00048.x (doi:10.1111/j.1476-5381.2008.00048.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Pertwee R. G., Thomas A. 2009. Therapeutic applications for agents that act at CB1 and CB2 receptors. In The cannabinoid receptors (ed. Reggio P. H.), pp. 361–392, 1st edn. New York, NY: Humana Press [Google Scholar]
  • 5.Pertwee R. G. 2007. Cannabinoids and multiple sclerosis. Mol. Neurobiol. 36, 45–59 10.1007/s12035-007-0005-2 (doi:10.1007/s12035-007-0005-2) [DOI] [PubMed] [Google Scholar]
  • 6.Petrosino S., Di Marzo V. 2010. FAAH and MAGL inhibitors: therapeutic opportunities from regulating endocannabinoid levels. Curr. Opin. Investig. Drugs 11, 51–62 [PubMed] [Google Scholar]
  • 7.Dziadulewicz E. K., et al. 2007. Naphthalen-1-yl-(4-pentyloxynaphthalen-1-yl)methanone: a potent, orally bioavailable human CB1/CB2 dual agonist with antihyperalgesic properties and restricted central nervous system penetration. J. Med. Chem. 50, 3851–3856 10.1021/jm070317a (doi:10.1021/jm070317a) [DOI] [PubMed] [Google Scholar]
  • 8.Boyce S. 2008. Targeting peripheral cannabinoid 1 (CB1) receptors for chronic pain. Fundam. Clin. Pharmacol. 22(Suppl. 2), 5–28 10.1111/j.1472-8206.2008.00591.x (doi:10.1111/j.1472-8206.2008.00591.x) [DOI] [Google Scholar]
  • 9.Groblewski T., et al. 2010. Pre-clinical pharmacological properties of novel peripherally-acting CB1-CB2 agonists In 20th Annu. Symp. on the Cannabinoids; 2010, p. 37 Research Triangle Park, NC: Int. Cannabinoid Research Society [Google Scholar]
  • 10.Yu X. H., Cao C. Q., Martino G., Puma C., Morinville A., St-Onge S., Lessard É., Perkins M. N., Laird J. M. A. 2010. A peripherally restricted cannabinoid receptor agonist produces robust anti-nociceptive effects in rodent models of inflammatory and neuropathic pain. Pain 151, 337–344 10.1016/j.pain.2010.07.019 (doi:10.1016/j.pain.2010.07.019) [DOI] [PubMed] [Google Scholar]
  • 11.Rahn E. J., Thakur G. A., Wood J. A. T., Zvonok A. M., Makriyannis A., Hohmann A. G. 2011. Pharmacological characterization of AM1710, a putative cannabinoid CB2 agonist from the cannabilactone class: antinociception without central nervous system side-effects. Pharmacol. Biochem. Behav. 98, 493–502 10.1016/j.pbb.2011.02.024 (doi:10.1016/j.pbb.2011.02.024) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.van der Stelt M., et al. 2011. Discovery and optimization of 1-(4-(pyridin-2-yl)benzyl)imidazolidine-2,4-dione derivatives as a novel class of selective cannabinoid CB2 receptor agonists. J. Med. Chem. 54, 7350–7362 10.1021/jm200916p (doi:10.1021/jm200916p) [DOI] [PubMed] [Google Scholar]
  • 13.Gijsen H. J. M., De Cleyn M. A. J., Surkyn M., Van Lommen G. R. E., Verbist B. M. P., Nijsen M. J. M. A., Meert T., Van Wauwe J., Aerssens J. 2012. 5-Sulfonyl-benzimidazoles as selective CB2 agonists: part 2. Bioorg. Med. Chem. Lett. 22, 547–552 10.1016/j.bmcl.2011.10.091 (doi:10.1016/j.bmcl.2011.10.091) [DOI] [PubMed] [Google Scholar]
  • 14.Cheng Y.-X., et al. 2012. γ-Carbolines: a novel class of cannabinoid agonists with high aqueous solubility and restricted CNS penetration. Bioorg. Med. Chem. Lett. 22, 1619–1624 10.1016/j.bmcl.2011.12.124 (doi:10.1016/j.bmcl.2011.12.124) [DOI] [PubMed] [Google Scholar]
  • 15.Adam J. M., et al. 2012. Low brain penetrant CB1 receptor agonists for the treatment of neuropathic pain. Bioorg. Med. Chem. Lett. 22, 2932–2937 10.1016/j.bmcl.2012.02.048 (doi:10.1016/j.bmcl.2012.02.048) [DOI] [PubMed] [Google Scholar]
  • 16.Groblewski T., et al. 2010. Peripherally-acting CB1-CB2 agonists for pain: do they still hold promise? In 20th Annu. Symp. on the Cannabinoids; 2010, p. 38 Research Triangle Park, NC: Int. Cannabinoid Research Society [Google Scholar]
  • 17.Cui J. H., Kim W. M., Lee H. G., Kim Y. O., Kim C. M., Yoon M. H. 2011. Antinociceptive effect of intrathecal cannabinoid receptor agonist WIN 55,212-2 in a rat bone tumor pain model. Neurosci. Lett. 493, 67–71 10.1016/j.neulet.2010.12.052 (doi:10.1016/j.neulet.2010.12.052) [DOI] [PubMed] [Google Scholar]
  • 18.Hama A., Sagen J. 2011. Centrally mediated antinociceptive effects of cannabinoid receptor ligands in rat models of nociception. Pharmacol. Biochem. Behav. 100, 340–346 10.1016/j.pbb.2011.09.004 (doi:10.1016/j.pbb.2011.09.004) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Potenzieri C., Harding-Rose C., Simone D. A. 2008. The cannabinoid receptor agonist, WIN 55, 212-2, attenuates tumor-evoked hyperalgesia through peripheral mechanisms. Brain. Res. 1215, 69–75 10.1016/j.brainres.2008.03.063 (doi:10.1016/j.brainres.2008.03.063) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Gu X. P., Mei F. M., Liu Y., Zhang R., Zhang J., Ma Z. L. 2011. Intrathecal administration of the cannabinoid 2 receptor agonist JWH015 can attenuate cancer pain and decrease mRNA expression of the 2B subunit of N-methyl-d-aspartic acid. Anesth. Analg. 113, 405–411 10.1213/ANE.0b013e31821d1062 (doi:10.1213/ANE.0b013e31821d1062) [DOI] [PubMed] [Google Scholar]
