Keywords

These keywords were added by machine and not by the authors. This process is experimental and the keywords may be updated as the learning algorithm improves.

Key Points

  • Cannabinoids are pharmacological agents of endogenous (endocannabinoids), botanical (phytocannabinoids), or synthetic origin.

  • Cannabinoids alleviate pain through a variety of receptor and non-receptor mechanisms including direct analgesic and anti-inflammatory effects, modulatory actions on neurotransmitters, and interactions with endogenous and administered opioids.

  • Cannabinoid agents are currently available in various countries for pain treatment, and even cannabinoids of botanical origin may be approvable by FDA, although this is distinctly unlikely for smoked cannabis.

  • An impressive body of literature supports cannabinoid analgesia, and recently, this has been supplemented by an increasing number of phase I–III clinical trials.

Introduction

Plants and Pain

It is a curious fact that we owe a great deal of our insight into pharmacological treatment of pain to the plant world [1]. Willow bark from Salix spp. led to development of aspirin and eventual elucidation of the analgesic effects of ­prostaglandins and their role in inflammation. The opium poppy (Papaver somniferum) provided the prototypic narcotic analgesic morphine, the first alkaloid discovered, and stimulated the much later discovery of the endorphin and enkephalin systems. Similarly, the pharmacological properties of cannabis (Cannabis sativa) prompted the isolation of Δ9-tetrahydrocannabinol (THC), the major psychoactive ingredient in cannabis, in 1964 [2]. It is this breakthrough that subsequently prompted the more recent discovery of the body’s own cannabis-like system, the endocannabinoid system (ECS), which modulates pain under physiological conditions. Pro-nociceptive mechanisms of the endovanilloid system were similarly revealed by phytochemistry of capsaicin, the pungent ingredient in hot chile peppers (Capsicum annuum etc.), which activates transient receptor potential vanilloid receptor-1 (TRPV1). Additional plant products such as the mints and mustards activate other TRP channels to produce their physiological effects.

The Endocannabinoid System

There are three recognized types of cannabinoids: (1) the phytocannabinoids [3] derived from the cannabis plant, (2) synthetic cannabinoids (e.g., ajulemic acid, nabilone, CP55940, WIN55, 212-2) based upon the chemical structure of THC or other ligands which bind cannabinoid receptors, and (3) the endogenous cannabinoids or endocannabinoids. Endocannabinoids are natural chemicals such as anandamide (AEA) and 2-arachidonoylglycerol (2-AG) found in animals whose basic functions are “relax, eat, sleep, forget, and ­protect” [4]. The endocannabinoid system encompasses the endocannabinoids themselves, their biosynthetic and catabolic enzymes, and their corresponding receptors [5]. AEA is hydrolyzed by the enzyme fatty-acid amide hydrolase (FAAH) into breakdown products arachidonic acid and ethanolamine [6]. By contrast, 2-AG is hydrolyzed primarily by the enzyme monoacylglycerol lipase (MGL) into breakdown products arachidonic acid and glycerol [7] and to a lesser extent by the enzymes ABHD6 and ABHD12. FAAH, a postsynaptic enzyme, may control anandamide levels near sites of synthesis, whereas MGL, a presynaptic enzyme [8], may terminate 2-AG signaling following CB1 receptor activation. These enzymes also represent therapeutic targets because inhibition of endocannabinoid deactivation will increase levels of endocannabinoids at sites with ongoing synthesis and release [9]. The pathways controlling formation of AEA remain poorly understood. However, 2-AG is believed to be formed from membrane phospholipid precursors through the sequential activation of two distinct enzymes, phospholipase C and diacylglycerol lipase-α. First, PLC catalyzes formation of the 2-AG precursor diacylglycerol (DAG) from membrane phosphoinositides. Then, DAG is hydrolyzed by the enzyme diacylglycerol lipase-α (DGL-α) to generate 2-AG [199].

There are currently two well-defined cannabinoid receptors, although additional candidate cannabinoid receptors have also been postulated. CB1, a seven transmembrane spanning G-protein-coupled receptor inhibiting cyclic AMP release, was identified in 1988 [10]. CB1 is the primary neuromodulatory receptor accounting for psychopharmacological effects of THC and most of its analgesic effects [11]. Endocannabinoids are produced on demand in postsynaptic cells and engage presynaptic CB1 receptors through a retrograde mechanism [12]. Activation of presynaptic CB1 receptors then acts as a synaptic circuit breaker to inhibit neurotransmitter release (either excitatory or inhibitory) from the presynaptic neuron (vide infra) (Fig. 18.1). CB2 was identified in 1992, and while thought of primarily as a peripheral immunomodulatory receptor, it also has important effects on pain. The role of CB2 in modulating persistent inflammatory and neuropathic pain [13] has been recently reviewed [14, 15]. Activation of CB2 suppresses neuropathic pain mechanisms through nonneuronal (i.e., microglia and astrocytes) and neuronal mechanisms that may involve interferon-gamma [16]. THC, the prototypical classical cannabinoid, is a weak partial agonist at both CB1 and CB2 receptors. Transgenic mice lacking cannabinoid receptors (CB1, CB2, GPR55), enzymes controlling endocannabinoid breakdown (FAAH, MGL, ABHD6), and endocannabinoid synthesis (DGL-α, DGL-β) have been generated [17]. These knockouts have helped elucidate the role of the endocannabinoid system in controlling nociceptive processing and facilitated development of inhibitors of endocannabinoid breakdown (FAAH, MGL) as novel classes of analgesics.

