ArticlePDF AvailableLiterature Review

Neuroprotection in Experimental Autoimmune Encephalomyelitis and Progressive Multiple Sclerosis by Cannabis-Based Cannabinoids

Authors:

Abstract

Multiple sclerosis (MS) is the major immune-mediated, demyelinating, neurodegenerative disease of the central nervous system. Compounds within cannabis, notably Δ9-tetrahydrocannabinol (Δ9-THC) can limit the inappropriate neurotransmissions that cause MS-related problems and medicinal cannabis is now licenced for the treatment of MS symptoms. However, the biology indicates that the endocannabinoid system may offer the potential to control other aspects of disease. Although there is limited evidence that the cannabinoids from cannabis are having significant immunosuppressive activities that will influence relapsing autoimmunity, we and others can experimentally demonstrate that they may limit neurodegeneration that drives progressive disability. Here we show that synthetic cannabidiol can slow down the accumulation of disability from the inflammatory penumbra during relapsing experimental autoimmune encephalomyelitis (EAE) in ABH mice, possibly via blockade of voltage-gated sodium channels. In addition, whilst non-sedating doses of Δ9-THC do not inhibit relapsing autoimmunity, they dose-dependently inhibit the accumulation of disability during EAE. They also appear to slow down clinical progression during MS in humans. Although a 3 year, phase III clinical trial did not detect a beneficial effect of oral Δ9-THC in progressive MS, a planned subgroup analysis of people with less disability who progressed more rapidly, demonstrated a significant slowing of progression by oral Δ9-THC compared to placebo. Whilst this may support the experimental and biological evidence for a neuroprotective effect by the endocannabinoid system in MS, it remains to be established whether this will be formally demonstrated in further trials of Δ9-THC/cannabis in progressive MS.
INVITED REVIEW
Neuroprotection in Experimental Autoimmune
Encephalomyelitis and Progressive Multiple Sclerosis
by Cannabis-Based Cannabinoids
Gareth Pryce &Dieter R. Riddall &David L. Selwood &
Gavin Giovannoni &David Baker
Received: 26 October 2014 /Accepted: 10 December 2014
#Springer Science+Business Media New York 2014
Abstract Multiple sclerosis (MS) is the major immune-me-
diated, demyelinating, neurodegenerative disease of the cen-
tral nervous system. Compounds within cannabis, notably
Δ9-tetrahydrocannabinol (Δ9-THC) can limit the inappropri-
ate neurotransmissions that cause MS-related problems and
medicinal cannabis is now licenced for the treatment of MS
symptoms. However, the biology indicates that the
endocannabinoid system may offer the potential to control
other aspects of disease. Although there is limited evidence
that the cannabinoids from cannabis are having significant
immunosuppressive activities that will influence relapsing
autoimmunity, we and others can experimentally demonstrate
that they may limit neurodegeneration that drives progressive
disability. Here we show that synthetic cannabidiol can slow
down the accumulation of disability from the inflammatory
penumbra during relapsing experimental autoimmune
encephalomyelitis (EAE) in ABH mice, possibly via blockade
of voltage-gated sodium channels. In addition, whilst non-
sedating doses of Δ9-THC do not inhibit relapsing autoim-
munity, they dose-dependently inhibit the accumulation of
disability during EAE. They also appear to slow down clinical
progression during MS in humans. Although a 3 year, phase
III clinical trial did not detect a beneficial effect of oral Δ9-
THC in progressive MS, a planned subgroup analysis of
people with less disability who progressed more rapidly, dem-
onstrated a significant slowing of progression by oral Δ9-
THC compared to placebo. Whilst this may support the ex-
perimental and biological evidence for a neuroprotective ef-
fect by the endocannabinoid system in MS, it remains to be
established whether this will be formally demonstrated in
further trials of Δ9-THC/cannabis in progressive MS.
Keywords Cannabinoid .Cannabidiol .Experimental
autoimmune encephalomyelitis .Multiple sclerosis .