  • 21.Khasabova I. A., Chandiramani A., Harding-Rose C., Simone D. A., Seybold V. S. 2011. Increasing 2-arachidonoyl glycerol signaling in the periphery attenuates mechanical hyperalgesia in a model of bone cancer pain. Pharmacol. Res. 64, 60–67 10.1016/j.phrs.2011.03.007 (doi:10.1016/j.phrs.2011.03.007) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Khasabova I. A., Gielissen J., Chandiramani A., Harding-Rose C., Abu Odeh D., Simone D. A., Seybold V. S. 2011. CB1 and CB2 receptor agonists promote analgesia through synergy in a murine model of tumor pain. Behav. Pharmacol. 22, 607–616 10.1097/FBP.0b013e3283474a6d (doi:10.1097/FBP.0b013e3283474a6d) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Dogrul A., Gul H., Akar A., Yildiz O., Bilgin F., Guzeldemir E. 2003. Topical cannabinoid antinociception: synergy with spinal sites. Pain 105, 11–16 10.1016/S0304-3959(03)00068-X (doi:10.1016/S0304-3959(03)00068-X) [DOI] [PubMed] [Google Scholar]
  • 24.Yesilyurt O., Dogrul A., Gul H., Seyrek M., Kusmez O., Ozkan Y., Yildiz O. 2003. Topical cannabinoid enhances topical morphine antinociception. Pain 105, 303–308 10.1016/S0304-3959(03)00245-8 (doi:10.1016/S0304-3959(03)00245-8) [DOI] [PubMed] [Google Scholar]
  • 25.Rukwied R., Watkinson A., McGlone F., Dvorak M. 2003. Cannabinoid agonists attenuate capsaicin-induced responses in human skin. Pain 102, 283–288 10.1016/S0304-3959(02)00401-3 (doi:10.1016/S0304-3959(02)00401-3) [DOI] [PubMed] [Google Scholar]
  • 26.Dvorak M., Watkinson A., McGlone F., Rukwied R. 2003. Histamine induced responses are attenuated by a cannabinoid receptor agonist in human skin. Inflamm. Res. 52, 238–245 10.1007/s00011-003-1162-z (doi:10.1007/s00011-003-1162-z) [DOI] [PubMed] [Google Scholar]
  • 27.Magina S., Esteves-Pinto C., Moura E., Serrão M. P., Moura D., Petrosino S., Di Marzo V., Vieira-Coelho M. A. 2011. Inhibition of basal and ultraviolet B-induced melanogenesis by cannabinoid CB1 receptors: a keratinocyte-dependent effect. Arch. Dermatol. Res. 303, 201–210 10.1007/s00403-011-1126-z (doi:10.1007/s00403-011-1126-z) [DOI] [PubMed] [Google Scholar]
  • 28.Whiteside G. T., Lee G. P., Valenzano K. J. 2007. The role of the cannabinoid CB2 receptor in pain transmission and therapeutic potential of small molecule CB2 receptor agonists. Curr. Med. Chem. 14, 917–936 10.2174/092986707780363023 (doi:10.2174/092986707780363023) [DOI] [PubMed] [Google Scholar]
  • 29.Guindon J., Hohmann A. G. 2008. Cannabinoid CB2 receptors: a therapeutic target for the treatment of inflammatory and neuropathic pain. Br. J. Pharmacol. 153, 319–334 10.1038/sj.bjp.0707531 (doi:10.1038/sj.bjp.0707531) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Beltramo M., Bernardini N., Bertorelli R., Campanella M., Nicolussi E., Fredduzzi S., Reggiani A. 2006. CB2 receptor-mediated antihyperalgesia: possible direct involvement of neural mechanisms. Eur. J. Neurosci. 23, 1530–1538 10.1111/j.1460-9568.2006.04684.x (doi:10.1111/j.1460-9568.2006.04684.x) [DOI] [PubMed] [Google Scholar]
  • 31.Gutierrez T., Crystal J. D., Zvonok A. M., Makriyannis A., Hohmann A. G. 2011. Self-medication of a cannabinoid CB2 agonist in an animal model of neuropathic pain. Pain 152, 1976–1987 10.1016/j.pain.2011.03.038 (doi:10.1016/j.pain.2011.03.038) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Paszcuk A. F., Dutra R. C., da Silva K. A. B. S., Quintão N. L. M., Campos M. M., Calixto J. B. 2011. Cannabinoid agonists inhibit neuropathic pain induced by brachial plexus avulsion in mice by affecting glial cells and MAP kinases. PLoS ONE 6, e24034. 10.1371/journal.pone.0024034 (doi:10.1371/journal.pone.0024034) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Bab I., Smoum R., Bradshaw H., Mechoulam R. 2011. Skeletal lipidomics: regulation of bone metabolism by fatty acid amide family. Br. J. Pharmacol. 163, 1441–1446 10.1111/j.1476-5381.2011.01474.x (doi:10.1111/j.1476-5381.2011.01474.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Guindon J., Hohmann A. G. 2011. The endocannabinoid system and cancer: therapeutic implication. Br. J. Pharmacol. 163, 1447–1463 10.1111/j.1476-5381.2011.01327.x (doi:10.1111/j.1476-5381.2011.01327.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Odan M., et al. 2012. Discovery of S-777469: an orally available CB2 agonist as an antipruritic agent. Bioorg. Med. Chem. Lett. 22, 2803–2806 10.1016/j.bmcl.2012.02.072 (doi:10.1016/j.bmcl.2012.02.072) [DOI] [PubMed] [Google Scholar]
  • 36.Little J. P., Villanueva E. B., Klegeris A. 2011. Therapeutic potential of cannabinoids in the treatment of neuroinflammation associated with Parkinson's disease. Mini-Rev. Med. Chem. 11, 582–590 [DOI] [PubMed] [Google Scholar]
