ChapterPDF Available

Role of Cannabinoids in Pain Management

Authors:
  • CReDO Science

Abstract and Figures

It is a curious fact that we owe a great deal of our insight into pharmacological treatment of pain to the plant world. 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. 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.
Content may be subject to copyright.
181
T.R. Deer et al. (eds.), Comprehensive Treatment of Chronic Pain by Medical, Interventional, and Integrative Approaches,
DOI 10.1007/978-1-4614-1560-2_18, © American Academy of Pain Medicine 2013
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 in ammation. The opium poppy ( Papaver som-
niferum ) provided the prototypic narcotic analgesic morphine,
the rst 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 D 9 -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 phytochem-
istry of capsaicin, the pungent ingredient in hot chile peppers
( Capsicum annuum etc.), which activates transient recep-
tor 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 cata-
bolic enzymes, and their corresponding receptors [ 5 ] . AEA
is hydrolyzed by the enzyme fatty-acid amide hydrolase
(FAAH) into breakdown products arachidonic acid and etha-
nolamine [ 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
Role of Cannabinoids in Pain
Management
Ethan B. Russo and Andrea G. Hohmann
1 8
E. B. Russo , M.D. (*)
GW Pharmaceuticals ,
20402 81st Avenue SW , Vashon , WA 98070 , USA
Pharmaceutical Sciences , University of Montana ,
Missoula , MT , USA
e-mail: ethanrusso@comcast.net
A. G. Hohmann , Ph.D.
Department of Psychological and Brain Sciences , Indiana University ,
101 East 10th Street , Bloomington , IN 47405 , USA
e-mail: hohmanna@indiana.edu
Key Points
Cannabinoids are pharmacological agents of endog-
enous (endocannabinoids), botanical (phytocan-
nabinoids), or synthetic origin.
Cannabinoids alleviate pain through a variety of
receptor and non-receptor mechanisms including
direct analgesic and anti-in ammatory effects,
modulatory actions on neurotransmitters, and inter-
actions 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.
182 E.B. Russo and A.G. Hohmann
postsynaptic enzyme, may control anandamide levels near
sites of synthesis, whereas MGL, a presynaptic enzyme [ 8 ] ,
may terminate 2-AG signaling following CB
1 receptor acti-
vation. 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 forma-
tion of AEA remain poorly understood. However, 2-AG is
believed to be formed from membrane phospholipid precur-
sors through the sequential activation of two distinct enzymes,
phospholipase C and diacylglycerol lipase- a . First, PLC
catalyzes formation of the 2-AG precursor diacylglycerol
(DAG) from membrane phosphoinositides. Then, DAG is
hydrolyzed by the enzyme diacylglycerol lipase- a (DGL- a )
to generate 2-AG [
199 ] .
There are currently two well-de ned cannabinoid recep-
tors, although additional candidate cannabinoid receptors
have also been postulated. CB
1 , a seven transmembrane
spanning G-protein-coupled receptor inhibiting cyclic AMP
release, was identi ed in 1988 [ 10 ] . CB 1 is the primary neu-
romodulatory receptor accounting for psychopharmacologi-
cal effects of THC and most of its analgesic effects [ 11 ] .
Endocannabinoids are produced on demand in postsynaptic
cells and engage presynaptic CB
1 receptors through a retro-
grade mechanism [ 12 ] . Activation of presynaptic CB
1 recep-
tors then acts as a synaptic circuit breaker to inhibit
neurotransmitter release (either excitatory or inhibitory)
from the presynaptic neuron ( vide infra ) (Fig. 18.1 ). CB 2 was
identi ed in 1992, and while thought of primarily as a periph-
eral immunomodulatory receptor, it also has important
Fig. 18.1 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 gen-
erate 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- a to generate 2-AG. 2-AG is
released from the postsynaptic neuron and acts as a retrograde signal-
ing molecule. Endocannabinoids activate presynaptic CB
1 receptors
which reside on terminals of glutamatergic and GABAergic neurons.
Activation of CB
1 by 2-AG, anandamide, or exogenous cannabinoids
(e.g., tetrahydrocannabinol, THC ) inhibits calcium in ux in the presyn-
aptic 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 endo-
cannabinoid 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 CB
1 in the pre-
synaptic terminal, whereas FAAH is localized to postsynaptic sites.
The existence of an endocannabinoid transporter remains controver-
sial. 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 endocan-
nabinoid signaling system in the treatment of pain (Figure by authors
with kind assistance of James Brodie, GW Pharmaceuticals)
18318 Rol e of Cannabi noids in Pain Management
effects on pain. The role of CB
2 in modulating persistent
in ammatory and neuropathic pain [
13 ] has been recently
reviewed [ 14, 15 ] . Activation of CB 2 suppresses neuropathic
pain mechanisms through nonneuronal (i.e., microglia and
astrocytes) and neuronal mechanisms that may involve inter-
feron-gamma [ 16 ] . THC, the prototypical classical cannabi-
noid, is a weak partial agonist at both CB
1 and CB
2 receptors.
Transgenic mice lacking cannabinoid receptors (CB
1 , CB 2 ,
GPR55), enzymes controlling endocannabinoid breakdown
(FAAH, MGL, ABHD6), and endocannabinoid synthesis
(DGL- a , DGL- b ) have been generated [ 17 ] . These knock-
outs 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.
A Brief Scienti c History of Cannabis and Pain
Centuries of Citations
Cannabis has been utilized in one form or another for treat-
ment of pain for longer than written history [ 18– 21 ] .
Although this documentation has been a major preoccupa-
tion of the lead author [ 22– 25 ] , 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 ef cacy of canna-
bis in pain treatment certainly underline the importance of
properly harnessing cannabinoid mechanisms therapeuti-
cally [ 26, 27 ] , prescription analgesics in the United States
necessitate Food and Drug Administration (FDA) approval.
This requires a rigorous development program proving con-
sistency, quality, ef cacy, and safety as de ned by basic
scienti c studies and randomized controlled trials (RCT)
[ 28 ] and generally adhering to recent IMMPACT recommen-
dations [ 29 ] , provoking our next question.
Can a Botanical Agent Become a Prescription
Medicine?
Most modern physicians fail to recognize that pharmacog-
nosy (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 cer-
tainly possible to standardize botanical agents and facilitate
their recommendation based on sound science [
31 ] . Botanical
medicines can even ful ll 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 dem-
onstrate the same quality, clinical analgesic bene t, and
appropriately safe adverse event pro le 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 pub-
lished [ 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 sys-
tem 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 sup-
press 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 CB
1 [ 43 ] and CB 2 mechanisms (Fig. 18.2 ). Indeed, path-
ological pain states have been postulated to arise, at least in
part, from a dysregulation of the endocannabinoid system.
Antinociceptive and Anti-in fl ammatory Pain
Mechanisms
Beyond the mechanisms previously mentioned, the ECS
plays a critical role in peripheral pain, in ammation, and
hyperalgesia [ 43 ] through both CB 1 and CB
2 mechanisms.
CB
1 and CB
2 mechanisms are also implicated in regulation
of contact dermatitis and pruritus [ 44 ] . A role for spinal CB
2
mechanisms, mediated by microglia and/or astrocytes, is
also revealed under conditions of in ammation [
45 ] . Both
THC and cannabidiol (CBD), a non-euphoriant phytocan-
nabinoid common in certain cannabis strains, are potent anti-
in ammatory antioxidants with activity exceeding that of
184 E.B. Russo and A.G. Hohmann
Central nervous system
Descending modulation:
PAG (presynaptic GABAergic,
glutamatergic mechanisms
RVM (ON and OFF cells)
Nociceptive transmission
(VPL, amygdala, anterior
cingulate cortex)
Enteric nervous
system
Gut
Modulation
of propulation
Periphery
Nonneuronal:
CB1,CB2(immune cells,
inflammatory cells,
keratinocytes)
Pruritus
Limbs
Autonomic
Nerve
Nerve
Vagus
Brain
Cord
Wind-up
Central
sensitization
Nociceptive
transmission
WDR, NS cell;
microglia ( by injury)
Contact dermatitisHyperalgesia
Allodynia
Primary afferent:
CB1;CB2( by injury)
Modulation
of secretion
Fig. 18.2 Cannabinoids suppress pain and other pathophysiological
(e.g., contact dermatitis, pruritis) and physiological (e.g., gastrointesti-
nal transit and secretion) processes through multiple mechanisms
involving CB
1 and CB
2 receptors. Peripheral, spinal, and supraspinal
sites of cannabinoid actions are shown. In the periphery, cannabinoids
act through both neuronal and nonneuronal mechanisms to control
in ammation, allodynia, and hyperalgesia. CB
1 and CB
2 have been
localized to both primary afferents and nonneuronal cells (e.g., kerati-
nocytes, 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 speci c ( NS ) cells. Similar pro-
cesses are observed at rostral levels of the neuraxis (e.g., ventropostero-
lateral nucleus of the thalamus, amygdala, anterior cingulate cortex).
Cannabinoids also actively modulate pain through descending mecha-
nisms. 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 ring pause of OFF cells, in response to noxious
stimulation to produce antinociception (Figure by authors with kind
assistance of James Brodie, GW Pharmaceuticals)
18518 Rol e of Cannabi noids in Pain Management
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 TRPV
1 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 vari-
ous pharmaceutical rms 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 attenu-
ating 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- a ) 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
ef cacious 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 impor-
tant in NMDA-glutamatergic mechanisms that become
hyperresponsive in chronic pain states. Cannabinoids
speci cally 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,
bromyalgia, and idiopathic bowel syndrome wherein these
mechanisms seem to operate pathophysiologically [ 63 ] ,
prompting the hypothesis of a “clinical endocannabinoid
de ciency.” Endocannabinoid modulators may therefore
restore homeostasis, leading to normalization of function in
these pathophysiological conditions. THC also has numer-
ous effects on serotonergic systems germane to migraine
[ 64 ] , increasing its production in the cerebrum while decreas-
ing reuptake [
65 ] . In fact, the ECS seems to modulate the
trigeminovascular system of migraine pathogenesis at
vascular and neurochemical levels [
66– 68 ] .
Cannabinoid-Opioid Interactions
Although endocannabinoids do not bind to opioid receptors,
the ECS may nonetheless work in parallel with the endoge-
nous 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 demon-
strate experimental opiate sparing [ 70 ] , prevent opioid toler-
ance and withdrawal [ 71 ] , and rekindle opioid analgesia after
loss of effect [ 72 ] . Adjunctive treatments that combine opi-
oids with cannabinoids may enhance the analgesic effects of
either agent. Such strategies may permit lower doses of anal-
gesics to be employed for therapeutic bene t 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 dif culties, 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 signi fi cant bene fi t was
evident and side effects led to prominent dropout rates (aver-
age doses 15–16.6 mg THC). Dronabinol produced bene t in
pain in multiple sclerosis [ 76 ] , but none was evident in post-
operative 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 ] , fol-
lowed by a 4-week open-label trial with continued improve-
ment (Table 18.1 ). Associated adverse events were prominent.
Methodological issues included lack of prescreening for can-
nabinoids, 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 nabi-
lone patients with 6 obtaining 22.6 % mean pain relief after
3 months, and 5 achieving 28.6 % relief after 6 months, com-
parable 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
ef cacy than with diphenhydramine [ 81 ] .