Neuroprotection .Δ9-tetrahydrocannabinol
Introduction
Multiple sclerosis (MS) is a major immune-mediated, demy-
elinating and neurodegenerative disease of the central nervous
system (CNS), which affects about 2-3 million people world-
wide (Compston and Coles 2002,2008). Disease is often
associated with relapsing-remitting neurological attacks and
the progressive, slow worsening of disability, typically over
many years. Demyelination and axonal and neuronal loss
leads to a variety of different cognitive, sensory and motor
problems that accumulate as disease progresses due to lesions
within different neural pathways of the CNS (Compston and
Coles 2002). At present there is no cure, although there are
some disease modifying therapies (DMT) that can slow down
the development of CNS lesions and neurological relapses
Electronic supplementary material The online version of this article
(doi:10.1007/s11481-014-9575-8) contains supplementary material,
which is available to authorized users.
G. Pryce :G. Giovannoni:D. Baker (*)
Neuroimmunology Unit, Blizard Institute, Barts and the London
School of Medicine and Dentistry, Queen Mary University of
London, 4 Newark Street, London E1 2AT, UK
e-mail: david.baker@qmul.ac.uk
G. Pryce
e-mail: g.pryce@qmul.ac.uk
G. Giovannoni
e-mail: g.giovannoni@qmul.ac.uk
D. R. Riddall :D. L. Selwood
Wolfson Institute of Biomedical Research, University College
London, London, UK
D. R. Riddall
e-mail: drr@ntlworld.com
D. L. Selwood
e-mail: d.selwood@ucl.ac.uk
J Neuroimmune Pharmacol
DOI 10.1007/s11481-014-9575-8
caused by the entry of cells of the peripheral immune system
into the CNS. These however, have relatively low efficacy, as
occurs with the beta interferons, glatiramer acetate and
teriflunomide, or higher efficacy which can be associated with
significant, sometimes life-threatening side effects, which has
been reported with fingolimod, natalizumab and alemtuzumab
(Marta and Giovannoni 2012). These can limit the nerve loss
that occurs as a consequence of these lesions (Gunnarsson
et al. 2011), however, these treatments if not started sufficient-
ly quickly following diagnosis, do not appear to control the
nerve loss associated with progressive MS. This is driven by
central inflammatory and other neurodegenerative effects that
underlie irreversible disability (Compston and Coles 2002;
Marta and Giovannoni 2012). Dysregulation of effective neu-
rotransmission leads to a number of troublesome symptoms
dependent on lesion location and include: incontinence;
spasms; spasticity and pain (Compston and Coles 2002).
These are controlled by a variety of different drugs, which
are often associated with significant sedating side effects
(Compston and Coles 2002). The failure to find adequate
treatments, leads people with MS (PwMS) to often seek
complementary or alternative medicines (CAM) to supple-
ment their prescribed medicines (Yadav et al. 2014;Masullo
et al. 2015). With the advent of the internet, use of CAM can
be widely publicised and adopted even before scientific evi-
dence can support or refute the claims of efficacy. Indeed
PwMS perceived benefit from taking cannabis for the control
of sleep disturbances, pain and spasticity (Consroeet al. 1997;
Clark et al. 2004; Chong et al. 2006). This was subsequently
supported by biology, experimental and clinical class I evi-
dence in humans to support the role of cannabinoid control of
spasticityandpaininPwMS(Bakeretal.2000,2012;
Novotna et al. 2011; Zajicek et al. 2012;Langfordetal.2013).
Symptom Control by Cannabinoids
The endocannabinoid systems regulates synaptic neurotrans-
mission and it is therefore not surprising that compounds
within cannabis can stimulate neuronal CB
1
cannabinoid re-
ceptors (CB
1
R) to control the excessive or inappropriate neu-
rotransmission that leads to symptoms of MS (Corey-Bloom
et al. 2012; Zajicek et al. 2012). Some places are now
supporting the use of medical marijuana, and cannabis ex-
tracts (Sativex/nabiximols) have become licensed medicines
for the treatment of spasticity and pain in MS (Novotna et al.
2011; Langford et al. 2013). Early reports from Europe and
the USA failed to distinguish any perceived therapeutic effi-
cacy in symptom control of MS (Consroe et al. 1997). How-
ever, in experimental models of MS-related spasticity that
occurs due to CNS autoimmunity, it could be shown that
delta9 tetrahydrocannabinol (Δ9-THC) and the CB
1
Rcon-
trolled symptoms, with no apparent effect of cannabinol
(CBD) on spasticity (Baker et al. 2000; Wilkinson et al.