  • 37.Fernández-Ruiz J., et al. 2011. Prospects for cannabinoid therapies in basal ganglia disorders. Br. J. Pharmacol. 163, 1365–1378 10.1111/j.1476-5381.2011.01365.x (doi:10.1111/j.1476-5381.2011.01365.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kim K., Moore D. H., Makriyannis A., Abood M. E. 2006. AM1241, a cannabinoid CB2 receptor selective compound, delays disease progression in a mouse model of amyotrophic lateral sclerosis. Eur. J. Pharmacol. 542, 100–105 10.1016/j.ejphar.2006.05.025 (doi:10.1016/j.ejphar.2006.05.025) [DOI] [PubMed] [Google Scholar]
  • 39.Shoemaker J. L., Seely K. A., Reed R. L., Crow J. P., Prather P. L. 2007. The CB2 cannabinoid agonist AM-1241 prolongs survival in a transgenic mouse model of amyotrophic lateral sclerosis when initiated at symptom onset. J. Neurochem. 101, 87–98 10.1111/j.1471-4159.2006.04346.x (doi:10.1111/j.1471-4159.2006.04346.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dittel B. N. 2008. Direct suppression of autoreactive lymphocytes in the central nervous system via the CB2 receptor. Br. J. Pharmacol. 153, 271–276 10.1038/sj.bjp.0707493 (doi:10.1038/sj.bjp.0707493) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Basu S., Dittel B. N. 2011. Unraveling the complexities of cannabinoid receptor 2 (CB2) immune regulation in health and disease. Immunol. Res. 51, 26–38 10.1007/s12026-011-8210-5 (doi:10.1007/s12026-011-8210-5) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Persidsky Y., Ho W. Z., Ramirez S. H., Potula R., Abood M. E., Unterwald E., Tuma R. 2011. HIV-1 infection and alcohol abuse: neurocognitive impairment, mechanisms of neurodegeneration and therapeutic interventions. Brain Behav. Immun. 25, S61–S70 10.1016/j.bbi.2011.03.001 (doi:10.1016/j.bbi.2011.03.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Busquets-Garcia A., Puighermanal E., Pastor A., de la Torre R., Maldonado R., Ozaita A. 2011. Differential role of anandamide and 2-arachidonoylglycerol in memory and anxiety-like responses. Biol. Psychiatry 70, 479–486 10.1016/j.biopsych.2011.04.022 (doi:10.1016/j.biopsych.2011.04.022) [DOI] [PubMed] [Google Scholar]
  • 44.Navarrete F., Pérez-Ortiz J. M., Manzanares J. 2012. Cannabinoid CB2 receptor-mediated regulation of impulsive-like behaviour in DBA/2 mice. Br. J. Pharmacol. 165, 260–273 10.1111/j.1476-5381.2011.01542.x (doi:10.1111/j.1476-5381.2011.01542.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Xi Z.-X., et al. 2011. Brain cannabinoid CB2 receptors modulate cocaine's actions in mice. Nat. Neurosci. 14, 1160–1166 10.1038/nn.2874 (doi:10.1038/nn.2874) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Elliott M. B., Tuma R. F., Amenta P. S., Barbe M. F., Jallo J. I. 2011. Acute effects of a selective cannabinoid-2 receptor agonist on neuroinflammation in a model of traumatic brain injury. J. Neurotrauma 28, 973–981 10.1089/neu.2010.1672 (doi:10.1089/neu.2010.1672) [DOI] [PubMed] [Google Scholar]
  • 47.Zhang M., Martin B. R., Adler M. W., Razdan R. K., Jallo J. I., Tuma R. F. 2007. Cannabinoid CB2 receptor activation decreases cerebral infarction in a mouse focal ischemia/reperfusion model. J. Cereb. Blood Flow Metab. 27, 1387–1396 10.1038/sj.jcbfm.9600447 (doi:10.1038/sj.jcbfm.9600447) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Pacher P., Haskó G. 2008. Endocannabinoids and cannabinoid receptors in ischaemia-reperfusion injury and preconditioning. Br. J. Pharmacol. 153, 252–262 10.1038/sj.bjp.0707582 (doi:10.1038/sj.bjp.0707582) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Steffens S., Veillard N. R., Arnaud C., Pelli G., Burger F., Staub C., Zimmer A., Frossard J.-L., Mach F. 2005. Low dose oral cannabinoid therapy reduces progression of atherosclerosis in mice. Nature 434, 782–786 10.1038/nature03389 (doi:10.1038/nature03389) [DOI] [PubMed] [Google Scholar]
  • 50.Hoyer F. F., Steinmetz M., Zimmer S., Becker A., Lütjohann D., Buchalla R., Zimmer A., Nickenig G. 2011. Atheroprotection via cannabinoid receptor-2 is mediated by circulating and vascular cells in vivo. J. Mol. Cell. Cardiol. 51, 1007–1014 10.1016/j.yjmcc.2011.08.008 (doi:10.1016/j.yjmcc.2011.08.008) [DOI] [PubMed] [Google Scholar]
  • 51.Wright K. L., Duncan M., Sharkey K. A. 2008. Cannabinoid CB2 receptors in the gastrointestinal tract: a regulatory system in states of inflammation. Br. J. Pharmacol. 153, 263–270 10.1038/sj.bjp.0707486 (doi:10.1038/sj.bjp.0707486) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Izzo A. A., Camilleri M. 2008. Emerging role of cannabinoids in gastrointestinal and liver diseases: basic and clinical aspects. Gut 57, 1140–1155 10.1136/gut.2008.148791 (doi:10.1136/gut.2008.148791) [DOI] [PubMed] [Google Scholar]