186 E.B. Russo and A.G. Hohmann
Table 18.1 Randomized controlled trials of cannabinoids in pain
Agent N = Indication Duration/type Outcomes/reference
Ajulemic acid 21 Neuropathic pain 7 day crossover Visual analogue pain scales improved
over placebo ( p = 0.02)/Karst et al. [
92 ]
Cannabis, smoked 50 HIV neuropathy 5 days/DB Decreased daily pain ( p = 0.03) and
hyperalgesia ( p = 0.05), 52 % with >30 %
pain reduction vs. placebo ( p = 0.04)/
Abrams et al. [
94 ]
Cannabis, smoked 23 Chronic neuropathic pain 5 days/DB Decreased pain vs. placebo only at 9.4 %
THC level ( p = 0.023)/Ware et al. [
98 ]
Cannabis, smoked 38 Neuropathic pain Single dose/DBC NSD in pain except at highest cannabis
dose ( p = 0.02), with prominent
psychoactive effects/Wilsey et al. [
95 ]
Cannabis, smoked 34 HIV neuropathy 5 days /DB DDS improved over placebo ( p = 0.016),
46 % vs. 18 % improved >30 %, 2 cases
toxic psychosis/Ellis et al. [
97 ]
Cannabis, vaporized 21 Chronic pain on opioids 5 days/DB 27 % decrement in pain/Abrams et al.
[ 118 ]
Cannador 419 Pain due to spasm in MS 15 weeks Improvement over placebo in subjective
pain associated with spasm ( p = 0.003)/
Zajicek et al. [
120 ]
Cannador 65 Postherpetic neuralgia 4 weeks No bene fi t observed/Ernst et al. [ 122 ]
Cannador 30 Postoperative pain Single doses, daily Decreasing pain intensity with increased
dose ( p = 0.01)/Holdcroft et al. [
123 ]
Marinol 24 Neuropathic pain in MS 15–21 days/DBC Median numerical pain ( p = 0.02),
median pain relief improved ( p = 0.035)
over placebo/Svendsen et al. [
76 ]
Marinol 40 Postoperative pain Single dose/DB No bene fi t observed over placebo/Buggy
et al. [
77 ]
Marinol 30 Chronic pain 3 doses, 1 day/DBC Total pain relief improved with 10 mg
( p < 0.05) and 20 mg ( p < 0.01) with
opioids, AE prominent/Narang et al. [
79 ]
Nabilone 41 Postoperative pain 3 doses in 24 h/DB NSD morphine consumption. Increased
pain at rest and on movement with
nabilone 1 or 2 mg/Beaulieu [
85 ]
Nabilone 31 Fibromyalgia 2 weeks/DBC Compared to amitriptyline, nabilone
improved sleep, decrease wakefulness,
had no effect on pain, and increased AE/
Ware et al. [
90 ]
Nabilone 96 Neuropathic pain 14 weeks/DBC vs.
dihydrocodeine
Dihydrocodeine more effective with
fewer AE/Frank et al. [
88 ]
Nabilone 13 Spasticity pain 9 weeks/DBC NRS decreased 2 points for nabilone
( p < 0.05)/Wissel et al. [
87 ]
Nabilone 40 Fibromyalgia 4 weeks/DBC VAS decreased in pain, Fibromyalgia
Impact Questionnaire, and anxiety over
placebo (all, p < 0.02)/Skrabek et al. [
89 ]
Sativex 20 Neurogenic pain Series of 2-week N-of-1
crossover blocks
Improvement with Tetranabinex and
Sativex on VAS pain vs. placebo
( p < 0.05), symptom control best with
Sativex ( p < 0.0001)/Wade et al. [
132 ]
Sativex 24 Chronic intractable pain 12 weeks, series of N-of-1
crossover blocks
VAS pain improved over placebo
( p < 0.001) especially in MS ( p < 0.0042)/
Notcutt et al. [
133 ]
Sativex 48 Brachial plexus avulsion 6 weeks in 3 two-week
crossover blocks
Bene ts noted in Box Scale-11 pain
scores with Tetranabinex ( p = 0.002) and
Sativex ( p = 0.005) over placebo/Berman
et al. [
134 ]
Sativex 66 Central neuropathic pain
in MS
5 weeks Numerical Rating Scale (NRS) analgesia
improved over placebo ( p = 0.009)/Rog
et al. [
135 ]
(continued)
18718 Rol e of Cannabino ids in Pain Management
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 dys-
phoria 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 uncon-
trolled 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 bene ts in other parame-
ters. 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 bene t,
but with dihydrocodeine proving clearly superior in ef cacy
and modestly superior in side-effect pro le. In an RCT in 40
patients of nabilone vs. placebo over 4 weeks, it showed
signi cant decreases in VAS of pain and anxiety (Table
18.1 )
[ 89 ] . A more recent study of nabilone vs. amitriptyline in
bromyalgia yielded bene ts 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 psychoac-
tive 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 [ 94– 97 ] .
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 experi-
enced. Symptoms of possible intoxication in the cannabis
group including anxiety (25 %), sedation (54 %), disorienta-
tion (16 %), paranoia (13 %), confusion (17 %), dizziness
(15 %), and nausea (11 %) were all statistically signi cantly
more common than in the placebo group. Despite these
ndings, the authors stated that the values do not represent
any serious safety concern in this short-term study. No dis-
cussion in the article addressed issues of the relative ef cacy
of blinding in the trial.
Wilsey et al. [ 95 ] examined neuropathic pain in 38 sub-
jects 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
Table 18.1 (continued)
Agent N = Indication Duration/type Outcomes/reference
Sativex 125 Peripheral neuropathic
pain
5 weeks Improvements in NRS pain levels
( p = 0.004), dynamic allodynia ( p = 0.042),
and punctuate allodynia ( p = 0.021) vs.
placebo/Nurmikko et al. [
136 ]
Sativex 56 Rheumatoid arthritis Nocturnal dosing for 5
weeks
Improvements over placebo morning
pain on movement ( p = 0.044), morning
pain at rest ( p = 0.018), DAS-28
( p = 0.002), and SF-MPQ pain at present
( p = 0.016)/Blake et al. [
138 ]
Sativex 117 Pain after spinal injury 10 days NSD in NRS pain scores, but improved
Brief Pain Inventory ( p = 0.032), and
Patients’ Global Impression of Change
( p = 0.001) (unpublished)
Sativex 177 Intractable cancer pain 2 weeks Improvements in NRS analgesia vs.
placebo ( p = 0.0142), Tetranabinex NSD/
Johnson et al. [
139 ]
Sativex 135 Intractable lower urinary
tract symptoms in MS
8 weeks Improved bladder severity symptoms
including pain over placebo ( p = 0.001)
[ 200 ]
Sativex 360 Intractable cancer pain 5 weeks/DB CRA of lower and middle-dose cohorts
improved over placebo ( p = 0.006)/ [ 201 ]
188 E.B. Russo and A.G. Hohmann
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, particu-
larly 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 signi cant
differences were noted in pain relief in the two cannabis
potency groups, but a signi cant 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 ana-
logue 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 signi cantly 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 promi-
nent feature in this cannabis-experienced population. This
study distinguished itself from some others in its inclusion of
speci c objective neuropsychological measures and demon-
strated neurocognitive impairment in attention, learning, and
memory, most noteworthy with 7 % THC cannabis. No com-
mentary on blinding ef cacy was included.
Ellis et al. [ 97 ] examined HIV-associated neuropathic
pain in a double-blind trial of placebo vs. 1–8 % THC can-
nabis 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-experi-
enced, 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 sub-
jects 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 psycho-
sis” at what proved to be his rst 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 inac-
curate at guessing the investigational product, 93 % of those
receiving cannabis guessed correctly. On safety issues, the
authors stated that the frequency of some nontreatment-lim-
iting side effects was greater for cannabis than placebo.
These included concentration dif culties, fatigue, sleepiness
or sedation, increased duration of sleep, reduced salivation,
and thirst.
A Canadian study [ 98 ] examined single 25-mg inhala-
tions 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 can-
nabis-free for 1 year, but were required to have some experi-
ence 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, numb-
ness, 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 ] gen-
erally suggest randomized controlled clinical trials of
12-week duration as a prerequisite to demonstrate ef cacy
and safety. While one might assume that the degree of pain
improvement demonstrated in these trials could be main-
tained 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, prescrip-
tion, and named-patient supplies, with vastly lower AE rates
(data on le, GW Pharmaceuticals) [ 28, 99 ] . Certainly, the
cognitive effects noted in California-smoked cannabis stud-
ies gure among many factors that would call the ef cacy 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 speci c chronic polyneuropathy
syndromes and their response to pharmacological therapies,
the presence of intolerable side effects did not differ in groups
receiving gabapentinoids, tricyclic antidepressants, anticon-
vulsants, 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 bene ts might
possibly accrue to some, but it is eminently likely that candi-
dates for such therapy might refuse it on any number of
18918 Role of Cannabinoids in Pain Management
grounds: not wishing to smoke, concern with respect to intox-
ication, 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 bron-
chitic symptoms [ 100 ] , if not obvious emphysematous degen-
eration [ 101 ] or increase in aerodigestive cancers [ 102 ] . Even
such smoked cannabis proponents as Lester Grinspoon has
acknowledged are the only well-con rmed deleterious physi-
cal 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 respira-
tory 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 con-
tained ammonia (NH
3 ) at a level of 720 m g per 775 mg ciga-
rette, a gure 20-fold higher than that found in tobacco
smoke. It was hypothesized that this nding was likely attrib-
utable 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 compari-
sons, 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 carci-
nogenic potential of these various compounds were men-
tioned. More recently, experimental analysis of cannabis
smoke with resultant acetaldehyde production has posited its
genotoxic potential to be attributable to reactions that pro-
duce DNA adducts [ 105 ] .
Vaporizers for cannabis have been offered as a harm reduc-
tion 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 com-
ponents with the goal of reducing or eliminating by-products
of combustion, including potentially carcinogenic polycyclic
aromatic hydrocarbons, benzene, acetaldehyde, carbon mon-
oxide, toluene, naphthaline, phenol, toluene, hydrogen cya-
nide, and ammonia. Space limitations permit only a cursory
review of available literature [ 107– 115 ] .
A pilot study of the Volcano vaporizer vs. smoking was
performed in the USA in 2007 in 18 active cannabis consum-
ers, 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 identi-
cal 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 under-
taken, the assertion is questionable. It was also stated that
there were no reported adverse events. Some 12 subjects pre-
ferred the Volcano, 2 chose smoking, and 2 had no prefer-
ence 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 lim-
ited 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 pro le of smoked or vaporized cannabis would
meet FDA requirements. The fact that all the vaporization tri-
als 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 pro les 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 signi cant amounts. Existing vaporizers still
lack portability or convenience [ 28 ] . A large Internet survey
revealed that only 2.2 % of cannabis users employed vapor-
ization as their primary cannabis intake method [ 118 ] . While
studies to date have established that lower temperature vapor-
ization in the Volcano, but not necessarily other devices, can
reduce the relative amounts of noxious by-products of com-
bustion, it has yet to be demonstrated that they are totally
eliminated. Until or unless this goal is achieved, along with
190 E.B. Russo and A.G. Hohmann
requisite benchmarks of herbal cannabis quality, safety, and
ef cacy 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 differ-
ing 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-associ-
ated pain. Both THC and Cannador improved pain scores in
follow-up [ 120 ] . Cannador was also employed for posther-
petic 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 nota-
ble (Table
18.1 ).
Sativex® is a whole-cannabis-based extract delivered as
an oromucosal spray that combines a CB
1 and CB
2 partial
agonist (THC) with a cannabinoid system modulator (CBD),
minor cannabinoids, and terpenoids plus ethanol and propyl-
ene glycol excipients and peppermint avoring [ 51, 123 ] .