Fig. 18.1
figure 1

Putative mechanism of endocannabinoid-mediated ­retrograde signaling in the nervous system. Activation of metabotropic glutamate receptors (mGluR) by glutamate triggers the activation of the phospholipase C (PLC)-diacylglycerol lipase (DGL) pathway to generate the endocannabinoid 2-arachidonoylglycerol (2-AG). First, the 2-AG precursor diacylglycerol (DAG) is formed from PLC-mediated hydrolysis of membrane phospholipid precursors (PIPx). DAG is then hydrolyzed by the enzyme DGL-α to generate 2-AG. 2-AG is released from the postsynaptic neuron and acts as a retrograde signaling ­molecule. Endocannabinoids activate presynaptic CB1 receptors which reside on terminals of glutamatergic and GABAergic neurons. Activation of CB1 by 2-AG, anandamide, or exogenous cannabinoids (e.g., ­tetrahydrocannabinol, THC) inhibits calcium influx in the presynaptic terminal, thereby inhibiting release of the primary neurotransmitter (i.e., glutamate or GABA) from the synaptic vesicle. Endocannabinoids are then rapidly deactivated by transport into cells (via a putative endocannabinoid transporter) followed by intracellular hydrolysis. 2-AG is metabolized by the enzyme monoacylglycerol lipase (MGL), whereas anandamide is metabolized by a distinct enzyme, fatty-acid amide hydrolase (FAAH). Note that MGL co-localizes with CB1 in the presynaptic terminal, whereas FAAH is localized to postsynaptic sites. The existence of an endocannabinoid transporter remains controversial. Pharmacological inhibitors of either endocannabinoid deactivation (e.g., FAAH and MGL inhibitors) or transport (i.e., uptake inhibitors) have been developed to exploit the therapeutic potential of the endocannabinoid signaling system in the treatment of pain (Figure by authors with kind assistance of James Brodie, GW Pharmaceuticals)

A Brief Scientific History of Cannabis and Pain

Centuries of Citations

Cannabis has been utilized in one form or another for treatment of pain for longer than written history [1821]. Although this documentation has been a major preoccupation of the lead author [2225], and such information can provide provocative direction to inform modern research on treatment of pain and other conditions, it does not represent evidence of form, content, or degree that is commonly acceptable to governmental regulatory bodies with respect to pharmaceutical development.

Anecdotes Versus Modern Proof of Concept

While thousands of compelling stories of efficacy of cannabis in pain treatment certainly underline the importance of properly harnessing cannabinoid mechanisms therapeutically [26, 27], prescription analgesics in the United States necessitate Food and Drug Administration (FDA) approval. This requires a rigorous development program proving consistency, quality, efficacy, and safety as defined by basic scientific studies and randomized controlled trials (RCT) [28] and generally adhering to recent IMMPACT recommendations [29], provoking our next question.

Can a Botanical Agent Become a Prescription Medicine?

Most modern physicians fail to recognize that pharmacognosy (study of medicinal plants) has led directly or indirectly to an estimated 25 % of modern pharmaceuticals [30]. While the plethora of available herbal agents yield an indecipherable cacophony to most clinicians and consumers alike, it is certainly possible to standardize botanical agents and facilitate their recommendation based on sound science [31]. Botanical medicines can even fulfill the rigorous dictates of the FDA and attain prescription drug status via a clear roadmap in the form of a blueprint document [32], henceforth termed the Botanical Guidance: http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm070491.pdf. To be successful and clinically valuable, botanicals, including cannabis-based medicines, must demonstrate the same quality, clinical analgesic benefit, and appropriately safe adverse event profile as available new chemical entities (NCE) [28].

The Biochemical and Neurophysiological Basis of Pain Control by Cannabinoids

Neuropathic Pain

Thorough reviews of therapeutic effects of cannabinoids in preclinical and clinical domains have recently been published [33, 34]. In essence, the endocannabinoid system (ECS) is active throughout the CNS and PNS in modulating pain at spinal, supraspinal, and peripheral levels. Endocannabinoids are produced on demand in the CNS to dampen sensitivity to pain [35]. The endocannabinoid system is operative in such key integrative pain centers as the periaqueductal grey matter [36, 37], the ventroposterolateral nucleus of the thalamus [38], and the spinal cord [39, 40]. Endocannabinoids are endogenous mediators of stress-induced analgesia and fear-conditioned analgesia and suppress pain-related phenomena such as windup [41] and allodynia [42]. In the periphery and PNS [13], the ECS has key effects in suppressing both hyperalgesia and allodynia via CB1 [43] and CB2 mechanisms (Fig. 18.2). Indeed, pathological pain states have been postulated to arise, at least in part, from a dysregulation of the endocannabinoid system.

Fig. 18.2
figure 2

Cannabinoids suppress pain and other pathophysiological (e.g., contact dermatitis, pruritis) and physiological (e.g., gastrointestinal transit and secretion) processes through multiple mechanisms involving CB1 and CB2 receptors. Peripheral, spinal, and supraspinal sites of cannabinoid actions are shown. In the periphery, cannabinoids act through both neuronal and nonneuronal mechanisms to control inflammation, allodynia, and hyperalgesia. CB1 and CB2 have been localized to both primary afferents and nonneuronal cells (e.g., keratinocytes, microglia), and expression can be regulated by injury. In the spinal cord, cannabinoids suppress nociceptive transmission, windup, and central sensitization by modulating activity in the ascending pain pathway of the spinothalamic tract, including responses of wide dynamic range (WDR) and nociceptive specific (NS) cells. Similar processes are observed at rostral levels of the neuraxis (e.g., ventroposterolateral nucleus of the thalamus, amygdala, anterior cingulate cortex). Cannabinoids also actively modulate pain through descending mechanisms. In the periaqueductal gray, cannabinoids act through presynaptic glutamatergic and GABAergic mechanisms to control nociception. In the rostral ventromedial medulla, cannabinoids suppress activity in ON cells and inhibit the firing pause of OFF cells, in response to noxious stimulation to produce antinociception (Figure by authors with kind assistance of James Brodie, GW Pharmaceuticals)

Antinociceptive and Anti-inflammatory Pain Mechanisms

Beyond the mechanisms previously mentioned, the ECS plays a critical role in peripheral pain, inflammation, and hyperalgesia [43] through both CB1 and CB2 mechanisms. CB1 and CB2 mechanisms are also implicated in regulation of contact dermatitis and pruritus [44]. A role for spinal CB2 mechanisms, mediated by microglia and/or astrocytes, is also revealed under conditions of inflammation [45]. Both THC and cannabidiol (CBD), a non-euphoriant phytocannabinoid common in certain cannabis strains, are potent anti-inflammatory antioxidants with activity exceeding that of vitamins C and E via non-cannabinoid mechanisms [46]. THC inhibits prostaglandin E-2 synthesis [47] and stimulates lipooxygenase [48]. Neither THC nor CBD affects COX-1 or COX-2 at relevant pharmacological dosages [49].