  • 53.Singh U. P., Singh N. P., Singh B., Price R. L., Nagarkatti M., Nagarkatti P. S. 2012. Cannabinoid receptor-2 (CB2) agonist ameliorates colitis in IL-10−/− mice by attenuating the activation of T cells and promoting their apoptosis. Toxicol. Appl. Pharmacol. 258, 256–267 10.1016/j.taap.2011.11.005 (doi:10.1016/j.taap.2011.11.005) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mallat A., Teixeira-Clerc F., Deveaux V., Lotersztajn S. 2007. Cannabinoid receptors as new targets of antifibrosing strategies during chronic liver diseases. Expert Opin. Ther. Targets 11, 403–409 10.1517/14728222.11.3.403 (doi:10.1517/14728222.11.3.403) [DOI] [PubMed] [Google Scholar]
  • 55.Lotersztajn S., et al. 2008. CB2 receptors as new therapeutic targets for liver diseases. Br. J. Pharmacol. 153, 286–289 10.1038/sj.bjp.0707511 (doi:10.1038/sj.bjp.0707511) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mallat A., Teixeira-Clerc F., Deveaux V., Manin S., Lotersztajn S. 2011. The endocannabinoid system as a key mediator during liver diseases: new insights and therapeutic openings. Br. J. Pharmacol. 163, 1432–1440 10.1111/j.1476-5381.2011.01397.x (doi:10.1111/j.1476-5381.2011.01397.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Huang L., Quinn M. A., Frampton G. A., Golden L. E., DeMorrow S. 2011. Recent advances in the understanding of the role of the endocannabinoid system in liver diseases. Dig. Liver Dis. 43, 188–193 10.1016/j.dld.2010.08.010 (doi:10.1016/j.dld.2010.08.010) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Louvet A., Teixeira-Clerc F., Chobert M. N., Deveaux V., Pavoine C., Zimmer A., Pecker F., Mallat A., Lotersztajn S. 2011. Cannabinoid CB2 receptors protect against alcoholic liver disease by regulating Kupffer cell polarization in mice. Hepatology 54, 1217–1226 10.1002/hep.24524 (doi:10.1002/hep.24524) [DOI] [PubMed] [Google Scholar]
  • 59.Barutta F., et al. 2011. Protective role of cannabinoid receptor type 2 in a mouse model of diabetic nephropathy. Diabetes 60, 2386–2396 10.2337/db10-1809 (doi:10.2337/db10-1809) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bab I., Zimmer A., Melamed E. 2009. Cannabinoids and the skeleton: from marijuana to reversal of bone loss. Ann. Med. 41, 560–567 10.1080/07853890903121025 (doi:10.1080/07853890903121025) [DOI] [PubMed] [Google Scholar]
  • 61.Maher S. A., Dubuis E. D., Belvisi M. G. 2011. G-protein coupled receptors regulating cough. Curr. Opin. Pharmacol. 11, 248–253 10.1016/j.coph.2011.06.005 (doi:10.1016/j.coph.2011.06.005) [DOI] [PubMed] [Google Scholar]
  • 62.Guzmán M. 2003. Cannabinoids: potential anticancer agents. Nat. Rev. Cancer 3, 745–755 10.1038/nrc1188 (doi:10.1038/nrc1188) [DOI] [PubMed] [Google Scholar]
  • 63.Malfitano A. M., Ciaglia E., Gangemi G., Gazzerro P., Laezza C., Bifulco M. 2011. Update on the endocannabinoid system as an anticancer target. Expert Opin. Ther. Targets 15, 297–308 10.1517/14728222.2011.553606 (doi:10.1517/14728222.2011.553606) [DOI] [PubMed] [Google Scholar]
  • 64.Vara D., Salazar M., Olea-Herrero N., Guzmán M., Velasco G., Díaz-Laviada I. 2011. Anti-tumoral action of cannabinoids on hepatocellular carcinoma: role of AMPK-dependent activation of autophagy. Cell Death Differ. 18, 1099–1111 10.1038/cdd.2011.32 (doi:10.1038/cdd.2011.32) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Nasser M. W., Qamri Z., Deol Y. S., Smith D., Shilo K., Zou X., Ganju R. K. 2011. Crosstalk between chemokine receptor CXCR4 and cannabinoid receptor CB2 in modulating breast cancer growth and invasion. PLoS ONE 6, e23901. 10.1371/journal.pone.0023901 (doi:10.1371/journal.pone.0023901) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gamaleddin I., Zvonok A., Makriyannis A., Goldberg S. R., Le Foll B. 2012. Effects of a selective cannabinoid CB2 agonist and antagonist on intravenous nicotine self administration and reinstatement of nicotine seeking. PLoS ONE 7, e29900. 10.1371/journal.pone.0029900 (doi:10.1371/journal.pone.0029900) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ramirez S. H., et al. 2012. Activation of cannabinoid receptor 2 attenuates leukocyte-endothelial cell interactions and blood-brain barrier dysfunction under inflammatory conditions. J. Neurosci. 32, 4004–4016 10.1523/JNEUROSCI.4628-11.2012 (doi:10.1523/JNEUROSCI.4628-11.2012) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Adhikary S., Li H., Heller J., Skarica M., Zhang M., Ganea D., Tuma R. F. 2011. Modulation of inflammatory responses by a cannabinoid-2-selective agonist after spinal cord injury. J. Neurotrauma 28, 2417–2427 10.1089/neu.2011.1853 (doi:10.1089/neu.2011.1853) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Pryce G., Baker D. 2007. Control of spasticity in a multiple sclerosis model is mediated by CB1, not CB2, cannabinoid receptors. Br. J. Pharmacol. 