2003; Pryce and Baker 2007; Pryce et al. 2014). This could
suggest that Δ9-THC is the major therapeutic chemical within
cannabis, based on the reports that cannabis in North America
may have a low CBD content (ElSohly et al. 2000; Wilkinson
et al. 2003; EMCDD 2008). However, pharmaceutical, med-
ical cannabis extracts being developed (Sativex & Cannador)
contain essentially equal proportions of Δ9-THC and CBD
(Novotna et al. 2011; Zajicek et al. 2012; Langford et al.
2013). Although it has been reported that CBD may limit
the side-effect potential of Δ9-THC within cannabis (Dalton
et al. 1976;RussoandGuy2006), little direct evidence has
been provided for such a specific ratio and contrasts with the
low CBD:Δ9-THC ratio (1:101:200) in many recreational
cannabis extracts (Burgdorf et al. 2011). However, it appears
that CBD is not inert and may have some medicinal value
(Mechoulam et al. 2002;RussoandGuy2006). Whilst me-
dicinal cannabis has become a licensed treatment for
symptom control, the question arises whether compounds
within cannabis have additional properties that could be
useful in the control of MS. We review the current
literature and present data to suggest that cannabis may
have utility in the control of nerve loss and disease
progression due to neuroimmunological disease.
Lack of Marked Immunosuppressive Effects
of Cannabinoids in EAE
Some studies have suggested that Δ9-THC and CBD may
have an immunosuppressive activity that could provide some
DMT function (Lyman et al. 1989;Mareszetal.2007;Kozela
et al. 2011). This is seen by a reduction in the incidence and
severity of disease and/or a delay in the onset of disease in
experimental autoimmune encephalomyelitis (EAE) models
of MS (Baker et al. 2011). In contrast to some immunosup-
pressive action of 5 mg/kg CBD reported in myelin-peptide
induced EAE in C57BL/6 mice (Kozela et al. 2011) and the
observation that 510 mg/kg CBD, but not 2.5 or 20 mg/kg
CBD, can inhibit the development of collagen-induced arthri-
tis in DBA-1 mice (Malfait et al. 2000), we have consistently
failed to detect any immunosuppressive effect in tissue ho-
mogenate induced EAE in ABH mice in multiple experiments
across a range of doses from 0.5 to 25 mg/kg. The lack of
immunosuppressive effects were found in an initial EAE
attack (Maresz et al. 2007) or as found here (Fig. 1)inan
induced-relapse; in the latter, essentially all the animals devel-
oped EAE of comparable severity and day of onset as found in
vehicle treated animals. This suggests that CBD is unlikely to
prevent relapsing neuroimmune-autoimmunity in MS.
Differences in the ease of immunosuppression in C57BL/6
(relatively EAE-resistant) and ABH (EAE susceptible) mice
have been seen previously (Sisay et al. 2013). As such,
apparent immunosuppression observed in EAE induced in
C57BL/6 mice, where disease induction can be inconsistent,
is lost once tested in ABH mice where robust and consistent
disease is induced (Sisay et al. 2013). Furthermore, in contrast
to the relapsing-remitting nature of EAE in ABH mice (Al-
Izki et al. 2012), myelin oligodendrocyte glycoprotein-
induced EAE in C57BL/6 is typically monophasic with poor
recovery (Sisay et al. 2013). This is related to inflammation
induced neurodegeneration. Thus the benefit of CBD may
relate to a neuroprotective effect rather than an immunosup-
pressive effect. Nevertheless, it is possible to induce immuno-
suppression of EAE with 5mg/kgΔ9-THC in both SJL
(Lyman et al. 1989) and ABH mice, via a neuronal CB
1
R-
dependent mechanism (Maresz et al. 2007;Croxfordetal.
A
Drug-Induced
Neuroprotection
Time Post-Disease Induction (days).
29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47
Mean Neurological Score ± SEM
0.0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
4.0
Vehicle
THC 2.5mg/kg i.p.
Period of Daily Treatment
B
C
Time of on Accelerating RotaRod (s)
0
50
100
150
200
250
300
Post- Relapse (Day 47)
**
Pre- Treatment (Day 27)
**
**
THC 10mg/kg i.p
CBD 10mg/kg i.p.