While THC is inactive at vanilloid receptors, CBD, like AEA, is a TRPV1 agonist. Like capsaicin, CBD is capable of inhibiting fatty-acid amide hydrolase (FAAH), the enzyme which hydrolyzes AEA and other fatty-acid amides that do not bind to cannabinoid receptors. CBD additionally inhibits AEA reuptake [50] though not potently. Thus, CBD acts as an endocannabinoid modulator [51], a mechanism that various pharmaceutical firms hope to emulate with new chemical entities (NCEs). CBD inhibits hepatic metabolism of THC to 11-hydroxy-THC, which is possibly more psychoactive, and prolongs its half-life, reducing its psychoactivity and attenuating attendant anxiety and tachycardia [51]; antagonizes psychotic symptoms [52]; and attenuates appetitive effects of THC [53] as well as its effects on short-term memory [54]. CBD also inhibits tumor necrosis factor-alpha (TNF-α) in a rodent model of rheumatoid arthritis [55]. Recently, CBD has been demonstrated to enhance adenosine receptor A2A signaling via inhibition of the adenosine transporter [56].

Recently, GPR18 has been proposed as a putative CBD receptor whose function relates to cellular migration [57]. Antagonism of GPR18 (by agents such as CBD) may be efficacious in treating pain of endometriosis, among other conditions, especially considering that such pain may be endocannabinoid-mediated [58]. Cannabinoids are also very active in various gastrointestinal and visceral sites mediating pain responses [59, 60].

Cannabinoid Interactions with Other Neurotransmitters Pertinent to Pain

As alluded to above, the ECS modulates neurotransmitter release via retrograde inhibition. This is particularly important in NMDA-glutamatergic mechanisms that become hyperresponsive in chronic pain states. Cannabinoids specifically inhibit glutamate release in the hippocampus [61]. THC reduces NMDA responses by 30–40 % [46]. Secondary and tertiary hyperalgesia mediated by NMDA [62] and by calcitonin gene-related peptide [40] may well be targets of cannabinoid therapy in disorders such as migraine, fibromyalgia, and idiopathic bowel syndrome wherein these mechanisms seem to operate pathophysiologically [63], prompting the hypothesis of a “clinical endocannabinoid deficiency.” Endocannabinoid modulators may therefore restore homeostasis, leading to normalization of function in these pathophysiological conditions. THC also has numerous effects on serotonergic systems germane to migraine [64], increasing its production in the cerebrum while decreasing reuptake [65]. In fact, the ECS seems to modulate the trigeminovascular system of migraine pathogenesis at ­vascular and neurochemical levels [6668].

Cannabinoid-Opioid Interactions

Although endocannabinoids do not bind to opioid receptors, the ECS may nonetheless work in parallel with the endogenous opioid system with numerous areas of overlap and interaction. Pertinent mechanisms include stimulation of beta-endorphin by THC [69] as well as its ability to demonstrate experimental opiate sparing [70], prevent opioid tolerance and withdrawal [71], and rekindle opioid analgesia after loss of effect [72]. Adjunctive treatments that combine opioids with cannabinoids may enhance the analgesic effects of either agent. Such strategies may permit lower doses of analgesics to be employed for therapeutic benefit in a manner that minimizes incidence or severity of adverse side effects.

Clinical Trials, Utility, and Pitfalls of Cannabinoids in Pain

Evidence for Synthetic Cannabinoids

Oral dronabinol (THC) has been available as the synthetic Marinol® since 1985 and is indicated for nausea associated with chemotherapy and appetite stimulation in HIV/AIDS. Issues with its cost, titration difficulties, delayed onset, and propensity to induce intoxicating and dysphoric effects have limited clinical application [73]. It was employed in two open-label studies of chronic neuropathic pain in case studies in 7 [74] and 8 patients [75], but no significant benefit was evident and side effects led to prominent dropout rates (average doses 15–16.6 mg THC). Dronabinol produced benefit in pain in multiple sclerosis [76], but none was evident in postoperative pain (Table 18.1) [77]. Dronabinol was reported to relieve pruritus in three case-report subjects with cholestatic jaundice [78]. Dronabinol was assessed in 30 chronic non-cancer pain patients on opioids in double-blind crossover single-day sessions vs. placebo with improvement [79], followed by a 4-week open-label trial with continued improvement (Table 18.1). Associated adverse events were prominent. Methodological issues included lack of prescreening for cannabinoids, 4 placebo subjects with positive THC assays, and 58 % of subjects correctly guessing Marinol dose on test day. An open-label comparison in polyneuropathy examined nabilone patients with 6 obtaining 22.6 % mean pain relief after 3 months, and 5 achieving 28.6 % relief after 6 months, comparable to conventional agents [80]. A pilot study of Marinol in seven spinal cord injury patients with neuropathic pain saw two withdraw, and the remainder appreciate no greater efficacy than with diphenhydramine [81].