150, 519–525 10.1038/sj.bjp.0707003 (doi:10.1038/sj.bjp.0707003) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Kinsey S. G., Naidu P. S., Cravatt B. F., Dudley D. T., Lichtman A. H. 2011. Fatty acid amide hydrolase blockade attenuates the development of collagen-induced arthritis and related thermal hyperalgesia in mice. Pharmacol. Biochem. Behav. 99, 718–725 10.1016/j.pbb.2011.06.022 (doi:10.1016/j.pbb.2011.06.022) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Anavi-Goffer S., Baillie G., Irving A. J., Gertsch J., Greig I. R., Pertwee R. G., Ross R. A. 2012. Modulation of L-α-lysophosphatidylinositol/GPR55 mitogen-activated protein kinase (MAPK) signaling by cannabinoids. J. Biol. Chem. 287, 91–104 10.1074/jbc.M111.296020 (doi:10.1074/jbc.M111.296020) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Finn D. P., Beckett S. R. G., Roe C. H., Madjd A., Fone K. C. F., Kendall D. A., Marsden C. A., Chapman V. 2004. Effects of coadministration of cannabinoids and morphine on nociceptive behaviour, brain monoamines and HPA axis activity in a rat model of persistent pain. Eur. J. Neurosci. 19, 678–686 10.1111/j.0953-816X.2004.03177.x (doi:10.1111/j.0953-816X.2004.03177.x) [DOI] [PubMed] [Google Scholar]
  • 73.Yoon M. H., Choi J. 2003. Pharmacologic interaction between cannabinoid and either clonidine or neostigmine in the rat formalin test. Anesthesiology 99, 701–707 10.1097/00000542-200309000-00027 (doi:10.1097/00000542-200309000-00027) [DOI] [PubMed] [Google Scholar]
  • 74.Kang S., Kim C. H., Lee H., Kim D. Y., Han J. I., Chung R. K., Lee G. Y. 2007. Antinociceptive synergy between the cannabinoid receptor agonist WIN 55,212-2 and bupivacaine in the rat formalin test. Anesth. Analg. 104, 719–725 10.1213/01.ane.0000255291.38637.26 (doi:10.1213/01.ane.0000255291.38637.26) [DOI] [PubMed] [Google Scholar]
  • 75.Guindon J., De Léan A., Beaulieu P. 2006. Local interactions between anandamide, an endocannabinoid, and ibuprofen, a nonsteroidal anti-inflammatory drug, in acute and inflammatory pain. Pain 121, 85–93 10.1016/j.pain.2005.12.007 (doi:10.1016/j.pain.2005.12.007) [DOI] [PubMed] [Google Scholar]
  • 76.Ruggieri V., Vitale G., Filaferro M., Frigeri C., Pini L. A., Sandrini M. 2010. The antinociceptive effect of acetylsalicylic acid is differently affected by a CB1 agonist or antagonist and involves the serotonergic system in rats. Life Sci. 86, 510–517 10.1016/j.lfs.2010.02.006 (doi:10.1016/j.lfs.2010.02.006) [DOI] [PubMed] [Google Scholar]
  • 77.Ahn D. K., Choi H. S., Yeo S. P., Woo Y. W., Lee M. K., Yang G. Y., Jeon H. J., Park J. S., Mokha S. S. 2007. Blockade of central cyclooxygenase (COX) pathways enhances the cannabinoid-induced antinociceptive effects on inflammatory temporomandibular joint (TMJ) nociception. Pain 132, 23–32 10.1016/j.pain.2007.01.015 (doi:10.1016/j.pain.2007.01.015) [DOI] [PubMed] [Google Scholar]
  • 78.Ulugöl A., Özyigit F., Yeşilyurt Ö., Dogrul A. 2006. The additive antinociceptive interaction between WIN 55,212-2, a cannabinoid agonist, and ketorolac. Anesth. Analg. 102, 443–447 10.1213/01.ane.0000194587.94260.1d (doi:10.1213/01.ane.0000194587.94260.1d) [DOI] [PubMed] [Google Scholar]
  • 79.Reche I., Fuentes J. A., Ruiz-Gayo M. 1996. Potentiation of Δ9-tetrahydrocannabinol-induced analgesia by morphine in mice: involvement of µ- and κ-opioid receptors. Eur. J. Pharmacol. 318, 11–16 10.1016/S0014-2999(96)00752-2 (doi:10.1016/S0014-2999(96)00752-2) [DOI] [PubMed] [Google Scholar]
  • 80.Pertwee R. G. 2001. Cannabinoid receptors and pain. Prog. Neurobiol. 63, 569–611 10.1016/S0301-0082(00)00031-9 (doi:10.1016/S0301-0082(00)00031-9) [DOI] [PubMed] [Google Scholar]
  • 81.Cichewicz D. L., McCarthy E. A. 2003. Antinociceptive synergy between Δ9-tetrahydrocannabinol and opioids after oral administration. J. Pharmacol. Exp. Ther. 304, 1010–1015 10.1124/jpet.102.045575 (doi:10.1124/jpet.102.045575) [DOI] [PubMed] [Google Scholar]
  • 82.Cichewicz D. L. 2004. Synergistic interactions between cannabinoid and opioid analgesics. Life Sci. 74, 1317–1324 10.1016/j.lfs.2003.09.038 (doi:10.1016/j.lfs.2003.09.038) [DOI] [PubMed] [Google Scholar]
  • 83.Tham S. M., Angus J. A., Tudor E. M., Wright C. E. 2005. Synergistic and additive interactions of the cannabinoid agonist CP55,940 with µ opioid receptor and α2-adrenoceptor agonists in acute pain models in mice. Br. J. Pharmacol. 144, 875–884 10.1038/sj.bjp.0706045 (doi:10.1038/sj.bjp.0706045) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cichewicz D. L., Welch S. P., Smith F. L. 2005. Enhancement of transdermal fentanyl and buprenorphine antinociception by transdermal Δ9-tetrahydrocannabinol. Eur. J. Pharmacol. 525, 74–82 10.1016/j.ejphar.2005.09.039 (doi:10.1016/j.ejphar.2005.09.039) [DOI] [PubMed] [Google Scholar]