It is approved in Canada for spasticity in MS and under a
Notice of Compliance with Conditions for central neuro-
pathic 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 pre-
dominating in THC and CBD, respectively [ 124 ] . Each
100 m l 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 [ 125– 127 ] . Sativex
effects begin within an interval allowing dose titration.
A very favorable adverse event pro le has been observed in
the development program [ 27, 128 ] . Most patients stabilize
at 8–10 sprays per day after 7–10 days, attaining symptom-
atic 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 com-
pleted [ 201 ] . Available clinical trials with Sativex have been
independently assessed [
129, 130 ] .
In a phase II study of 20 patients with neurogenic symp-
toms [ 131 ] , signi cant 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 com-
pared 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 symp-
toms [ 99 ] without development of any tolerance requiring
dose escalation or withdrawal effects in those who volun-
tarily 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 signi cant improvements in analgesia for Sativex
only (Table 18.1 ). This suggests that the CBD component in
Sativex was necessary for bene t.
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 con-
ventional agents [ 80 ] .
A recently completed RCT of Sativex in intractable can-
cer 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 pla-
cebo 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).
19118 Rol e of Cannabino ids in Pain Management
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) pro le of Sativex has been quite
benign with bad taste, oral stinging, dry mouth, dizziness, nau-
sea, or fatigue most common, but not usually prompting dis-
continuation [ 128 ] . Most psychoactive sequelae are early and
transient and have been notably lowered by more recent appli-
cation 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 pro le
compared to smoked cannabis based on safety-extension stud-
ies 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 indica-
tions [ 141, 142 ] over a period of several months or more. All
AEs are more frequent with smoked cannabis, except for nau-
sea 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 medica-
tions did not differ from placebo and thus could not be attribut-
able 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 nonsteroi-
dal anti-in ammatory 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 pro-
duce 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 pro-
duces 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 neces-
sity or advantage for treatment of chronic pain [ 28 ] . In con-
trast, 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 indistin-
guishable from placebo, in the single digits out of 100 [ 100 ] .
It is clear from RCTs that such psychoactivity is not a neces-
sary 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 demonstra-
bly 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 ef cacy and side effect
pro fi le [ 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 simultane-
ously in schedule I (as THC), schedule II (as nabilone), and
schedule III (as Marinol). DAL is assessed on the basis of
ve elements: intoxication, reinforcement, tolerance, with-
drawal, 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 psycho-
activity of THC [ 28 ] . RCT AE pro les do not indicate eupho-
ria or other possible reinforcing psychoactive indicia as
common problems with its use [ 99 ] . Similarly, acute THC
effects such as tachycardia, hypothermia, orthostatic hypoten-
sion, dry mouth, ocular injection, and intraocular pressure
decreases undergo prominent tachyphylaxis with regular
usage [ 150 ] . Despite that observation, Sativex has not dem-
onstrated dose tolerance to its therapeutic bene ts on pro-
longed administration, and ef cacy has been maintained for
up to several years in pain conditions [ 99 ] .
The existence or severity of a cannabis withdrawal syn-
drome 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 meet-
ing 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 com-
pleted and demonstrate lower scores on drug liking and simi-
lar 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 signi cant dif-
ferences. In a recent RCT of Sativex vs. placebo in MS
patients, no cognitive differences of note were observed
192 E.B. Russo and A.G. Hohmann
[ 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 epi-
demiological data [ 158– 161 ] , 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 con-
cerns. Few cases of related symptoms have been reported in
SAFEX studies of Sativex.
Immune function becomes impaired in experimental ani-
mals 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 demon-
strated 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 signi cant drug-drug interactions of any type have
been observed in the entire development program to date.
No studies have demonstrated signi cant 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 ther-
apy) 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 compro-
mise between therapeutic and adverse effects that may both be
mediated via CB
1 mechanisms [ 34 ] . Mechanisms to avoid
psychoactive sequelae could include peripherally active syn-
thetic 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 monoa-
cylycerol lipase (MGL). CBD also has this effect [
50 ] and cer-
tainly 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 ef cacy 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 CB
1 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 sup-
press neuropathic and in ammatory 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 pro les.
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-
in fl ammatory [ 175 ] and analgesic properties, though less
potently than THC [ 176 ] . CBC, like CBD, is a weak inhibi-
tor of AEA reuptake [ 177 ] . CBC is additionally a potent
TRPA1 agonist [ 178 ] . Cannabigerol (CBG), another phyto-
cannabinoid, displays weak binding at both CB
1 and CB
2
[ 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, dis-
plays weak CB
1 antagonism [ 184 ] and a variety of anticon-
vulsant activities [ 185 ] that might prove useful in chronic
neuropathic pain treatment. THCV also reduced in ammation
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 demon-
strate pain reducing activity [ 123, 163 ] . Myrcene displays an
opioid-type analgesic effect blocked by naloxone [ 191 ] and
reduces in ammation via PGE-2 [ 192 ] . b -Caryophyllene
displays anti-in ammatory activity on par with phenylbuta-
zone via PGE-1 [ 193 ] , but contrasts by displaying gastric
cytoprotective activity [
194 ] . Surprisingly, b -caryophyllene
has proven to be a phytocannabinoid in its own right as a
selective CB
2 agonist [ 195 ] . a -Pinene inhibits PGE-1 [ 196 ] ,
and linalool acts as a local anesthetic [
197 ] .
19318 Role of Cannabinoids in Pain Management
Summary
Basic science and clinical trials support the theoretical and
practical basis of cannabinoid agents as analgesics for
chronic pain. Their unique pharmacological pro les with
multimodality effects and generally favorable ef cacy and
safety pro les render cannabinoid-based medicines promis-
ing agents for adjunctive treatment, particularly for neuro-
pathic 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.
References
1. Di Marzo V, Bisogno T, De Petrocellis L. Endocannabinoids and
related compounds: walking back and forth between plant natural
products and animal physiology. Chem Biol. 2007;14(7):741–56.
2. Gaoni Y, Mechoulam R. Isolation, structure and partial synthesis of
an active constituent of hashish. J Am Chem Soc. 1964;86:
1646–7.
3. Pate D. Chemical ecology of cannabis. J Int Hemp Assoc. 1994;2:
32–7.
4. Di Marzo V, Melck D, Bisogno T, De Petrocellis L.
Endocannabinoids: endogenous cannabinoid receptor ligands with
neuromodulatory action. Trends Neurosci. 1998;21(12):521–8.
5. Pacher P, Batkai S, Kunos G. The endocannabinoid system as an
emerging target of pharmacotherapy. Pharmacol Rev. 2006;58(3):
389–462.
6. Cravatt BF, Giang DK, May eld SP, Boger DL, Lerner RA, Gilula
NB. Molecular characterization of an enzyme that degrades neuro-
modulatory fatty-acid amides. Nature. 1996;384(6604):83–7.
7. Dinh TP, Freund TF, Piomelli D. A role for monoglyceride lipase in
2-arachidonoylglycerol inactivation. Chem Phys Lipids.
2002;121(1–2):149–58.
8. Gulyas AI, Cravatt BF, Bracey MH, et al. Segregation of two endo-
cannabinoid-hydrolyzing enzymes into pre- and postsynaptic com-
partments in the rat hippocampus, cerebellum and amygdala. Eur J
Neurosci. 2004;20(2):441–58.
9. Mangieri RA, Piomelli D. Enhancement of endocannabinoid sig-
naling and the pharmacotherapy of depression. Pharmacol Res.
2007;56(5):360–6.
10. Howlett AC, Johnson MR, Melvin LS, Milne GM. Nonclassical
cannabinoid analgetics inhibit adenylate cyclase: development of a
cannabinoid receptor model. Mol Pharmacol. 1988;33(3):
297–302.
11. Zimmer A, Zimmer AM, Hohmann AG, Herkenham M, Bonner TI.
Increased mortality, hypoactivity, and hypoalgesia in cannabinoid CB1
receptor knockout mice. Proc Natl Acad Sci USA. 1999;96(10):5780–5.
12. Wilson RI, Nicoll RA. Endogenous cannabinoids mediate retro-
grade signalling at hippocampal synapses. Nature. 2001;410(6828):
588–92.
13. Ibrahim MM, Porreca F, Lai J, et al. CB2 cannabinoid receptor acti-
vation produces antinociception by stimulating peripheral release
of endogenous opioids. Proc Natl Acad Sci USA. 2005;102(8):
3093–8.
14. Guindon J, Hohmann AG. Cannabinoid CB2 receptors: a therapeu-
tic target for the treatment of in ammatory and neuropathic pain.
Br J Pharmacol. 2008;153(2):319–34.
15. Pacher P, Mechoulam R. Is lipid signaling through cannabinoid 2 recep-
tors part of a protective system? Prog Lipid Res. 2011;50:193–211.
16. Racz I, Nadal X, Alferink J, et al. Interferon-gamma is a critical
modulator of CB(2) cannabinoid receptor signaling during neuro-
pathic pain. J Neurosci. 2008;28(46):12136–45.
17. Guindon J, Hohmann AG. The endocannabinoid system and pain.
CNS Neurol Disord Drug Targets. 2009;8(6):403–21.
18. Fankhauser M. History of cannabis in Western medicine. In:
Grotenhermen F, Russo EB, editors. Cannabis and cannabinoids:
pharmacology, toxicology and therapeutic potential. Binghamton:
Haworth Press; 2002. p. 37–51.
19. Russo EB. History of cannabis as medicine. In: Guy GW, Whittle
BA, Robson P, editors. Medicinal uses of cannabis and cannabi-
noids. London: Pharmaceutical Press; 2004. p. 1–16.
20. Russo EB. History of cannabis and its preparations in saga, science
and sobriquet. Chem Biodivers. 2007;4(8):2624–48.
21. Mechoulam R. The pharmacohistory of Cannabis sativa . In:
Mechoulam R, editor. Cannabinoids as therapeutic agents. Boca
Raton: CRC Press; 1986. p. 1–19.
22. Russo E. Cannabis treatments in obstetrics and gynecology: a his-
torical review. J Cannabis Ther. 2002;2(3–4):5–35.
23. Russo EB. Hemp for headache: an in-depth historical and scienti c
review of cannabis in migraine treatment. J Cannabis Ther.
2001;1(2):21–92.
24. Russo EB. The role of cannabis and cannabinoids in pain manage-
ment. In: Cole BE, Boswell M, editors. Weiner’s pain management:
a practical guide for clinicians. 7th ed. Boca Raton: CRC Press;
2006. p. 823–44.
25. Russo EB. Cannabis in India: ancient lore and modern medicine.
In: Mechoulam R, editor. Cannabinoids as therapeutics. Basel:
Birkhäuser Verlag; 2005. p. 1–22.
26. ABC News, USA Today, Stanford Medical Center Poll. Broad
experience with pain sparks search for relief. 9 May 2005.
27. Russo EB. Cannabinoids in the management of dif fi cult to treat
pain. Ther Clin Risk Manag. 2008;4(1):245–59.
28. Russo EB. The solution to the medicinal cannabis problem. In:
Schatman ME, editor. Ethical issues in chronic pain management.
Boca Raton: Taylor & Francis; 2006. p. 165–94.
29. Dworkin RH, Turk DC, Farrar JT, et al. Core outcome measures for
chronic pain clinical trials: IMMPACT recommendations. Pain.
2005;113(1–2):9–19.
30. Tyler VE. Phytomedicines in Western Europe: potential impact on
herbal medicine in the United States. In: Kinghorn AD, Balandrin MF,
editors. Human medicinal agents from plants (ACS symposium, No.