Table 18.1 Randomized controlled trials of cannabinoids in pain

Nabilone, or Cesamet®, is a semisynthetic analogue of THC that is about tenfold more potent, and longer lasting [82]. It is indicated as an antiemetic in chemotherapy in the USA. Prior case reports in neuropathic pain [83] and other pain disorders [84] have been published. Sedation and dysphoria are prominent associated adverse events. An RCT of nabilone in 41 postoperative subjects dosed TID actually resulted in increased pain scores (Table 18.1) [85]. An uncontrolled study of 82 cancer patients on nabilone noted improved pain scores [86], but retention rates were limited. Nabilone improved pain (p  <  0.05) vs. placebo in patients with mixed spasticity syndromes in a small double-blind trial (Table 18.1) [87], but was without benefits in other parameters. In a double-blind crossover comparison of nabilone to dihydrocodeine (schedule II opioid) in chronic neuropathic pain (Table 18.1) [88], both drugs produced marginal benefit, but with dihydrocodeine proving clearly superior in efficacy and modestly superior in side-effect profile. In an RCT in 40 patients of nabilone vs. placebo over 4 weeks, it showed significant decreases in VAS of pain and anxiety (Table 18.1) [89]. A more recent study of nabilone vs. amitriptyline in fibromyalgia yielded benefits on sleep, but not pain, mood, or quality of life (Table 18.1) [90]. An open-label trial of nabilone vs. gabapentin found them comparable in pain and other symptom relief in peripheral neuropathic pain [91].

Ajulemic acid (CT3), another synthetic THC analogue in development, was utilized in a phase II RCT in peripheral neuropathic pain in 21 subjects with apparent improvement (Table 18.1) [92]. Whether or not ajulemic acid is psychoactive is the subject of some controversy [93].

Evidence for Smoked or Vaporized Cannabis

Few randomized controlled clinical trials (RCTs) of pain with smoked cannabis have been undertaken to date [9497]. One of these [96] examined cannabis effects on experimental pain in normal volunteers.

Abrams et al. [94] studied inpatient adults with painful HIV neuropathy in 25 subjects in double-blind fashion to receive either smoked cannabis as 3.56 % THC cigarettes or placebo cigarettes three times daily for 5 days (Table 18.1). The smoked cannabis group had a 34 % reduction in daily pain vs. 17 % in the placebo group (p  =  0.03). The cannabis cohort also had a 52 % of subjects report a >30 % reduction in pain scores over the 5 days vs. 24 % in the placebo group (p  =  0.04) (Table 18.1). The authors rated cannabis as “well tolerated” due to an absence of serious adverse events (AE) leading to withdrawal, but all subjects were cannabis experienced. Symptoms of possible intoxication in the cannabis group including anxiety (25 %), sedation (54 %), disorientation (16 %), paranoia (13 %), confusion (17 %), dizziness (15 %), and nausea (11 %) were all statistically significantly more common than in the placebo group. Despite these findings, the authors stated that the values do not represent any serious safety concern in this short-term study. No discussion in the article addressed issues of the relative efficacy of blinding in the trial.

Wilsey et al. [95] examined neuropathic pain in 38 subjects in a double-blind crossover study comparing 7 % THC cannabis, 3.5 % THC cannabis, and placebo cigarettes via a complex cumulative dosing scheme with each dosage given once, in random order, with at least 3 day intervals separating sessions (Table 18.1). A total of 9 puffs maximum were allowed over several hours per session. Authors stated, “Psychoactive effects were minimal and well-tolerated, but neuropsychological impairment was problematic, particularly with the higher concentration of study medication.” Again, only cannabis-experienced subjects were allowed entry. No withdrawals due to AE were reported, but 1 subject was removed due to elevated blood pressure. No significant differences were noted in pain relief in the two cannabis potency groups, but a significant separation of pain reduction from placebo (p  =  0.02) was not evident until a cumulative 9 puffs at 240 min elapsed time. Pain unpleasantness was also reduced in both active treatment groups (p  <  0.01). Subjectively, an “any drug effect” demonstrated a visual analogue scale (VAS) of 60/100 in the high-dose group, but even the low-dose group registered more of a “good drug effect” than placebo (p  <  0.001). “Bad drug effect” was also evident. “Feeling high” and “feeling stoned” were greatest in the high-dose sessions (p  <  0.001), while both high- and low-dose differentiated significantly from placebo (p  <  0.05). Of greater concern, both groups rated impairment as 30/100 on VAS vs. placebo (p  =  0.003). Sedation also demarcated both groups from placebo (p  <  0.01), as did confusion (p  =  0.03), and hunger (p  <  0.001). Anxiety was not considered a prominent feature in this cannabis-experienced population. This study distinguished itself from some others in its inclusion of specific objective neuropsychological measures and demonstrated neurocognitive impairment in attention, learning, and memory, most noteworthy with 7 % THC cannabis. No commentary on blinding efficacy was included.

Ellis et al. [97] examined HIV-associated neuropathic pain in a double-blind trial of placebo vs. 1–8 % THC cannabis administered four times daily over 5 days with a 2-week washout (Table 18.1). Subjects were started at 4 % THC and then titrated upward or downward in four smoking sessions dependent upon their symptom relief and tolerance of the dose. In this study, 96 % of subjects were cannabis-experienced, and 28 out of 34 subjects completed the trial. The primary outcome measure (Descriptor Differential Scale, DDS) was improved in the active group over placebo (p  =  0.016), with >30 % relief noted in 46 % of cannabis subjects vs. 18 % of placebo. While most adverse events (AE) were considered mild and self-limited, two subjects had to leave the trial due to toxicity. One cannabis-naïve subject was withdrawn due to “an acute cannabis-induced psychosis” at what proved to be his first actual cannabis exposure. The other subject suffered intractable cough. Pain reduction was greater in the cannabis-treated group (p  =  0.016) among completers, as was the proportion of subjects attaining >30 % pain reduction (46 % vs. 18 %, p  =  0.043). Blinding was assessed in this study; whereas placebo patients were inaccurate at guessing the investigational product, 93 % of those receiving cannabis guessed correctly. On safety issues, the authors stated that the frequency of some nontreatment-limiting side effects was greater for cannabis than placebo. These included concentration difficulties, fatigue, sleepiness or sedation, increased duration of sleep, reduced salivation, and thirst.