  • 85.Williams I. J., Edwards S., Rubo A., Haller V. L., Stevens D. L., Welch S. P. 2006. Time course of the enhancement and restoration of the analgesic efficacy of codeine and morphine by Δ9-tetrahydrocannabinol. Eur. J. Pharmacol. 539, 57–63 10.1016/j.ejphar.2006.04.003 (doi:10.1016/j.ejphar.2006.04.003) [DOI] [PubMed] [Google Scholar]
  • 86.Cox M. L., Haller V. L., Welch S. P. 2007. Synergy between Δ9-tetrahydrocannabinol and morphine in the arthritic rat. Eur. J. Pharmacol. 567, 125–130 10.1016/j.ejphar.2007.04.010 (doi:10.1016/j.ejphar.2007.04.010) [DOI] [PubMed] [Google Scholar]
  • 87.Smith P. A., Selley D. E., Sim-Selley L. J., Welch S. P. 2007. Low dose combination of morphine and Δ9-tetrahydrocannabinol circumvents antinociceptive tolerance and apparent desensitization of receptors. Eur. J. Pharmacol. 571, 129–137 10.1016/j.ejphar.2007.06.001 (doi:10.1016/j.ejphar.2007.06.001) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Li J.-X., McMahon L. R., Gerak L. R., Becker G. L., France C. P. 2008. Interactions between Δ9-tetrahydrocannabinol and µ opioid receptor agonists in rhesus monkeys: discrimination and antinociception. Psychopharmacology 199, 199–208 10.1007/s00213-008-1157-0 (doi:10.1007/s00213-008-1157-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Welch S. P. 2009. Interaction of the cannabinoid and opioid systems in the modulation of nociception. Int. Rev. Psych. 21, 143–151 10.1080/09540260902782794 (doi:10.1080/09540260902782794) [DOI] [PubMed] [Google Scholar]
  • 90.Fischer B. D., Ward S. J., Henry F. E., Dykstra L. A. 2010. Attenuation of morphine antinociceptive tolerance by a CB1 receptor agonist and an NMDA receptor antagonist: interactive effects. Neuropharmacology 58, 544–550 10.1016/j.neuropharm.2009.08.005 (doi:10.1016/j.neuropharm.2009.08.005) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Fang Q., Han Z.-L., Li N., Wang Z.-L., He N., Wang R. 2012. Effects of neuropeptide FF system on CB1 and CB2 receptors mediated antinociception in mice. Neuropharmacology 62, 855–864 10.1016/j.neuropharm.2011.09.013 (doi:10.1016/j.neuropharm.2011.09.013) [DOI] [PubMed] [Google Scholar]
  • 92.Holdcroft A., Smith M., Jacklin A., Hodgson H., Smith B., Newton M., Evans F. 1997. Pain relief with oral cannabinoids in familial Mediterranean fever. Anaesthesia 52, 483–488 10.1111/j.1365-2044.1997.139-az0132.x (doi:10.1111/j.1365-2044.1997.139-az0132.x) [DOI] [PubMed] [Google Scholar]
  • 93.Narang S., Gibson D., Wasan A. D., Ross E. L., Michna E., Nedelikovic S. S., Jamison R. N. 2008. Efficacy of dronabinol as an adjuvant treatment for chronic pain patients on opioid therapy. J. Pain 9, 254–264 10.1016/j.jpain.2007.10.018 (doi:10.1016/j.jpain.2007.10.018) [DOI] [PubMed] [Google Scholar]
  • 94.Abrams D. I., Couey P., Shade S. B., Kelly M. E., Benowitz N. L. 2011. Cannabinoid-opioid interaction in chronic pain. Clin. Pharmacol. Ther. 90, 844–851 10.1038/clpt.2011.188 (doi:10.1038/clpt.2011.188) [DOI] [PubMed] [Google Scholar]
  • 95.Seeling W., et al. 2006. Δ9-tetrahydrocannabinol and the opioid receptor agonist piritramide do not act synergistically in postoperative pain. Anaesthesist 55, 391–400 10.1007/s00101-005-0963-6 (doi:10.1007/s00101-005-0963-6) [DOI] [PubMed] [Google Scholar]
  • 96.Naef M., Curatolo M., Petersen-Felix S., Arendt-Nielsen L., Zbinden A., Brenneisen R. 2003. The analgesic effect of oral Δ-9-tetrahydrocannabinol (THC), morphine, and a THC-morphine combination in healthy subjects under experimental pain conditions. Pain 105, 79–88 10.1016/S0304-3959(03)00163-5 (doi:10.1016/S0304-3959(03)00163-5) [DOI] [PubMed] [Google Scholar]
  • 97.Roberts J. D., Gennings C., Shih M. 2006. Synergistic affective analgesic interaction between Δ-9-tetrahydrocannabinol and morphine. Eur. J. Pharmacol. 530, 54–58 10.1016/j.ejphar.2005.11.036 (doi:10.1016/j.ejphar.2005.11.036) [DOI] [PubMed] [Google Scholar]
  • 98.Naderi N., Haghparast A., Saber-Tehrani A., Rezaii N., Alizadeh A.-M., Khani A., Motamedi F. 2008. Interaction between cannabinoid compounds and diazepam on anxiety-like behaviour of mice. Pharmacol. Biochem. Behav. 89, 64–75 10.1016/j.pbb.2007.11.001 (doi:10.1016/j.pbb.2007.11.001) [DOI] [PubMed] [Google Scholar]
  • 99.Valjent E., Mitchell J. M., Besson M.-J., Caboche J., Maldonado R. 2002. Behavioural and biochemical evidence for interactions between Δ9-tetrahydrocannabinol and nicotine. Br. J. Pharmacol. 135, 564–578 10.1038/sj.bjp.0704479 (doi:10.1038/sj.bjp.0704479) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Balerio G. N., Aso E., Maldonado R. 2006. Role of the cannabinoid system in the effects induced by nicotine on anxiety-like behaviour in mice. Psychopharmacology 184, 504–513 10.1007/s00213-005-0251-9 (doi:10.1007/s00213-005-0251-9) [DOI] [PubMed] [Google Scholar]