534). Washington, D.C.: American Chemical Society; 1993. p. 25–37.
31. Russo EB. Handbook of psychotropic herbs: a scienti fi c analysis of
herbal remedies for psychiatric conditions. Binghamton: Haworth
Press; 2001.
32. Food and Drug Administration. Guidance for industry: botanical
drug products. In: Services UDoHaH, editor. US Government; 2004.
p. 48.
http://www.fda.gov/downloads/Drugs/GuidanceCompliance
RegulatoryInformation/Guidances/ucm070491.pdf.
33. Walker JM, Hohmann AG. Cannabinoid mechanisms of pain sup-
pression. Handb Exp Pharmacol. 2005;168:509–54.
34. Rahn EJ, Hohmann AG. Cannabinoids as pharmacotherapies for
neuropathic pain: from the bench to the bedside. Neurotherapeutics.
2009;6(4):713–37.
35. Richardson JD, Aanonsen L, Hargreaves KM. SR 141716A, a can-
nabinoid receptor antagonist, produces hyperalgesia in untreated
mice. Eur J Pharmacol. 1997;319(2–3):R3–4.
36. Walker JM, Huang SM, Strangman NM, Tsou K, Sanudo-Pena
MC. Pain modulation by the release of the endogenous cannabi-
noid anandamide. Proc Natl Acad Sci. 1999;96(21):12198–203.
37. Walker JM, Hohmann AG, Martin WJ, Strangman NM, Huang SM,
Tsou K. The neurobiology of cannabinoid analgesia. Life Sci.
1999;65(6–7):665–73.
38. Martin WJ, Hohmann AG, Walker JM. Suppression of noxious
stimulus-evoked activity in the ventral posterolateral nucleus of the
194 E.B. Russo and A.G. Hohmann
thalamus by a cannabinoid agonist: correlation between electro-
physiological and antinociceptive effects. J Neurosci. 1996;16:
6601–11.
39. Hohmann AG, Martin WJ, Tsou K, Walker JM. Inhibition of nox-
ious stimulus-evoked activity of spinal cord dorsal horn neurons by
the cannabinoid WIN 55,212-2. Life Sci. 1995;56(23–24):2111–8.
40. Richardson JD, Aanonsen L, Hargreaves KM. Antihyperalgesic effects
of spinal cannabinoids. Eur J Pharmacol. 1998;345(2):145–53.
41. Strangman NM, Walker JM. Cannabinoid WIN 55,212-2 inhibits
the activity-dependent facilitation of spinal nociceptive responses.
J Neurophysiol. 1999;82(1):472–7.
42. Rahn EJ, Makriyannis A, Hohmann AG. Activation of cannabinoid
CB(1) and CB(2) receptors suppresses neuropathic nociception
evoked by the chemotherapeutic agent vincristine in rats. Br J
Pharmacol. 2007;152:765–77.
43. Richardson JD, Kilo S, Hargreaves KM. Cannabinoids reduce
hyperalgesia and in ammation via interaction with peripheral CB1
receptors. Pain. 1998;75(1):111–9.
44. Karsak M, Gaffal E, Date R, et al. Attenuation of allergic contact
dermatitis through the endocannabinoid system. Science.
2007;316(5830):1494–7.
45. Luongo L, Palazzo E, Tambaro S, et al. 1-(2 ¢ ,4 ¢ -Dichlorophenyl)-6-
methyl-N-cyclohexylamine-1,4-dihydroindeno[1,2-c]pyrazole-3-
carboxamide, a novel CB2 agonist, alleviates neuropathic pain
through functional microglial changes in mice. Neurobiol Dis.
2010;37(1):177–85.
46. Hampson AJ, Grimaldi M, Axelrod J, Wink D. Cannabidiol and (-)
Delta9-tetrahydrocannabinol are neuroprotective antioxidants. Proc
Natl Acad Sci USA. 1998;95(14):8268–73.
47. Burstein S, Levin E, Varanelli C. Prostaglandins and cannabis. II.
Inhibition of biosynthesis by the naturally occurring cannabinoids.
Biochem Pharmacol. 1973;22(22):2905–10.
48. Fimiani C, Liberty T, Aquirre AJ, Amin I, Ali N, Stefano GB.
Opiate, cannabinoid, and eicosanoid signaling converges on com-
mon intracellular pathways nitric oxide coupling. Prostaglandins
Other Lipid Mediat. 1999;57(1):23–34.
49. Stott CG, Guy GW, Wright S, Whittle BA. The effects of cannabis
extracts Tetranabinex & Nabidiolex on human cyclo-oxygenase
(COX) activity. Paper presented at: Symposium on the Cannabinoids,
Clearwater, June 2005.
50. Bisogno T, Hanus L, De Petrocellis L, et al. Molecular targets for
cannabidiol and its synthetic analogues: effect on vanilloid VR1
receptors and on the cellular uptake and enzymatic hydrolysis of
anandamide. Br J Pharmacol. 2001;134(4):845–52.
51. Russo EB, Guy GW. A tale of two cannabinoids: the therapeutic
rationale for combining tetrahydrocannabinol and cannabidiol.
Med Hypotheses. 2006;66(2):234–46.
52. Morgan CJ, Curran HV. Effects of cannabidiol on schizophrenia-
like symptoms in people who use cannabis. Br J Psychiatry.
2008;192(4):306–7.
53. Morgan CJ, Freeman TP, Schafer GL, Curran HV. Cannabidiol
attenuates the appetitive effects of delta 9-tetrahydrocannabinol in
humans smoking their chosen cannabis. Neuropsychopharmacology.
2010;35(9):1879–85.
54. Morgan CJ, Schafer G, Freeman TP, Curran HV. Impact of canna-
bidiol on the acute memory and psychotomimetic effects of smoked
cannabis: naturalistic study. Br J Psychiatry. 2010;197(4):285–90.
55. Malfait AM, Gallily R, Sumariwalla PF, et al. The nonpsychoactive
cannabis constituent cannabidiol is an oral anti-arthritic therapeutic
in murine collagen-induced arthritis. Proc Natl Acad Sci USA.
2000;97(17):9561–6.
56. Carrier EJ, Auchampach JA, Hillard CJ. Inhibition of an equilibra-
tive nucleoside transporter by cannabidiol: a mechanism of can-
nabinoid immunosuppression. Proc Natl Acad Sci USA.
2006;103(20):7895–900.
57. McHugh D, Hu SS, Rimmerman N, et al. N-arachidonoyl glycine,
an abundant endogenous lipid, potently drives directed cellular
migration through GPR18, the putative abnormal cannabidiol
receptor. BMC Neurosci. 2010;11:44.
58. Dmitrieva N, Nagabukuro H, Resuehr D, et al. Endocannabinoid
involvement in endometriosis. Pain. 2010;151(3):703–10.
59. Izzo AA, Camilleri M. Emerging role of cannabinoids in gastroin-
testinal and liver diseases: basic and clinical aspects. Gut.
2008;57(8):1140–55.
60. Izzo AA, Sharkey KA. Cannabinoids and the gut: new developments
and emerging concepts. Pharmacol Ther. 2010;126(1):21–38.
61. Shen M, Piser TM, Seybold VS, Thayer SA. Cannabinoid receptor
agonists inhibit glutamatergic synaptic transmission in rat hip-
pocampal cultures. J Neurosci. 1996;16(14):4322–34.
62. Nicolodi M, Volpe AR, Sicuteri F. Fibromyalgia and headache.
Failure of serotonergic analgesia and N-methyl-D-aspartate-
mediated neuronal plasticity: their common clues. Cephalalgia.
1998;18 Suppl 21:41–4.
63. Russo EB. Clinical endocannabinoid de fi ciency (CECD): Can this
concept explain therapeutic bene ts of cannabis in migraine,
bromyalgia, irritable bowel syndrome and other treatment-resis-
tant conditions? Neuroendocrinol Lett. 2004;25(1–2):31–9.
64. Russo E. Cannabis for migraine treatment: the once and future pre-
scription? An historical and scienti c review. Pain. 1998;76(1–2):3–8.
65. Spadone C. Neurophysiologie du cannabis [neurophysiology of
cannabis]. Encéphale. 1991;17(1):17–22.
66. Akerman S, Holland PR, Goadsby PJ. Cannabinoid (CB1) receptor
activation inhibits trigeminovascular neurons. J Pharmacol Exp
Ther. 2007;320(1):64–71.
67. Akerman S, Kaube H, Goadsby PJ. Anandamide is able to inhibit
trigeminal neurons using an in vivo model of trigeminovascular-
mediated nociception. J Pharmacol Exp Ther. 2003;309(1):56–63.
68. Akerman S, Kaube H, Goadsby PJ. Anandamide acts as a vasodila-
tor of dural blood vessels in vivo by activating TRPV1 receptors. Br
J Pharmacol. 2004;142:1354–60.
69. Manzanares J, Corchero J, Romero J, Fernandez-Ruiz JJ, Ramos
JA, Fuentes JA. Chronic administration of cannabinoids regulates
proenkephalin mRNA levels in selected regions of the rat brain.
Brain Res Mol Brain Res. 1998;55(1):126–32.
70. Cichewicz DL, Martin ZL, Smith FL, Welch SP. Enhancement of
mu opioid antinociception by oral delta9-tetrahydrocannabinol:
dose-response analysis and receptor identi cation. J Pharmacol
Exp Ther. 1999;289(2):859–67.
71. Cichewicz DL, Welch SP. Modulation of oral morphine antinocicep-
tive tolerance and naloxone-precipitated withdrawal signs by oral delta
9-tetrahydrocannabinol. J Pharmacol Exp Ther. 2003;305(3):812–7.
72. Cichewicz DL, McCarthy EA. Antinociceptive synergy between
delta(9)-tetrahydrocannabinol and opioids after oral administration.
J Pharmacol Exp Ther. 2003;304(3):1010–5.
73. Calhoun SR, Galloway GP, Smith DE. Abuse potential of dronabi-
nol (Marinol). J Psychoactive Drugs. 1998;30(2):187–96.
74. Clermont-Gnamien S, Atlani S, Attal N, Le Mercier F, Guirimand F,
Brasseur L. Utilisation thérapeutique du delta-9-tétrahydrocannabi-
nol (dronabinol) dans les douleurs neuropathiques réfractaires. The
therapeutic use of D9-tetrahydrocannabinol (dronabinol) in refrac-
tory neuropathic pain. Presse Med. 2002;31(39 Pt 1):1840–5.
75. Attal N, Brasseur L, Guirimand D, Clermond-Gnamien S, Atlami
S, Bouhassira D. Are oral cannabinoids safe and effective in refrac-
tory neuropathic pain? Eur J Pain. 2004;8(2):173–7.
76. Svendsen KB, Jensen TS, Bach FW. Does the cannabinoid dronabi-
nol reduce central pain in multiple sclerosis? Randomised double
blind placebo controlled crossover trial. BMJ. 2004;329(7460):253.
77. Buggy DJ, Toogood L, Maric S, Sharpe P, Lambert DG, Rowbotham
DJ. Lack of analgesic ef cacy of oral delta-9-tetrahydrocannabinol
in postoperative pain. Pain. 2003;106(1–2):169–72.
78. Neff GW, O’Brien CB, Reddy KR, et al. Preliminary observation
with dronabinol in patients with intractable pruritus secondary to
cholestatic liver disease. Am J Gastroenterol. 2002;97(8):
2117–9.
19518 Role of Cannabinoi ds in Pain Management
79. Narang S, Gibson D, Wasan AD, et al. Ef cacy of dronabinol as an
adjuvant treatment for chronic pain patients on opioid therapy.