A Canadian study [98] examined single 25-mg inhalations of various cannabis potencies (0–9.4 % THC) three times daily for 5 days per cycle in 23 subjects with chronic neuropathic pain (Table 18.1). Patients were said to be cannabis-free for 1 year, but were required to have some experience of the drug. Only the highest potency demarcated from placebo on decrements in average daily pain score (5.4 vs. 6.1, p  =  0.023). The most frequent AE in the high-dose group were headache, dry eyes, burning sensation, dizziness, numbness, and cough, but with “high” or “euphoria” reported only once in each cannabis potency group.

The current studies of smoked cannabis are noteworthy for their extremely short-term exposure and would be of uncertain relevance in a regulatory environment. The IMMPACT recommendations on chronic neuropathic pain clinical trials that are currently favored by the FDA [29] generally suggest randomized controlled clinical trials of 12-week duration as a prerequisite to demonstrate efficacy and safety. While one might assume that the degree of pain improvement demonstrated in these trials could be maintained over this longer interval, it is only reasonable to assume that cumulative adverse events would also increase to at least some degree. The combined studies represent only a total of 1,106 patient-days of cannabis exposure (Abrams: 125, Wilsey: 76, Ellis: 560, Ware 345) or 3 patient-years of experience. In contrast, over 6,000 patient-years of data have been analyzed for Sativex between clinical trials, prescription, and named-patient supplies, with vastly lower AE rates (data on file, GW Pharmaceuticals) [28, 99]. Certainly, the cognitive effects noted in California-smoked cannabis studies figure among many factors that would call the efficacy of blinding into question for investigations employing such an approach. However, it is also important to emphasize that unwanted side effects are not unique to cannabinoids. In a prospective evaluation of specific chronic polyneuropathy syndromes and their response to pharmacological therapies, the presence of intolerable side effects did not differ in groups receiving gabapentinoids, tricyclic antidepressants, anticonvulsants, cannabinoids (including nabilone, Sativex), and topical agents [80]. Moreover, no serious adverse events were related to any of the medications.

The current studies were performed in a very select subset of patients who almost invariably have had prior experience of cannabis. Their applicability to cannabis-naïve populations is, thus, quite unclear. At best, the observed benefits might possibly accrue to some, but it is eminently likely that candidates for such therapy might refuse it on any number of grounds: not wishing to smoke, concern with respect to intoxication, etc. Sequelae of smoking in therapeutic outcomes have had little discussion in these brief RCTs [28]. Cannabis smoking poses substantial risk of chronic cough and bronchitic symptoms [100], if not obvious emphysematous degeneration [101] or increase in aerodigestive cancers [102]. Even such smoked cannabis proponents as Lester Grinspoon has acknowledged are the only well-confirmed deleterious physical effect of marihuana is harm to the pulmonary system [103]. However, population-based studies of cannabis trials have failed to show any evidence for increased risk of respiratory symptoms/chronic obstructive pulmonary disease [100] or lung cancer [102] associated with smoking cannabis.

A very detailed analysis and comparison of mainstream and sidestream smoke for cannabis vs. tobacco smoke was performed in Canada [104]. Of note, cannabis smoke contained ammonia (NH3) at a level of 720 μg per 775 mg cigarette, a figure 20-fold higher than that found in tobacco smoke. It was hypothesized that this finding was likely attributable to nitrate fertilizers. Formaldehyde and acetaldehyde were generally lower in cannabis smoke than in tobacco, but butyraldehyde was higher. Polycyclic aromatic hydrocarbon (PAH) contents were qualitatively similar in the comparisons, but total yield was lower for cannabis mainstream smoke, but higher than tobacco for sidestream smoke. Additionally, NO, NO x , hydrogen cyanide, and aromatic amines concentrations were 3–5 times higher in cannabis smoke than that from tobacco. Possible mutagenic and carcinogenic potential of these various compounds were mentioned. More recently, experimental analysis of cannabis smoke with resultant acetaldehyde production has posited its genotoxic potential to be attributable to reactions that produce DNA adducts [105].

Vaporizers for cannabis have been offered as a harm reduction technique that would theoretically eliminate products of combustion and associated adverse events. The Institute of Medicine (IOM) examined cannabis issues in 1999 [106], and among their conclusions was the following (p. 4): “Recommendation 2: Clinical trials of cannabinoid drugs for symptom management should be conducted with the goal of developing rapid-onset, reliable, and safe delivery systems.” One proposed technique is vaporization, whereby cannabis is heated to a temperature that volatilizes THC and other components with the goal of reducing or eliminating by-products of combustion, including potentially carcinogenic polycyclic aromatic hydrocarbons, benzene, acetaldehyde, carbon monoxide, toluene, naphthaline, phenol, toluene, hydrogen cyanide, and ammonia. Space limitations permit only a cursory review of available literature [107115].

A pilot study of the Volcano vaporizer vs. smoking was performed in the USA in 2007 in 18 active cannabis consumers, with only 48 h of presumed abstinence [116]. NIDA 900-mg cannabis cigarettes were employed (1.7, 3.4, and 6.8 % THC) with each divided in two, so that one-half would be smoked or vaporized in a series of double-blind sessions. The Volcano vaporizer produced comparable or slightly higher THC plasma concentrations than smoking. Measured CO in exhaled vapor sessions diminished very slightly, while it increased after smoking (p  <  0.001). Self-reported visual analogue scales of the associated high were virtually identical in vaporization vs. smoking sessions and increased with higher potency material. A contention was advanced that the absence of CO increase after vaporization can be equated to “little or no exposure to gaseous combustion toxins.” Given that no measures of PAH or other components were undertaken, the assertion is questionable. It was also stated that there were no reported adverse events. Some 12 subjects preferred the Volcano, 2 chose smoking, and 2 had no preference as to technique, making the vaporizer “an acceptable system” and providing “a safer way to deliver THC.”