  • 101.Braida D., Limonta V., Malabarba L., Zani A., Sala M. 2007. 5-HT1A receptors are involved in the anxiolytic effect of Δ9-tetrahydrocannabinol and AM 404, the anandamide transport inhibitor, in Sprague-Dawley rats. Eur. J. Pharmacol. 555, 156–163 10.1016/j.ejphar.2006.10.038 (doi:10.1016/j.ejphar.2006.10.038) [DOI] [PubMed] [Google Scholar]
  • 102.Adamczyk P., Golda A., McCreary A. C., Filip M., Przegalński E. 2008. Activation of endocannabinoid transmission induces antidepressant-like effects in rats. J. Physiol. Pharmacol. 59, 217–228 [PubMed] [Google Scholar]
  • 103.Bahremand A., Shafaroodi H., Ghasemi M., Nasrabady S. E., Gholizadeh S., Dehpour A. R. 2008. The cannabinoid anticonvulsant effect on pentylenetetrazole-induced seizure is potentiated by ultra-low dose naltrexone in mice. Epilepsy Res. 81, 44–51 10.1016/j.eplepsyres.2008.04.010 (doi:10.1016/j.eplepsyres.2008.04.010) [DOI] [PubMed] [Google Scholar]
  • 104.Luszczki J. J., Andres-Mach M., Barcicka-Klosowska B., Florek-Luszczki M., Haratym-Maj A., Czuczwar S. J. 2011. Effects of WIN 55,212–2 mesylate (a synthetic cannabinoid) on the protective action of clonazepam, ethosuximide, phenobarbital and valproate against pentylenetetrazole-induced clonic seizures in mice. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 35, 1870–1876 10.1016/j.pnpbp.2011.07.001 (doi:10.1016/j.pnpbp.2011.07.001) [DOI] [PubMed] [Google Scholar]
  • 105.Luszczki J. J., Misiuta-Krzesinska M., Florek M., Tutka P., Czuczwar S. J. 2011. Synthetic cannabinoid WIN 55,212-2 mesylate enhances the protective action of four classical antiepileptic drugs against maximal electroshock-induced seizures in mice. Pharmacol. Biochem. Behav. 98, 261–267 10.1016/j.pbb.2011.01.002 (doi:10.1016/j.pbb.2011.01.002) [DOI] [PubMed] [Google Scholar]
  • 106.Luszczki J. J., Czuczwar P., Cioczek-Czuczwar A., Dudra-Jastrzebska M., Andres-Mach M., Czuczwar S. J. 2010. Effect of arachidonyl-2 ‘-chloroethylamide, a selective cannabinoid CB1 receptor agonist, on the protective action of the various antiepileptic drugs in the mouse maximal electroshock-induced seizure model. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 34, 18–25 10.1016/j.pnpbp.2009.09.005 (doi:10.1016/j.pnpbp.2009.09.005) [DOI] [PubMed] [Google Scholar]
  • 107.Naderi N., Ahari F. A., Shafaghi B., Najarkolaei A. H., Motamedi F. 2008. Evaluation of interactions between cannabinoid compounds and diazepam in electroshock-induced seizure model in mice. J. Neural. Transm. 115, 1501–1511 10.1007/s00702-008-0076-x (doi:10.1007/s00702-008-0076-x) [DOI] [PubMed] [Google Scholar]
  • 108.Bhattacharjee H., Nadipuram A., Kosanke S., Kiani M. F., Moore B. M. 2011. Low-volume binary drug therapy for the treatment of hypovolemia. Shock 35, 590–596 10.1097/SHK.0b013e3182150e80 (doi:10.1097/SHK.0b013e3182150e80) [DOI] [PubMed] [Google Scholar]
  • 109.Szczesniak A.-M., Maor Y., Robertson H., Hung O., Kelly M. E. M. 2011. Nonpsychotropic cannabinoids, abnormal cannabidiol and cannabigerol-dimethyl heptyl, act at novel cannabinoid receptors to reduce intraocular pressure. J. Ocular Pharmacol. Ther. 27, 427–435 10.1089/jop.2011.0041 (doi:10.1089/jop.2011.0041) [DOI] [PubMed] [Google Scholar]
  • 110.Torres S., Lorente M., Rodríguez-Fornés F., Hernández-Tiedra S., Salazar M., García-Taboada E., Barcia J., Guzmán M., Velasco G. 2011. A combined preclinical therapy of cannabinoids and temozolomide against glioma. Mol. Cancer Ther. 10, 90–103 10.1158/1535-7163.mct-10-0688 (doi:10.1158/1535-7163.mct-10-0688) [DOI] [PubMed] [Google Scholar]
  • 111.Kwiatkowska M., Parker L. A., Burton P., Mechoulam R. 2004. A comparative analysis of the potential of cannabinoids and ondansetron to suppress cisplatin-induced emesis in the Suncus murinus (house musk shrew). Psychopharmacology 174, 254–259 10.1007/s00213-003-1739-9 (doi:10.1007/s00213-003-1739-9) [DOI] [PubMed] [Google Scholar]
  • 112.Donadelli M., et al. 2011. Gemcitabine/cannabinoid combination triggers autophagy in pancreatic cancer cells through a ROS-mediated mechanism. Cell Death Dis. 2, e152. 10.1038/cddis.2011.36 (doi:10.1038/cddis.2011.36) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Paquette J. J., Wang H.-Y., Bakshi K., Olmstead M. C. 2007. Cannabinoid-induced tolerance is associated with a CB1 receptor G protein coupling switch that is prevented by ultra-low dose rimonabant. Behav. Pharmacol. 18, 767–776 10.1097/FBP.0b013e3282f15890 (doi:10.1097/FBP.0b013e3282f15890) [DOI] [PubMed] [Google Scholar]
  • 114.Gholizadeh S., Shafaroodi H., Ghasemi M., Bahremand A., Sharifzadeh M., Dehpour A. R. 2007. Ultra-low dose cannabinoid antagonist AM251 enhances cannabinoid anticonvulsant effects in the pentylenetetrazole-induced seizure in mice. Neuropharmacology 53, 763–770 10.1016/j.neuropharm.2007.08.005 (doi:10.1016/j.neuropharm.2007.08.005) [DOI] [PubMed] [Google Scholar]