J Pain. 2008;9(3):254–64.
80. Toth C, Au S. A prospective identi cation of neuropathic pain in
speci c chronic polyneuropathy syndromes and response to phar-
macological therapy. Pain. 2008;138(3):657–66.
81. Rintala DH, Fiess RN, Tan G, Holmes SA, Bruel BM. Effect of
dronabinol on central neuropathic pain after spinal cord injury: a
pilot study. Am J Phys Med Rehabil. 2010;89(10):840–8.
82. Lemberger L, Rubin A, Wolen R, et al. Pharmacokinetics, metabo-
lism and drug-abuse potential of nabilone. Cancer Treat Rev.
1982;9(Suppl B):17–23.
83. Notcutt W, Price M, Chapman G. Clinical experience with nabilone
for chronic pain. Pharm Sci. 1997;3:551–5.
84. Berlach DM, Shir Y, Ware MA. Experience with the synthetic can-
nabinoid nabilone in chronic noncancer pain. Pain Med.
2006;7(1):25–9.
85. Beaulieu P. Effects of nabilone, a synthetic cannabinoid, on postop-
erative pain: Les effets de la nabilone, un cannabinoide synthetique,
sur la douleur postoperatoire. Can J Anaesth. 2006;53(8):769–75.
86. Maida V. The synthetic cannabinoid nabilone improves pain and
symptom management in cancer patietns. Breast Cancer Res Treat.
2007;103(Part 1):121–2.
87. Wissel J, Haydn T, Muller J, et al. Low dose treatment with the
synthetic cannabinoid nabilone signi cantly reduces spasticity-
related pain: a double-blind placebo-controlled cross-over trial.
J Neurol. 2006;253(10):1337–41.
88. Frank B, Serpell MG, Hughes J, Matthews JN, Kapur D. Comparison
of analgesic effects and patient tolerability of nabilone and dihydro-
codeine for chronic neuropathic pain: randomised, crossover, dou-
ble blind study. BMJ. 2008;336(7637):199–201.
89. Skrabek RQ, Galimova L, Ethans K, Perry D. Nabilone for the
treatment of pain in bromyalgia. J Pain. 2008;9(2):164–73.
90. Ware MA, Fitzcharles MA, Joseph L, Shir Y. The effects of nabi-
lone on sleep in bromyalgia: results of a randomized controlled
trial. Anesth Analg. 2010;110(2):604–10.
91. Bestard JA, Toth CC. An open-label comparison of nabilone and
gabapentin as adjuvant therapy or monotherapy in the management
of neuropathic pain in patients with peripheral neuropathy. Pain
Pract. 2011;11:353–68. Epub 2010 Nov 18.
92. Karst M, Salim K, Burstein S, Conrad I, Hoy L, Schneider U. Analgesic
effect of the synthetic cannabinoid CT-3 on chronic neuropathic pain:
a randomized controlled trial. JAMA. 2003;290(13):1757–62.
93. Dyson A, Peacock M, Chen A, et al. Antihyperalgesic properties of
the cannabinoid CT-3 in chronic neuropathic and in ammatory
pain states in the rat. Pain. 2005;116(1–2):129–37.
94. Abrams DI, Jay CA, Shade SB, et al. Cannabis in painful HIV-
associated sensory neuropathy: a randomized placebo-controlled
trial. Neurology. 2007;68(7):515–21.
95. Wilsey B, Marcotte T, Tsodikov A, et al. A randomized, placebo-
controlled, crossover trial of cannabis cigarettes in neuropathic
pain. J Pain. 2008;9(6):506–21.
96. Wallace M, Schulteis G, Atkinson JH, et al. Dose-dependent effects
of smoked cannabis on capsaicin-induced pain and hyperalgesia in
healthy volunteers. Anesthesiology. 2007;107(5):785–96.
97. Ellis RJ, Toperoff W, Vaida F, et al. Smoked medicinal cannabis
for neuropathic pain in HIV: a randomized, crossover clinical trial.
Neuropsychopharmacology. 2009;34(3):672–80.
98. Ware MA, Wang T, Shapiro S, et al. Smoked cannabis for chronic
neuropathic pain: a randomized controlled trial. CMAJ.
2010;182(14):E694–701.
99. Wade DT, Makela PM, House H, Bateman C, Robson PJ. Long-
term use of a cannabis-based medicine in the treatment of spasticity
and other symptoms in multiple sclerosis. Mult Scler. 2006;12:
639–45.
100. Tashkin DP. Smoked marijuana as a cause of lung injury. Monaldi
Arch Chest Dis. 2005;63(2):93–100.
101. Tashkin DP, Simmons MS, Sherrill DL, Coulson AH. Heavy habit-
ual marijuana smoking does not cause an accelerated decline in
FEV1 with age. Am J Respir Crit Care Med. 1997;155(1):141–8.
102. Hashibe M, Morgenstern H, Cui Y, et al. Marijuana use and the
risk of lung and upper aerodigestive tract cancers: results of a
population-based case-control study. Cancer Epidemiol
Biomarkers Prev. 2006;15(10):1829–34.
103. Grinspoon L, Bakalar JB. Marihuana, the forbidden medicine.
Rev. and exp. edn. New Haven: Yale University Press; 1997.
104. Moir D, Rickert WS, Levasseur G, et al. A comparison of main-
stream and sidestream marijuana and tobacco cigarette smoke
produced under two machine smoking conditions. Chem Res
Toxicol. 2008;21(2):494–502.
105. Singh R, Sandhu J, Kaur B, et al. Evaluation of the DNA damag-
ing potential of cannabis cigarette smoke by the determination of
acetaldehyde derived N2-ethyl-2 ¢ -deoxyguanosine adducts. Chem
Res Toxicol. 2009;22(6):1181–8.
106. Joy JE, Watson SJ, Benson Jr JA. Marijuana and medicine: assessing
the science base. Washington D.C.: Institute of Medicine; 1999.
107. Gieringer D. Marijuana waterpipe and vaporizer study. MAPS
Bull. 1996;6(3):59–66.
108. Gieringer D. Cannabis “vaporization”: a promising strategy for
smoke harm reduction. J Cannabis Ther. 2001;1(3–4):153–70.
109. Storz M, Russo EB. An interview with Markus Storz. J Cannabis
Ther. 2003;3(1):67–78.
110. Gieringer D, St. Laurent J, Goodrich S. Cannabis vaporizer com-
bines ef cient delivery of THC with effective suppression of pyro-
lytic compounds. J Cannabis Ther. 2004;4(1):7–27.
111. Hazekamp A, Ruhaak R, Zuurman L, van Gerven J, Verpoorte R.
Evaluation of a vaporizing device (Volcano) for the pulmonary
administration of tetrahydrocannabinol. J Pharm Sci. 2006;95(6):
1308–17.
112. Van der Kooy F, Pomahacova B, Verpoorte R. Cannabis smoke
condensate I: the effect of different preparation methods on tetra-
hydrocannabinol levels. Inhal Toxicol. 2008;20(9):801–4.
113. Bloor RN, Wang TS, Spanel P, Smith D. Ammonia release from
heated ‘street’ cannabis leaf and its potential toxic effects on can-
nabis users. Addiction. 2008;103(10):1671–7.
114. Zuurman L, Roy C, Schoemaker RC, et al. Effect of intrapulmonary
tetrahydrocannabinol administration in humans. J Psychopharmacol
(Oxford, England). 2008;22(7):707–16.
115. Pomahacova B, Van der Kooy F, Verpoorte R. Cannabis smoke
condensate III: the cannabinoid content of vaporised Cannabis
sativa . Inhal Toxicol. 2009;21(13):1108–12.
116. Abrams DI, Vizoso HP, Shade SB, Jay C, Kelly ME, Benowitz
NL. Vaporization as a smokeless cannabis delivery system: a pilot
study. Clin Pharmacol Ther. 2007;82(5):572–8.
117. Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL.
Cannabinoid-opioid interaction in chronic pain. Clinical pharma-
cology and therapeutics. 2011;90(6):844–51.
118. Earleywine M, Barnwell SS. Decreased respiratory symptoms in
cannabis users who vaporize. Harm Reduct J. 2007;4:11.
119. Zajicek J, Fox P, Sanders H, et al. Cannabinoids for treatment of
spasticity and other symptoms related to multiple sclerosis (CAMS
study): multicentre randomised placebo-controlled trial. Lancet.
2003;362(9395):1517–26.
120. Zajicek JP, Sanders HP, Wright DE, et al. Cannabinoids in multi-
ple sclerosis (CAMS) study: safety and ef cacy data for 12
months follow up. J Neurol Neurosurg Psychiatry. 2005;76(12):
1664–9.
121. Ernst G, Denke C, Reif M, Schnelle M, Hagmeister H. Standardized
cannabis extract in the treatment of postherpetic neuralgia: a ran-
domized, double-blind, placebo-controlled cross-over study. Paper
presented at: international association for cannabis as medicine,
Leiden, 9 Sept 2005.
122. Holdcroft A, Maze M, Dore C, Tebbs S, Thompson S. A multi-
center dose-escalation study of the analgesic and adverse effects
196 E.B. Russo and A.G. Hohmann
of an oral cannabis extract (Cannador) for postoperative pain man-
agement. Anesthesiology. 2006;104(5):1040–6.
123. McPartland JM, Russo EB. Cannabis and cannabis extracts:
greater than the sum of their parts? J Cannabis Ther. 2001;
1(3–4):103–32.
124. de Meijer E. The breeding of cannabis cultivars for pharmaceuti-
cal end uses. In: Guy GW, Whittle BA, Robson P, editors.
Medicinal uses of cannabis and cannabinoids. London:
Pharmaceutical Press; 2004. p. 55–70.
125. Guy GW, Robson P. A phase I, double blind, three-way crossover
study to assess the pharmacokinetic pro le of cannabis based
medicine extract (CBME) administered sublingually in variant
cannabinoid ratios in normal healthy male volunteers
(GWPK02125). J Cannabis Ther. 2003;3(4):121–52.
126. Karschner EL, Darwin WD, McMahon RP, et al. Subjective and
physiological effects after controlled Sativex and oral THC admin-
istration. Clin Pharmacol Ther. 2011;89(3):400–7.
127. Karschner EL, Darwin WD, Goodwin RS, Wright S, Huestis MA.
Plasma cannabinoid pharmacokinetics following controlled oral
delta9-tetrahydrocannabinol and oromucosal cannabis extract
administration. Clin Chem. 2011;57(1):66–75.
128. Russo EB, Etges T, Stott CG. Comprehensive adverse event pro le
of Sativex. 18th annual symposium on the cannabinoids. Vol
Aviemore, Scotland: International Cannabinoid Research Society;
2008. p. 136.
129. Barnes MP. Sativex: clinical ef cacy and tolerability in the treat-
ment of symptoms of multiple sclerosis and neuropathic pain.
Expert Opin Pharmacother. 2006;7(5):607–15.
130. Pérez J. Combined cannabinoid therapy via na oromucosal spray.
Drugs Today. 2006;42(8):495–501.
131. Wade DT, Robson P, House H, Makela P, Aram J. A preliminary
controlled study to determine whether whole-plant cannabis
extracts can improve intractable neurogenic symptoms. Clin
Rehabil. 2003;17:18–26.
132. Notcutt W, Price M, Miller R, et al. Initial experiences with
medicinal extracts of cannabis for chronic pain: results from 34
“N of 1” studies. Anaesthesia. 2004;59:440–52.