A recent [202, 117] examined interactions of 3.2 % THC NIDA cannabis vaporized in the Volcano in conjunction with opioid treatment in a 5-day inpatient trial in 21 patients with chronic pain (Table 18.1). All subjects were prior cannabis smokers. Overall, pain scores were reduced from 39.6 to 29.1 on a VAS, a 27 % reduction, by day 5. Pain scores in subjects on morphine fell from 34.8 to 24.1, while in subjects taking oxycodone, scores dropped from 43.8 to 33.6.

The clinical studies performed with vaporizers to date have been very small pilot studies conducted over very limited timeframes (i.e., for a maximum of 5 days). Thus, these studies cannot contribute in any meaningful fashion toward possible FDA approval of vaporized cannabis as a delivery technique, device, or drug under existing policies dictated by the Botanical Guidance [32]. It is likewise quite unlikely that the current AE profile of smoked or vaporized cannabis would meet FDA requirements. The fact that all the vaporization trials to date have been undertaken only in cannabis-experienced subjects does not imply that results would generalize to larger patient populations. Moreover, there is certainly no reason to expect AE profiles to be better in cannabis-naïve patients. Additionally, existing standardization of cannabis product and delivery via vaporization seem far off the required marks. Although vaporizers represent an alternate delivery method devoid of the illegality associated with smoked cannabis, the presence of toxic ingredients such as PAH, ammonia, and acetaldehyde in cannabis vapor are unlikely to be acceptable to FDA in any significant amounts. Existing vaporizers still lack portability or convenience [28]. A large Internet survey revealed that only 2.2 % of cannabis users employed vaporization as their primary cannabis intake method [118]. While studies to date have established that lower temperature vaporization in the Volcano, but not necessarily other devices, can reduce the relative amounts of noxious by-products of combustion, it has yet to be ­demonstrated that they are totally eliminated. Until or unless this goal is achieved, along with requisite benchmarks of herbal cannabis quality, safety, and efficacy in properly designed randomized clinical trials, vaporization remains an unproven technology for therapeutic cannabinoid administration.

Evidence for Cannabis-Based Medicines

Cannador is a cannabis extract in oral capsules, with differing THC:CBD ratios [51]. Cannador was utilized in a phase III RCT of spasticity in multiple sclerosis (CAMS) (Table 18.1) [119]. While no improvement was evident in the Ashworth Scale, reduction was seen in spasm-associated pain. Both THC and Cannador improved pain scores in follow-up [120]. Cannador was also employed for postherpetic neuralgia in 65 patients, but without success (Table 18.1) [121, 122]. Slight pain reduction was observed in 30 subjects with postoperative pain (CANPOP) not receiving opiates, but psychoactive side effects were notable (Table 18.1).

Sativex® is a whole-cannabis-based extract delivered as an oromucosal spray that combines a CB1 and CB2 partial agonist (THC) with a cannabinoid system modulator (CBD), minor cannabinoids, and terpenoids plus ethanol and propylene glycol excipients and peppermint flavoring [51, 123]. It is approved in Canada for spasticity in MS and under a Notice of Compliance with Conditions for central neuropathic pain in multiple sclerosis and treatment of cancer pain unresponsive to opioids. Sativex is also approved in MS in the UK, Spain, and New Zealand, for spasticity in multiple sclerosis, with further approvals expected soon in some 22 countries around the world. Sativex is highly standardized and is formulated from two Cannabis sativa chemovars predominating in THC and CBD, respectively [124]. Each 100 μl pump-action oromucosal spray of Sativex yields 2.7 mg of THC and 2.5 mg of CBD plus additional components. Pharmacokinetic data are available [125127]. Sativex effects begin within an interval allowing dose titration. A very favorable adverse event profile has been observed in the development program [27, 128]. Most patients stabilize at 8–10 sprays per day after 7–10 days, attaining symptomatic control without undue psychoactive sequelae. Sativex was added to optimized drug regimens in subjects with uncontrolled pain in every RCT (Table 18.1). An Investigational New Drug (IND) application to study Sativex in advanced clinical trials in the USA was approved by the FDA in January 2006 in patients with intractable cancer pain. One phase IIB dose-ranging study has already been completed [201]. Available clinical trials with Sativex have been independently assessed [129, 130].

In a phase II study of 20 patients with neurogenic symptoms [131], significant improvement was seen with both Tetranabinex (high-THC extract without CBD) and Sativex on pain, with Sativex displaying better symptom control (p  <  0.0001), with less intoxication (Table 18.1).

In a phase II study of intractable chronic pain in 24 patients [132], Sativex again produced the best results compared to Tetranabinex (p  <  0.001), especially in MS (p  <  0.0042) (Table 18.1).

In a phase III study of brachial plexus avulsion (N  =  48) [133], pain reduction with Tetranabinex and Sativex was about equal (Table 18.1).

In an RCT of 66 MS subjects, mean Numerical Rating Scale (NRS) analgesia favored Sativex over placebo (Table 18.1) [134].

In a phase III trial (N  =  125) of peripheral neuropathic pain with allodynia [135], Sativex notably alleviated pain levels and dynamic and punctate allodynia (Table 18.1).

In a safety-extension study in 160 subjects with various symptoms of MS [136], 137 patients showed sustained improvements over a year or more in pain and other symptoms [99] without development of any tolerance requiring dose escalation or withdrawal effects in those who voluntarily discontinued treatment suddenly. Analgesia was quickly reestablished upon Sativex resumption.

In a phase II RCT in 56 rheumatoid arthritis sufferers over 5 weeks with Sativex [137], medicine was limited to only 6 evening sprays (16.2 mg THC  +  15 mg CBD). By study end, morning pain on movement, morning pain at rest, DAS-28 measure of disease activity, and SF-MPQ pain all favored Sativex (Table 18.1).

In a phase III RCT in intractable cancer pain on opioids (N  =  177), Sativex, Tetranabinex THC-predominant extract, and placebo were compared [138] demonstrating strongly statistically significant improvements in analgesia for Sativex only (Table 18.1). This suggests that the CBD component in Sativex was necessary for benefit.