  • 115.Bátkai S., et al. 2012. Δ8-Tetrahydrocannabivarin protects against hepatic ischemia/reperfusion injury by attenuating oxidative stress and inflammatory response via CB2 receptors. Br. J. Pharmacol. 165, 2450–2461 10.1111/j.1476-5381.2011.01410.x (doi:10.1111/j.1476-5381.2011.01410.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.García C., Palomo-Garo C., García-Arencibia M., Ramos J. A., Pertwee R. G., Fernandez-Ruiz J. 2011. Symptom-relieving and neuroprotective effects of the phytocannabinoid Δ9-THCV in animal models of Parkinson's disease. Br. J. Pharmacol. 163, 1495–1506 10.1111/j.1476-5381.2011.01278.x (doi:10.1111/j.1476-5381.2011.01278.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Marquart S., et al. 2010. Inactivation of the cannabinoid receptor CB1 prevents leukocyte infiltration and experimental fibrosis. Arthritis Rheum. 62, 3467–3476 10.1002/art.27642 (doi:10.1002/art.27642) [DOI] [PubMed] [Google Scholar]
  • 118.Zhang M., Martin B. R., Adler M. W., Razdan R. K., Ganea D., Tuma R. F. 2008. Modulation of the balance between cannabinoid CB1 and CB2 receptor activation during cerebral ischemic/reperfusion injury. Neuroscience 152, 753–760 10.1016/j.neuroscience.2008.01.022 (doi:10.1016/j.neuroscience.2008.01.022) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Le Foll B., Goldberg S. R. 2005. Cannabinoid CB1 receptor antagonists as promising new medications for drug dependence. J. Pharmacol. Exp. Ther. 312, 875–883 10.1124/jpet.104.077974 (doi:10.1124/jpet.104.077974) [DOI] [PubMed] [Google Scholar]
  • 120.Bolognini D., et al. 2010. The plant cannabinoid Δ9-tetrahydrocannabivarin can decrease signs of inflammation and inflammatory pain in mice. Br. J. Pharmacol. 160, 677–687 10.1111/j.1476-5381.2010.00756.x (doi:10.1111/j.1476-5381.2010.00756.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Pertwee R. G. 2008. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: Δ9-tetrahydrocannabinol, cannabidiol and Δ9-tetrahydrocannabivarin. Br. J. Pharmacol. 153, 199–215 10.1038/sj.bjp.0707442 (doi:10.1038/sj.bjp.0707442) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Manzanedo C., Aguilar M. A., Rodríguez-Arias M., Navarro M., Miñarro J. 2004. Cannabinoid agonist-induced sensitisation to morphine place preference in mice. Neuroreport 15, 1373–1377 10.1097/01.wnr.000012621787116.8c (doi:10.1097/01.wnr.000012621787116.8c) [DOI] [PubMed] [Google Scholar]
  • 123.Ellgren M., Spano S. M., Hurd Y. L. 2007. Adolescent cannabis exposure alters opiate intake and opioid limbic neuronal populations in adult rats. Neuropsychopharmacology 32, 607–615 10.1038/sj.npp.1301127 (doi:10.1038/sj.npp.1301127) [DOI] [PubMed] [Google Scholar]
  • 124.Pertwee R. G. 1992. In vivo interactions between psychotropic cannabinoids and other drugs involving central and peripheral neurochemical mediators. In Marijuana/cannabinoids: neurobiology and neurophysiology (eds Murphy L., Bartke A.), pp. 165–218, 1st edn. Boca Raton, FL: CRC Press [Google Scholar]
  • 125.Marchese G., Casti P., Ruiu S., Saba P., Sanna A., Casu G., Pani L. 2003. Haloperidol, but not clozapine, produces dramatic catalepsy in Δ9-THC-treated rats: possible clinical implications. Br. J. Pharmacol. 140, 520–526 10.1038/sj.bjp.0705478 (doi:10.1038/sj.bjp.0705478) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Tumati S., Largent-Milnes T. M., Keresztes A., Ren J. Y., Roeske W. R., Vanderah T. W., Varga E. V. 2012. Repeated morphine treatment-mediated hyperalgesia, allodynia and spinal glial activation are blocked by co-administration of a selective cannabinoid receptor type-2 agonist. J. Neuroimmunol. 244, 23–31 10.1016/j.jneuroim.2011.12.021 (doi:10.1016/j.jneuroim.2011.12.021) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Romero-Sandoval A., Eisenach J. C. 2007. Spinal cannabinoid receptor type 2 activation reduces hypersensitivity and spinal cord glial activation after paw incision. Anesthesiology 106, 787–794 10.1097/01.anes.0000264765.33673.6c (doi:10.1097/01.anes.0000264765.33673.6c) [DOI] [PubMed] [Google Scholar]
  • 128.Yamamoto W., Mikami T., Iwamura H. 2008. Involvement of central cannabinoid CB2 receptor in reducing mechanical allodynia in a mouse model of neuropathic pain. Eur. J. Pharmacol. 583, 56–61 10.1016/j.ejphar.2008.01.010 (doi:10.1016/j.ejphar.2008.01.010) [DOI] [PubMed] [Google Scholar]
  • 129.Wilkerson J. L., et al. 2012. Intrathecal cannabilactone CB2R agonist, AM1710, controls pathological pain and restores basal cytokine levels. Pain 153, 1091–1106 10.1016/j.pain.2012.02.015 (doi:10.1016/j.pain.2012.02.015) [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Philosophical Transactions of the Royal Society B: Biological Sciences are provided here courtesy of The Royal Society

RESOURCES