133. Berman JS, Symonds C, Birch R. Ef cacy of two cannabis based
medicinal extracts for relief of central neuropathic pain from bra-
chial plexus avulsion: results of a randomised controlled trial.
Pain. 2004;112(3):299–306.
134. Rog DJ, Nurmiko T, Friede T, Young C. Randomized controlled
trial of cannabis based medicine in central neuropathic pain due to
multiple sclerosis. Neurology. 2005;65(6):812–9.
135. Nurmikko TJ, Serpell MG, Hoggart B, Toomey PJ, Morlion BJ,
Haines D. Sativex successfully treats neuropathic pain character-
ised by allodynia: a randomised, double-blind, placebo-controlled
clinical trial. Pain. 2007;133(1–3):210–20.
136. Wade DT, Makela P, Robson P, House H, Bateman C. Do canna-
bis-based medicinal extracts have general or speci c effects on
symptoms in multiple sclerosis? A double-blind, randomized,
placebo-controlled study on 160 patients. Mult Scler.
2004;10(4):434–41.
137. Blake DR, Robson P, Ho M, Jubb RW, McCabe CS. Preliminary
assessment of the ef cacy, tolerability and safety of a cannabis-
based medicine (Sativex) in the treatment of pain caused by rheu-
matoid arthritis. Rheumatology (Oxford). 2006;45(1):50–2.
138. Johnson JR, Burnell-Nugent M, Lossignol D, Ganae-Motan ED,
Potts R, Fallon MT. Multicenter, double-blind, randomized, pla-
cebo-controlled, parallel-group study of the ef cacy, safety, and
tolerability of THC:CBD extract and THC extract in patients with
intractable cancer-related pain. J Pain Symptom Manage.
2010;39(2):167–79.
139. Russo EB, Guy GW, Robson PJ. Cannabis, pain, and sleep: les-
sons from therapeutic clinical trials of Sativex, a cannabis-based
medicine. Chem Biodivers. 2007;4(8):1729–43.
140. Lynch ME, Young J, Clark AJ. A case series of patients using
medicinal marihuana for management of chronic pain under the
Canadian Marihuana Medical Access Regulations. J Pain
Symptom Manage. 2006;32(5):497–501.
141. Janse AFC, Breekveldt-Postma NS, Erkens JA, Herings RMC.
Medicinal gebruik van cannabis: PHARMO instituut. Institute for
Drug Outcomes Research; 2004.
142. Gorter RW, Butorac M, Cobian EP, van der Sluis W. Medical use
of cannabis in the Netherlands. Neurology. 2005;64(5):917–9.
143. Fitzgerald GA. Coxibs and cardiovascular disease. N Engl J Med.
2004;10:6.
144. Topol EJ. Failing the public health – rofecoxib, Merck, and the
FDA. N Engl J Med. 2004;10:6.
145. Grotenhermen F. Pharmacokinetics and pharmacodynamics of
cannabinoids. Clin Pharmacokinet. 2003;42(4):327–60.
146. Huestis MA, Henning fi eld JE, Cone EJ. Blood cannabinoids. I.
Absorption of THC and formation of 11-OH-THC and THCCOOH
during and after smoking marijuana. J Anal Toxicol. 1992;16(5):
276–82.
147. Wright S. GWMS001 and GWMS0106: maintenance of blinding.
London: GW Pharmaceuticals; 2005.
148. Clark P, Altman D. Assessment of blinding in phase III Sativex
spasticity studies. GW Pharmaceuticals; 2006.
149. Samaha AN, Robinson TE. Why does the rapid delivery of drugs
to the brain promote addiction? Trends Pharmacol Sci.
2005;26(2):82–7.
150. Jones RT, Benowitz N, Bachman J. Clinical studies of cannabis
tolerance and dependence. Ann N Y Acad Sci. 1976;282:221–39.
151. Budney AJ, Hughes JR, Moore BA, Vandrey R. Review of the
validity and signi cance of cannabis withdrawal syndrome. Am
J Psychiatry. 2004;161(11):1967–77.
152. Smith NT. A review of the published literature into cannabis
withdrawal symptoms in human users. Addiction. 2002;97(6):
621–32.
153. Solowij N, Stephens RS, Roffman RA, et al. Cognitive function-
ing of long-term heavy cannabis users seeking treatment. JAMA.
2002;287(9):1123–31.
154. Schoedel KA, Chen N, Hilliard A, et al. A randomized, double-
blind, placebo-controlled, crossover study to evaluate the abuse
potential of nabiximols oromucosal spray in subjects with a his-
tory of recreational cannabis use. Hum Psychopharmacol.
2011;26:224–36.
155. Russo EB, Mathre ML, Byrne A, et al. Chronic cannabis use in the
Compassionate Use Investigational New Drug Program: an exami-
nation of bene ts and adverse effects of legal clinical cannabis. J
Cannabis Ther. 2002;2(1):3–57.
156. Fride E, Russo EB. Neuropsychiatry: schizophrenia, depression,
and anxiety. In: Onaivi E, Sugiura T, Di Marzo V, editors.
Endocannabinoids: the brain and body’s marijuana and beyond.
Boca Raton: Taylor & Francis; 2006. p. 371–82.
157. Aragona M, Onesti E, Tomassini V, et al. Psychopathological and
cognitive effects of therapeutic cannabinoids in multiple sclerosis:
a double-blind, placebo controlled, crossover study. Clin
Neuropharmacol. 2009;32(1):41–7.
158. Degenhardt L, Hall W, Lynskey M. Testing hypotheses about the
relationship between cannabis use and psychosis. Drug Alcohol
Depend. 2003;71(1):37–48.
159. Macleod J, Davey Smith G, Hickman M. Does cannabis use cause
schizophrenia? Lancet. 2006;367(9516):1055.
160. Macleod J, Hickman M. How ideology shapes the evidence and
the policy: what do we know about cannabis use and what should
we do? Addiction. 2010;105:1326–30.
161. Hickman M, Vickerman P, Macleod J, et al. If cannabis caused
schizophrenia–how many cannabis users may need to be prevented
in order to prevent one case of schizophrenia? England and Wales
calculations. Addiction. 2009;104(11):1856–61.
19718 Role of Cannabinoids in Pain Management
162. Zuardi AW, Guimaraes FS. Cannabidiol as an anxiolytic and
antipsychotic. In: Mathre ML, editor. Cannabis in medical prac-
tice: a legal, historical and pharmacological overview of the thera-
peutic use of marijuana. Jefferson: McFarland; 1997. p. 133–41.
163. Russo EB. Taming THC: potential cannabis synergy and
phytocannabinoid-terpenoid entourage effects. Br J Pharmacol.
2011;163:1344–64.
164. Cabral G. Immune system. In: Grotenhermen F, Russo EB, editors.
Cannabis and cannabinoids: pharmacology, toxicology and thera-
peutic potential. Binghamton: Haworth Press; 2001. p. 279–87.
165. Katona S, Kaminski E, Sanders H, Zajicek J. Cannabinoid in uence
on cytokine pro le in multiple sclerosis. Clin Exp Immunol. 2005;
140(3):580–5.
166. Abrams DI, Hilton JF, Leiser RJ, et al. Short-term effects of can-
nabinoids in patients with HIV-1 infection. A randomized, placbo-
controlled clinical trial. Ann Intern Med. 2003;139:258–66.
167. Stott CG, Guy GW, Wright S, Whittle BA. The effects of cannabis
extracts Tetranabinex and Nabidiolex on human cytochrome
P450-mediated metabolism. Paper presented at: Symposium on
the Cannabinoids, Clearwater, 27 June 2005.
168. Stott CG, Ayerakwa L, Wright S, Guy G. Lack of human cyto-
chrome P450 induction by Sativex. 17th annual symposium on
the cannabinoids. Saint-Sauveur, Quebec: International
Cannabinoid Research Society; 2007. p. 211.
169. Grotenhermen F, Leson G, Berghaus G, et al. Developing limits
for driving under cannabis. Addiction. 2007;102(12):1910–7.
170. Hohmann AG, Suplita 2nd RL. Endocannabinoid mechanisms of
pain modulation. AAPS J. 2006;8(4):E693–708.
171. Clapper JR, Moreno-Sanz G, Russo R, et al. Anandamide sup-
presses pain initiation through a peripheral endocannabinoid
mechanism. Nat Neurosci. 2010;13:1265–70.
172. Schlosburg JE, Blankman JL, Long JZ, et al. Chronic monoacylg-
lycerol lipase blockade causes functional antagonism of the endo-
cannabinoid system. Nat Neurosci. 2010;13(9):1113–9.
173. Yu XH, Cao CQ, Martino G, et al. A peripherally restricted can-
nabinoid receptor agonist produces robust anti-nociceptive effects
in rodent models of in ammatory and neuropathic pain. Pain.
2010;151(2):337–44.
174. Izzo AA, Borrelli F, Capasso R, Di Marzo V, Mechoulam R. Non-
psychotropic plant cannabinoids: new therapeutic opportunities
from an ancient herb. Trends Pharmacol Sci. 2009;30(10):515–27.
175. Wirth PW, Watson ES, ElSohly M, Turner CE, Murphy JC. Anti-
in ammatory properties of cannabichromene. Life Sci. 1980;
26(23):1991–5.
176. Davis WM, Hatoum NS. Neurobehavioral actions of can-
nabichromene and interactions with delta 9-tetrahydrocannabinol.
Gen Pharmacol. 1983;14(2):247–52.
177. Ligresti A, Moriello AS, Starowicz K, et al. Antitumor activity of plant
cannabinoids with emphasis on the effect of cannabidiol on human
breast carcinoma. J Pharmacol Exp Ther. 2006;318(3):1375–87.
178. De Petrocellis L, Starowicz K, Moriello AS, Vivese M, Orlando P,
Di Marzo V. Regulation of transient receptor potential channels of
melastatin type 8 (TRPM8): effect of cAMP, cannabinoid CB(1)
receptors and endovanilloids. Exp Cell Res. 2007;313(9):1911–20.
179. Gauson LA, Stevenson LA, Thomas A, Baillie GL, Ross RA,
Pertwee RG. Cannabigerol behaves as a partial agonist at both
CB1 and CB2 receptors. 17th annual symposium on the cannabi-
noids. Vol Saint-Sauveur, Quebec: International Cannabinoid
Research Society; 2007, p. 206.
180. Cascio MG, Gauson LA, Stevenson LA, Ross RA, Pertwee RG.
Evidence that the plant cannabinoid cannabigerol is a highly
potent alpha2-adrenoceptor agonist and moderately potent 5HT1A
receptor antagonist. Br J Pharmacol. 2010;159(1):129-41.
181. Banerjee SP, Snyder SH, Mechoulam R. Cannabinoids: in fl uence
on neurotransmitter uptake in rat brain synaptosomes. J Pharmacol
Exp Ther. 1975;194(1):74–81.
182. Evans FJ. Cannabinoids: the separation of central from peripheral
effects on a structural basis. Planta Med. 1991;57(7):S60–7.
183. Evans AT, Formukong E, Evans FJ. Activation of phospholipase
A2 by cannabinoids. Lack of correlation with CNS effects. FEBS
Lett. 1987;211(2):119–22.
184. Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of
three plant cannabinoids: delta9-tetrahydrocannabinol, cannabid-
iol and delta9-tetrahydrocannabivarin. Br J Pharmacol. 2008;
153(2):199–215.
185. Hill AJ, Weston SE, Jones NA, et al. Delta-tetrahydrocannabivarin
suppresses in vitro epileptiform and in vivo seizure activity in
adult rats. Epilepsia. 2010;51(8):1522–32.