In a 2-week study of spinal cord injury pain, NRS of pain was not statistically different from placebo, probably due to the short duration of the trial, but secondary endpoints were positive (Table 18.1). Additionally, an RCT of intractable lower urinary tract symptoms in MS also demonstrated pain reduction (Table 18.1).

The open-label study of various polyneuropathy patients included Sativex patients with 3 obtaining 21.56 % mean pain relief after 3 months (2/3  >  30 %), and 4 achieving 27.6 % relief after 6 months (2/4  >  30 %), comparable to conventional agents [80].

A recently completed RCT of Sativex in intractable cancer pain unresponsive to opioids over 5 weeks was performed in 360 subjects (Table 18.1). Results of a Continuous Response Analysis (CRA) showed improvements over placebo in the low-dose (p  =  0.08) and middle-dose cohorts (p  =  0.038) or combined (p  =  0.006). Pain NRS improved over placebo in the low-dose (p  =  0.006) and combined cohorts (p  =  0.019).

Sleep has improved markedly in almost all Sativex RCTs in chronic pain based on symptom reduction, not a hypnotic effect [139].

The adverse event (AE) profile of Sativex has been quite benign with bad taste, oral stinging, dry mouth, dizziness, nausea, or fatigue most common, but not usually prompting discontinuation [128]. Most psychoactive sequelae are early and transient and have been notably lowered by more recent application of a slower, less aggressive titration schedule. While no direct comparative studies have been performed with Sativex and other agents, AE rates were comparable or greater with Marinol than with Sativex employing THC dosages some 2.5 times higher, likely due to the presence of accompanying CBD [28, 51]. Similarly, Sativex displayed a superior AE profile compared to smoked cannabis based on safety-extension studies of Sativex [28, 99], as compared to chronic use of cannabis with standardized government-supplied material in Canada for chronic pain [140] and the Netherlands for various indications [141, 142] over a period of several months or more. All AEs are more frequent with smoked cannabis, except for nausea and dizziness, both early and usually transiently reported with Sativex [27, 28, 128]. A recent meta-analysis suggested that serious AEs associated with cannabinoid-based medications did not differ from placebo and thus could not be attributable to cannabinoid use, further reinforcing the low toxicity associated with activation of cannabinoid systems.

Cannabinoid Pitfalls: Are They Surmountable?

The dangers of COX-1 and COX-2 inhibition by nonsteroidal anti-inflammatory drugs (NSAIDS) of various design (e.g., gastrointestinal ulceration and bleeding vs. coronary and cerebrovascular accidents, respectively) [143, 144] are unlikely to be mimicked by either THC or CBD, which produce no such activity at therapeutic dosages [49].

Natural cannabinoids require polar solvents and may be associated with delayed and sometimes erratic absorption after oral administration. Smoking of cannabis invariably produces rapid spikes in serum THC levels; cannabis smoking attains peak levels of serum THC above 140 ng/ml [145, 146], which, while desirable to the recreational user, has no necessity or advantage for treatment of chronic pain [28]. In contrast, comparable amounts of THC derived from oromucosal Sativex remained below 2 ng/ml with much lower propensity toward psychoactive sequelae [28, 125], with subjective intoxication levels on visual analogue scales that are indistinguishable from placebo, in the single digits out of 100 [100]. It is clear from RCTs that such psychoactivity is not a necessary accompaniment to pain control. In contrast, intoxication has continued to be prominent with oral THC [73].

In comparison to the questionable clinical trial blinding with smoked and vaporized cannabis discussed above, all indications are that such study blinding has been demonstrably effective with Sativex [147, 148] by utilizing a placebo spray with identical taste and color. Some 50 % of Sativex subjects in RCTs have had prior cannabis exposure, but results of two studies suggest that both groups exhibited comparable results in both treatment efficacy and side effect profile [134, 135].

Controversy continues to swirl around the issue of the potential dangers of cannabis use medicinally, particularly its drug abuse liability (DAL). Cannabis and cannabinoids are currently DEA schedule I substances and are forbidden in the USA (save for Marinol in schedule III and nabilone in schedule II) [73]. This is noteworthy in itself because the very same chemical compound, THC, appears simultaneously in schedule I (as THC), schedule II (as nabilone), and schedule III (as Marinol). DAL is assessed on the basis of five elements: intoxication, reinforcement, tolerance, withdrawal, and dependency plus the drug’s overall observed rates of abuse and diversion. Drugs that are smoked or injected are commonly rated as more reinforcing due to more rapid delivery to the brain [149]. Sativex has intermediate onset. It is claimed that CBD in Sativex reduces the psychoactivity of THC [28]. RCT AE profiles do not indicate euphoria or other possible reinforcing psychoactive indicia as common problems with its use [99]. Similarly, acute THC effects such as tachycardia, hypothermia, orthostatic hypotension, dry mouth, ocular injection, and intraocular pressure decreases undergo prominent tachyphylaxis with regular usage [150]. Despite that observation, Sativex has not demonstrated dose tolerance to its therapeutic benefits on prolonged administration, and efficacy has been maintained for up to several years in pain conditions [99].

The existence or severity of a cannabis withdrawal syndrome remains under debate [151, 152]. In contrast to reported withdrawal sequelae in recreational users [153], 24 subjects with MS who volunteered to discontinue Sativex after a year or more suffered no withdrawal symptoms meeting Budney criteria. While symptoms such as pain recurred after some 7–10 days without Sativex, symptom control was rapidly reattained upon resumption [99].

Finally, no known abuse or diversion incidents have been reported with Sativex to date (March 2011). Formal DAL studies of Sativex vs. Marinol and placebo have been completed and demonstrate lower scores on drug liking and similar measures at comparable doses [155].