186. Bolognini D, Costa B, Maione S, et al. The plant cannabinoid
delta9-tetrahydrocannabivarin can decrease signs of in ammation
and in ammatory pain in mice. Br J Pharmacol. 2010;
160(3):677–87.
187. Mehmedic Z, Chandra S, Slade D, et al. Potency trends of delta(9)-
THC and other cannabinoids in con scated cannabis preparations
from 1993 to 2008. J Forensic Sci. 2010;55:1209–17.
188. King LA, Carpentier C, Grif ths P. Cannabis potency in Europe.
Addiction. 2005;100(7):884–6.
189. Potter DJ, Clark P, Brown MB. Potency of delta 9-THC and other
cannabinoids in cannabis in England in 2005: implications for
psychoactivity and pharmacology. J Forensic Sci. 2008;53(1):
90–4.
190. Potter D. Growth and morphology of medicinal cannabis. In: Guy
GW, Whittle BA, Robson P, editors. Medicinal uses of cannabis
and cannabinoids. London: Pharmaceutical Press; 2004.
p. 17–54.
191. Rao VS, Menezes AM, Viana GS. Effect of myrcene on nocicep-
tion in mice. J Pharm Pharmacol. 1990;42(12):877–8.
192. Lorenzetti BB, Souza GE, Sarti SJ, Santos Filho D, Ferreira SH.
Myrcene mimics the peripheral analgesic activity of lemongrass
tea. J Ethnopharmacol. 1991;34(1):43–8.
193. Basile AC, Sertie JA, Freitas PC, Zanini AC. Anti-in ammatory
activity of oleoresin from Brazilian Copaifera. J Ethnopharmacol.
1988;22(1):101–9.
194. Tambe Y, Tsujiuchi H, Honda G, Ikeshiro Y, Tanaka S. Gastric
cytoprotection of the non-steroidal anti-in ammatory sesquiter-
pene, beta-caryophyllene. Planta Med. 1996;62(5):469–70.
195. Gertsch J, Leonti M, Raduner S, et al. Beta-caryophyllene is a
dietary cannabinoid. Proc Natl Acad Sci USA. 2008;105(26):
9099–104.
197. Gil ML, Jimenez J, Ocete MA, Zarzuelo A, Cabo MM. Comparative
study of different essential oils of Bupleurum gibraltaricum
Lamarck. Pharmazie. 1989;44(4):284–7.
198. Re L, Barocci S, Sonnino S, et al. Linalool modi es the nicotinic
receptor-ion channel kinetics at the mouse neuromuscular junc-
tion. Pharmacol Res. 2000;42(2):177–82.
199. Gregg, L.C, Jung, K.M., Spradley, J.M., Nyilas, R., Suplita II,
R.L., Zimmer, A., Watanabe, M., Mackie, K., Katona, I., Piomelli,
D. and Hohmann, A.G. (2012) Activation of type-5 metabotropic
glutamate receptors and diacylglycerol lipase-alpha initiates
2-arachidonoylglycerol formation and endocannabinoid-mediated
analgesia in vivo. The Journal of Neuroscience, in press
[DOI:10.1523/JNEUROSCI.0013-12.2012].
200. Kavia R, De Ridder D, Constantinescu C, Stott C, Fowler C.
Randomized controlled trial of Sativex to treat detrusor overactiv-
ity in multiple sclerosis. Mult Scler. 2010;16(11):1349–59.
201. Portenoy RK, Ganae-Motan ED, Allende S, Yanagihara R, Shaiova
L, Weinstein S, et al. Nabiximols for opioid-treated cancer patients
with poorly-controlled chronic pain: a randomized, placebo-con-
trolled, graded-dose trial. J Pain. 2012;13(5):438–49.
202. Abrams DI, Couey P, Shade SB, Kelly ME, Benowitz NL.
Cannabinoid-opioid interaction in chronic pain. Clinical pharma-
cology and therapeutics. 2011;90(6):844–51.
... The Cα RMSD and Cα RMSF data provide insight into the dynamic structural behaviour of p-CBR. NADAs stable Cα RMSD and the unique conformational changes induced by CBD on CB1 yet CB2 displays an opposite effect which highlights the potential of these ligands in modulating receptor activities differently The differential responses of CB1 and CB2 receptors to these ligands, as indicated by the distinct structural changes observed particularly in membrane-bound regions, emphasise the complexity of receptor-ligand interactions and the necessity to consider this in drug design [63,64]. Since, additions were made the crystal structure in the ICL3, the membrane-bound region of the protein was assessed using both temporal simulation snapshots depicting changes over 250 ns and Cα backbone data. ...
Preprint
Full-text available
This study provides a comprehensive analysis of the conformational dynamics of pseudo-endocannabinoid receptors (p-CBRs) CB1 and CB2 in response to ligand binding. The ligands studied include N-arachidonoyl dopamine (NADA), Cannabidiol (CBD), known agonists (FUB-MDMB and E3R), and inverse agonists (Taranabant and AM630). Using homology models based on crystal structures (PDB ID: 5U09 for CB1 and 5ZTY for CB2), molecular docking, and molecular dynamics (MD) simulations, the research validates the structural integrity and dynamic behaviours of these pseudo-receptors. Induced-fit docking revealed NADA as having the highest binding affinity for both receptors, with docking scores of −13.92 kcal/mol for CB1 and −13.32 kcal/mol for CB2, facilitated by key hydrogen bond interactions. MD simulations over 250 ns at ~298 K showed successful equilibration, indicating the most stable interaction for NADA, thereby further validating the models. CB1 and CB2 exhibited distinct conformational responses to ligands, with Cα RMSD and Cα RMSF analyses suggesting a more rigid binding pocket for CB1 and a flexible binding pocket for CB2. These findings highlight the significance of receptor behaviour and dynamics in the development of cannabinoid-based therapeutics, supporting the potential of CBD and NADA as modulators for CB1 and CB2, respectively.
Article
Full-text available
Neuropathic pain, a complex and often devastating condition, poses significant challenges for its effective management. Despite promising research on various cannabis formulations and delivery methods for neuropathic pain, significant gaps remain in our knowledge. While inhaled cannabis shows analgesic effects and alternative delivery methods may improve bioavailability, oral formulations have yielded mixed results, often limited by small sample sizes and placebo effects. Therefore, further research is essential to optimize cannabis formulations, identify responder profiles to tailor treatments effectively, and, most critically, confirm the long-term safety and efficacy of cannabis-based therapies in managing NP. This review article aims to provide a comprehensive analysis of the therapeutic potential of cannabis-based medicines, with a particular focus on cannabinoids. This review, though not systematic, examines 11 clinical studies, specifically Randomised Clinical Trials) published from 2014 to 2024, highlighting the efficacy of numerous cannabis formulations, in alleviating neuropathic pain. Key findings show that cannabinoids can reduce pain perception, improve patient quality of life, and mitigate other symptoms associated with neuropathic pain. The synergistic effects of tetrahydrocannabinol and cannabidiol are discussed, emphasizing their ability to enhance analgesic effects, while potentially reducing the psychoactive side effects of tetrahydrocannabinol. This review emphasizes the importance of the personalized approach to improve therapeutic outcomes. Limitations of the existing research focusing on cannabis for neuropathic pain are limited by heterogeneity, lack of standardization, small sample sizes, and reliance on subjective outcomes, impacting the reliability and generalizability of findings. However, this exhaustive review aims to inform clinicians and researchers about the evolving role of cannabis in contemporary pain management strategies, illustrating the diverse pharmacological profiles of cannabinoids and their potential as adjunct therapies for neuropathic pain management.
Article
Cannabinoids, particularly cannabidiol (CBD), have been gaining attention for their numerous potential health benefits and are employed in various industries. However, there are unresolved challenges in CBD extraction including low yields, impurity issues, and environmental concerns, suggesting the requirement for green methods. Hence, our review objectives are to assess the efficacy, and the impact of novel solvents used for CBD extraction, considering green and sustainable techniques. The traditional extraction methods such as maceration and Soxhlet extraction used for CBD extraction have limitations such as low efficiency, long extraction times, high energy consumption, and substantial CO₂ emissions, raising environmental concerns. Emerging green extraction techniques, such as supercritical fluid extraction, deep eutectic solvents, and microwave-assisted extraction, offer promising alternatives by reducing solvent use, minimizing processing time, and enhancing extraction yields. Supercritical CO2 extraction, utilizing supercritical fluids' unique properties offers efficient and safe CBD extraction. Emerging green solvents such as ionic solvents and deep eutectic solvents provide promising alternatives for CBD extraction due to their properties such as no or low toxicity compared to the traditionally used solvents. Pressurized liquid extraction, such as subcritical water extraction, and techniques like microwave-assisted and ultrasonic-assisted extraction provide rapid and efficient alternatives for CBD extraction. To fulfill the growing demand for CBD extracts, future research can aim at developing efficient and sustainable extraction techniques while reducing the potential degradation of CBD, removing impurities produced during extraction, and considering concepts of sustainability and the One Health approach.
Article
Human security outcomes of cannabis decriminalisation are important for small island developing states. This study uses a convergent parallel mixed-methods research design and comparative case analysis of Jamaica and St. Vincent and the Grenadines to develop a results matrix to evaluate the human security outcomes of cannabis decriminalisation. The findings indicate that St. Vincent and the Grenadines' context-sensitive approach has been more effective in providing livelihood opportunities for traditional farmers. In comparison, Jamaica's reform, using a people-centred approach and supported by a revised four-point plan, has struggled to integrate traditional farmers into its regulatory framework. Both states demonstrated sensitivity to cannabis use through social justice via expungement legislation, and health security through medical access. However, challenges remain in measuring the impact on health security in St. Vincent and the Grenadines. Biennial surveys capturing key variables can address this deficit by identifying policy gaps and guiding adaptive approaches. The study contributes to the discourse on drug reform and evaluation methodology as the development of a results matrix can assess human well-being and establish benchmarks for evaluating state progress in enhancing human security. The findings offer insights for policymakers, stakeholders, and evaluators to establish a secure cannabis industry aligned with human security principles.
Article
Full-text available
As propriedades terapêuticas da planta Cannabis sativa L. é de conhecimento popular desde os povos antigos e, devido a mudanças socioculturais, sua utilização para fins terapêuticos foi reduzida. Entretanto, nos últimos anos, a busca por pesquisas científicas sobre esses efeitos terapêuticos e o incentivo de seu uso tem crescido largamente em todo mundo. O objetivo desta revisão bibliográfica é apresentar a história da planta Cannabis com uso medicinal e evidenciar seus benefícios para a odontologia. Com base em artigos encontrados nas plataformas SciELO, Pubmed e Google acadêmico, foram reunidas informações a respeito da trajetória do uso da planta na medicina e como ela interage com o nosso organismo. Deste modo, as evidências demonstram muitos benefícios para odontologia, como tratamentos de mucosites orais, devido ao efeito antioxidante e anti-inflamatório dos derivados da planta, de doenças periodontais, também pelo potencial de combate a inflamação e pela capacidade antimicrobiana e dor orofacial, uma vez que são considerados analgésicos potentes. Em geral, esta coletânea sugere que as substâncias advindas da planta, como o canabidiol, possuem propriedades biológicas promissoras e o crescente interesse nas pesquisas sobre plantas medicinais e seus fitocompostos fomentam a divulgação e os estudos sobre as aplicações da Cannabis sativa L. em odontologia.