Cognitive effects of cannabis also remain at issue [155, 156], but less data are available in therapeutic applications. Studies of Sativex in neuropathic pain with allodynia have revealed no changes vs. placebo on Sativex in portions of the Halstead-Reitan Battery [135], or in central neuropathic pain in MS [134], where 80 % of tests showed no significant differences. In a recent RCT of Sativex vs. placebo in MS patients, no cognitive differences of note were observed [157]. Similarly, chronic Sativex use has not produced observable mood disorders.

Controversies have also arisen regarding the possible association of cannabis abuse and onset of psychosis [156]. However, an etiological relationship is not supported by epidemiological data [158161], but may well be affected by dose levels and duration, if pertinent. One may speculate that lower serum levels of Sativex combined with antipsychotic properties of CBD [52, 162, 163] might attenuate such concerns. Few cases of related symptoms have been reported in SAFEX studies of Sativex.

Immune function becomes impaired in experimental animals at cannabinoid doses 50–100 times necessary to produce psychoactive effects [164]. In four patients smoking cannabis medicinally for more than 20 years, no changes were evident in leukocyte, CD4, or CD8 cell counts [155]. MS patients on Cannador demonstrated no immune changes of note [165] nor were changes evident in subjects smoking cannabis in a brief trial in HIV patients [166]. Sativex RCTs have demonstrated no hematological or immune dysfunction.

No effects of THC extract, CBD extract, or Sativex were evident on the hepatic cytochrome P450 complex [167] or on human CYP450 [168]. Similarly, while Sativex might be expected to have additive sedative effects with other drugs or alcohol, no significant drug-drug interactions of any type have been observed in the entire development program to date.

No studies have demonstrated significant problems in relation to cannabis affecting driving skills at plasma levels below 5 ng/ml of THC [169]. Four oromucosal sprays of Sativex (exceeding the average single dose employed in therapy) produced serum levels well below this threshold [28]. As with other cannabinoids in therapy, it is recommended that patients not drive nor use dangerous equipment until accustomed to the effects of the drug.

Future Directions: An Array of Biosynthetic and Phytocannabinoid Analgesics

Inhibition of Endocannabinoid Transport and Degradation: A Solution?

It is essential that any cannabinoid analgesic strike a compromise between therapeutic and adverse effects that may both be mediated via CB1 mechanisms [34]. Mechanisms to avoid psychoactive sequelae could include peripherally active synthetic cannabinoids that do not cross the blood-brain barrier or drugs that boost AEA levels by inhibiting fatty-acid amide hydrolase (FAAH) [170] or that of 2-AG by inhibiting monoacylycerol lipase (MGL). CBD also has this effect [50] and certainly seems to increase the therapeutic index of THC [51].

In preclinical studies, drugs inhibiting endocannabinoid hydrolysis [171, 172] and peripherally acting agonists [173] all show promise for suppressing neuropathic pain. AZ11713908, a peripherally restricted mixed cannabinoid agonist, reduces mechanical allodynia with efficacy comparable to the brain penetrant mixed cannabinoid agonist WIN55,212-2 [173]. An irreversible inhibitor of the 2-AG hydrolyzing enzyme MGL suppresses nerve injury-induced mechanical allodynia through a CB1 mechanism, although these anti-allodynic effects undergo tolerance following repeated administration [172]. URB937, a brain impermeant inhibitor of FAAH, has recently been shown to elevate anandamide outside the brain and suppress neuropathic and inflammatory pain behavior without producing tolerance or unwanted CNS side effects [171]. These observations raise the possibility that peripherally restricted endocannabinoid modulators may show therapeutic potential as analgesics with limited side-effect profiles.

The Phytocannabinoid and Terpenoid Pipeline

Additional phytocannabinoids show promise in treatment of chronic pain [123, 163, 174]. Cannabichromene (CBC), another prominent phytocannabinoid, also displays anti-inflammatory [175] and analgesic properties, though less potently than THC [176]. CBC, like CBD, is a weak inhibitor of AEA reuptake [177]. CBC is additionally a potent TRPA1 agonist [178]. Cannabigerol (CBG), another phytocannabinoid, displays weak binding at both CB1 and CB2 [179, 180] but is a more potent GABA reuptake inhibitor than either THC or CBD [181]. CBG is a stronger analgesic, anti-erythema, and lipooxygenase agent than THC [182]. CBG likewise inhibits AEA uptake and is a TRPV1 agonist [177], a TRPA1 agonist, and a TRPM8 antagonist [178]. CBG is also a phospholipase A2 modulator that reduces PGE-2 release in synovial cells [183]. Tetrahydrocannabivarin, a phytocannabinoid present in southern African strains, displays weak CB1 antagonism [184] and a variety of anticonvulsant activities [185] that might prove useful in chronic neuropathic pain treatment. THCV also reduced inflammation and attendant pain in mouse experiments [187]. Most North American [187] and European [188, 189] cannabis strains have been bred to favor THC over a virtual absence of other phytocannabinoid components, but the latter are currently available in abundance via selective breeding [124, 190].

Aromatic terpenoid components of cannabis also demonstrate pain reducing activity [123, 163]. Myrcene displays an opioid-type analgesic effect blocked by naloxone [191] and reduces inflammation via PGE-2 [192]. β-Caryophyllene displays anti-inflammatory activity on par with phenylbutazone via PGE-1 [193], but contrasts by displaying gastric cytoprotective activity [194]. Surprisingly, β-caryophyllene has proven to be a phytocannabinoid in its own right as a selective CB2 agonist [195]. α-Pinene inhibits PGE-1 [196], and linalool acts as a local anesthetic [197].

Summary

Basic science and clinical trials support the theoretical and practical basis of cannabinoid agents as analgesics for chronic pain. Their unique pharmacological profiles with multimodality effects and generally favorable efficacy and safety profiles render cannabinoid-based medicines promising agents for adjunctive treatment, particularly for neuropathic pain. It is our expectation that the coming years will mark the advent of numerous approved cannabinoids with varying mechanisms of action and delivery techniques that should offer the clinician useful new tools for treating pain.