Article
Introduction Cannabis sativa is a highly versatile plant with a long history of cultivation and domestication. It produces multiple compounds that exert distinct and valuable therapeutic effects by modulating diverse biological systems, including the endocannabinoid system (ECS). Access to standardized, metabolically diverse, and reproducible C. sativa chemotypes and chemovars is essential for physicians to optimize individualized patient treatment and for industries to conduct drug‐discovery campaigns. Objective This study aimed to characterize and assess the phytochemical diversity of C. sativa chemotypes in diverse ecological regions of Colombia, South America. Methodology Ten cannabinoids and 23 terpenes were measured using liquid and gas chromatography, in addition to other phenotypic traits, in 156 C. sativa plants that were grown in diverse ecological regions in Colombia, a hotspot for global biodiversity. Results Our results reveal significant phytochemical diversity in Colombian‐grown C. sativa plants, with four distinct chemotypes based on cannabinoid profile. The significant amount of usually uncommon terpenes suggests that Colombia's environments may have unique capabilities that allow the plant to express these compounds. Colombia's diverse climates offer enormous cultivation potential, making it a key player in both domestic and international medicinal and recreational C. sativa trade. Conclusion These findings underscore Colombia's capacity to pioneer global C. sativa production diversification, particularly in South America with new emerging markets.
Chapter
This chapter includes psychotropic drugs used in veterinary psychiatry that do not belong to the families presented previously. It concerns venlafaxine (anxiolytic), trazodone (anxiolytic, antidepressant, and hypnotic), buspirone (anxiolytic), bupropion (antidepressant, also used in nicotine addiction in humans), methylphenidate (stimulant), NMDA antagonists, including ketamine (anesthetic, analgesic), dextromethorphan, and memantine (both anticompulsive), atomoxetine (stimulant used in the treatment of ADHD in humans), and finally cannabinoids (analgesics and anxiolytics in humans). None of these molecules exist as licensed veterinary medications.
Chapter
Full-text available
The herb cannabis is derived from the Old World species Cannabis sativa L. Cannabis indica and C. ruderalis may also merit species status. Cannabis has a history as an analgesic agent that spans at least 4000 years, including a century of usage in mainstream Western medicine. Quality control issues, and ultimately political fiat eliminated this agent from the modern pharmacopoeia, but it is now resurgent. The reasons lie in the remarkable pharmacological properties of the herb and new scientific research that reveals the inextricable link that cannabinoids possess with our own internal biochemistry. In essence, the cannabinoids form a system in parallel with that of the endogenous opioids in modulating pain. More important, cannabis and its endogenous and synthetic counterparts may be uniquely effective in pain syndromes in which opiates and other analgesics fail.
Article
Full-text available
Activation of cannabinoid receptors inhibits voltage-gated Ca ²⁺ channels and activates K ⁺ channels, reminiscent of other G-protein-coupled signaling pathways that produce presynaptic inhibition. We tested cannabinoid receptor agonists for effects on excitatory neurotransmission between cultured rat hippocampal neurons. Reducing the extracellular Mg ²⁺ concentration to 0.1 m m elicited repetitive, transient increases in intracellular Ca ²⁺ concentration ([Ca ²⁺ ] i spikes) that resulted from bursts of action potentials, as measured by combined whole-cell current clamp and indo-1-based microfluorimetry. Pharmacological characterization indicated that the [Ca ²⁺ ] i spikes required glutamatergic synaptic transmission. Cannabinoid receptor ligands inhibited stereoselectively the frequency of [Ca ²⁺ ] i spiking in the rank order of potency: CP 54,939 > CP 55,940 > Win 55,212-2 > anandamide, with EC 50 values of 0.36, 1.2, 2.7, and 71 n m , respectively. CP 55,940 was potent, but not efficacious, and reversed the inhibition produced by Win 55,212-2, indicating that it is a partial agonist. Inhibition of [Ca ²⁺ ] i spiking by Win 55,212-2 was prevented by treatment of cultures with active, but not heat-treated, pertussis toxin. Win 55,212-2 (100 n m ) inhibited stereoselectively CNQX-sensitive excitatory postsynaptic currents (EPSCs) elicited by presynaptic stimulation with an extracellular electrode, but did not affect the presynaptic action potential or currents elicited by direct application of kainate. Consistent with a presynaptic site of action, Win 55,212-2 increased both the number of response failures and the coefficient of variation of the evoked EPSCs. In contrast, cannabimimetics did not affect bicuculline-sensitive inhibitory postsynaptic currents. Thus, activation of cannabinoid receptors inhibits the presynaptic release of glutamate via an inhibitory G-protein.
Article
Full-text available
Sativex(®), a cannabis extract oromucosal spray containing Δ(9)-tetrahydrocannabinol (THC) and cannabidiol (CBD), is currently in phase III trials as an adjunct to opioids for cancer pain treatment, and recently received United Kingdom approval for treatment of spasticity. There are indications that CBD modulates THC's effects, but it is unclear if this is due to a pharmacokinetic and/or pharmacodynamic interaction. Cannabis smokers provided written informed consent to participate in this randomized, controlled, double-blind, double-dummy institutional review board-approved study. Participants received 5 and 15 mg synthetic oral THC, low-dose (5.4 mg THC and 5.0 mg CBD) and high-dose (16.2 mg THC and 15.0 mg CBD) Sativex, and placebo over 5 sessions. CBD, THC, 11-hydroxy-THC, and 11-nor- 9-carboxy-THC were quantified in plasma by 2-dimensional GC-MS. Lower limits of quantification were ≤0.25 μg/L. Nine cannabis smokers completed all 5 dosing sessions. Significant differences (P < 0.05) in maximum plasma concentrations (C(max)) and areas under the curve from 0-10.5 h postdose (AUC(0→10.5)) for all analytes were found between low and high doses of synthetic THC and Sativex. There were no statistically significant differences in C(max), time to maximum concentration or in the AUC(0→10.5) between similar oral THC and Sativex doses. Relative bioavailability was calculated to determine the relative rate and extent of THC absorption; 5 and 15 mg oral THC bioavailability was 92.6% (13.1%) and 98.8% (11.0%) of low- and high-dose Sativex, respectively. These data suggest that CBD modulation of THC's effects is not due to a pharmacokinetic interaction at these therapeutic doses.
Article
Full-text available
This article reviews recent research on cannabinoid analgesia via the endocannabinoid system and non-receptor mechanisms, as well as randomized clinical trials employing canna- binoids in pain treatment. Tetrahydrocannabinol (THC, Marinol ® ) and nabilone (Cesamet ® ) are currently approved in the United States and other countries, but not for pain indications. Other synthetic cannabinoids, such as ajulemic acid, are in development. Crude herbal cannabis remains illegal in most jurisdictions but is also under investigation. Sativex ® , a cannabis derived oromucosal spray containing equal proportions of THC (partial CB 1 receptor agonist ) and can- nabidiol (CBD, a non-euphoriant, anti-infl ammatory analgesic with CB 1 receptor antagonist and endocannabinoid modulating effects) was approved in Canada in 2005 for treatment of central neuropathic pain in multiple sclerosis, and in 2007 for intractable cancer pain. Numer- ous randomized clinical trials have demonstrated safety and effi cacy for Sativex in central and peripheral neuropathic pain, rheumatoid arthritis and cancer pain. An Investigational New Drug application to conduct advanced clinical trials for cancer pain was approved by the US FDA in January 2006. Cannabinoid analgesics have generally been well tolerated in clinical trials with acceptable adverse event profi les. Their adjunctive addition to the pharmacological armamentarium for treatment of pain shows great promise.
Article
Full-text available
OBJECTIVES: This study examines the concept of clinical endocannabinoid deficiency (CECD), and the prospect that it could underlie the pathophysiology of migraine, fibromyalgia, irritable bowel syndrome, and other functional conditions alleviated by clinical cannabis. METHODS: Available literature was reviewed, and literature searches pursued via the National Library of Medicine database and other resources. RESULTS: Migraine has numerous relationships to endocannabinoid function. Anandamide (AEA) potentiates 5-HT1A and inhibits 5-HT2A receptors supporting therapeutic efficacy in acute and preventive migraine treatment. Cannabinoids also demonstrate dopamine-blocking and anti-inflammatory effects. AEA is tonically active in the periaqueductal gray matter, a migraine generator. THC modulates glutamatergic neurotransmission via NMDA receptors. Fibromyalgia is now conceived as a central sensitization state with secondary hyperalgesia. Cannabinoids have similarly demonstrated the ability to block spinal, peripheral and gastrointestinal mechanisms that promote pain in headache, fibromyalgia, IBS and related disorders. The past and potential clinical utility of cannabis-based medicines in their treatment is discussed, as are further suggestions for experimental investigation of CECD via CSF examination and neuro-imaging. CONCLUSION: Migraine, fibromyalgia, IBS and related conditions display common clinical, biochemical and pathophysiological patterns that suggest an underlying clinical endocannabinoid deficiency that may be suitably treated with cannabinoid medicines.
Article
The antinociceptive effects of various mu opioids given p.o. alone and in combination with Delta-9-tetrahydrocannabinol (Delta(9)-THC) were evaluated using the tail-flick test. Morphine preceded by Delta(9)-THC treatment (20 mg/kg) was significantly more potent than morphine alone, with an ED50 shift from 28.8 to 13.1 mg/kg. Codeine showed the greatest shift in ED,, value when administered after Delta(9)-IHC (139.9 to 5.9 mg/kg). The dose-response curves for oxymorphone acid hydromorphone were shifted 5- and 12.6-fold, respectively. Methadone was enhanced 4-fold, whereas its derivative, I-alpha-acetylmethadol, was enhanced 3-fold. The potency ratios after pretreatment with Delta(9)-THC for heroin and meperidine indicated significant enhancement (4.1 and 8.9, respectively). Pentazocine did not show a parallel shift in its dose-response curve with Delta(9)-THC. Naloxone administration (1 mg/kg s.c.) completely blocked the antinociceptive effects of morphine p.o. and codeine p.o. The Delta(9)-THC-induced enhancement of morphine and codeine was also significantly decreased by naloxone administration. Naltrindole (2 mg/kg s.c.) did not affect morphine or codeine antinociception but did block the enhancement of these two opioids by Delta(9)-THC. No effect was seen when nor-binaltorphimine was administered 2 mg/kg s.c. before morphine or codeine. Furthermore, the enhancements of morphine and codeine were not blocked by nor-binaltorphimine. We find that many mu opioids are enhanced by an inactive dose of Delta(9)-THC p.o. The exact nature of this enhancement is unknown. We show evidence of involvement of mu and possibly delta opioid receptors as a portion of this signaling pathway that leads to a decrease in pain perception.
Article
Delta(9)-tetrahydrocannabinol binds cannabinoid (CB(1) and CB(2)) receptors, which are activated by endogenous compounds (endocannabinoids) and are involved in a wide range of physiopathological processes (e.g. modulation of neurotransmitter release, regulation of pain perception, and of cardiovascular, gastrointestinal and liver functions). The well-known psychotropic effects of Delta(9)-tetra hydrocannabinol, which are mediated by activation of brain CB(1) receptors, have greatly limited its clinical use. However, the plant Cannabis contains many cannabinoids with weak or no psychoactivity that, therapeutically, might be more promising than Delta(9)-tetra hydrocannabinol. Here, we provide an overview of the recent pharmacological advances, novel mechanisms of action, and potential therapeutic applications of such non-psychotropic plant-derived cannabinoids. Special emphasis is given to cannabidiol, the possible applications of which have recently emerged in inflammation, diabetes, cancer, affective and neurodegenerative diseases, and to Delta(9)-tetrahydrocannabivarin, a novel CB(1) antagonist which exerts potentially useful actions in the treatment of epilepsy and obesity.