CBD 5mg/kg i.p.
Vehicle
Drug-Induced
Neuroprotection
Time Post-Disease Induction (days).
29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47
Mean Neurological Score ± SEM
0.0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
4.0
Vehicle
CBD 5mg/kg i.p
CBD 10mg/kg i.p.
Period of Daily Treatment
Fig. 1 Neuroprotective potential
of cannabinoids during induce-
relapsing autoimmune
encephalomyelitis. EAE was
induced in Biozzi ABH mice
following immunization with
spinal cord homogenate
emulsified in Freunds complete
adjuvant on day 0 and 7. (Al-Izki
et al. 2012). Animals were
allowed to undergo a paralytic
inflammatory attack (all animals
scored 34) and a relapse was
induced by re-immunization on
day 28 during the first remission
(RM1). Animals (79/group)
were injected i.p. with either: (A,
C) 2.5 mg/kg Δ9-THC, (B, C)
5 mg/kg CBD, 10 mg/kg CBD or
ethanol:cremophor:phosphate
buffered saline from day 33
onwards. (a,b)Theresults
represent the mean± SEM daily
score based on a 05 scoring scale
(Al-Izki et al. 2012). The
differences between the minimal
disease score at the termination of
the experiment were analysed
using Mann Whitney U statistics.
(c) The mean±SEM activity on
an accelerating rotorod (4
40 rpm) (Al-Izki et al. 2012)
measured on day 27 and day 47
during the second remission
(RM2). Differences between
vehicle and treatment groups
were analysed using Students t
tests ** P<0.01 compared to
vehicle treated animals
2008;deLagoetal.2012). However, we do not believe
that these effects are particularly relevant to the clinical
use of cannabis (Baker et al. 2012). This is because the
immunosuppression only occurs as doses of CB
1
Rago-
nists that cause marked sedative, cannabimimetic
(hypomotility; hypothermia and sometimes seizures) ef-
fects (Croxford et al. 2008). These probably cause
significant stress-responses that are known to be immu-
nosuppressive in EAE (Bolton et al. 1997). Importantly,
there is no solid data to suggest that doses of medical
cannabis cause significant immunosuppressive effects in
MS, following analysis of peripheral immune responses
(Killestein et al. 2003;Katonaetal.2005;Sextonetal.
2014).
Cannabinoid Dose
THC 0.0mg/kg 0.25mg/kg 2.5mg/kg 0.0mg/kg 0.0mg/kg 2.5mg/kg 2.5mg/kg
CBD 0mg/kg 0mg/kg 0mg/kg 5mg/kg 10mg/kg 5mg/kg 10mg/kg
Mean Clinical Score During Remission
0.0
0.5
1.0
1.5
2.0
2.5
3.0
RM1 Pre-Relapse
RM2 Post-Relapse
*
**
A
*
*
Cannabinoid Dose
THC 0.0mg/kg 0.25mg/kg 2.5mg/kg 0.0mg/kg 0.0mg/kg 2.5mg/kg 2.5mg/kg
CBD 0mg/kg 0mg/kg 0mg/kg 5mg/kg 10mg/kg 5mg/kg 10mg/kg
Mean Rotorod Score During Remission
0
50
100
150
200
250
300
RM1 Pre-Relapse
RM2 Post-Relapse
*
*
B
Fig. 2 Neuroprotective potential
of cannabinoids during induce-
relapsing autoimmune
encephalomyelitis. EAE was
induced in Biozzi ABH mice
following immunization with
spinal cord homogenate
emulsified in Freunds complete
adjuvant on day 0 and 7. (Al-Izki
et al. 2012) Animals were allowed
to undergo a paralytic
inflammatory attack (all score 3
4) and a relapse was induced by
re-immunization on day 28
during the first remission (RM1).
Animals (5-9/group) were
injected i.p. with either
0.25 mg/kg Δ9-THC, 2.5 mg/kg
Δ9-THC, 5 mg/kg CBD,
10 mg/kg CBD a combination of
Δ9-THC and CBD or
ethanol:cremophor:phosphate
buffered saline from day 33
onwards. (a) The results represent
the mean minimum score during
remission±SEM daily score
based on a 05 scoring scale (Al-
Izki et al. 2012). These were
analysed using Mann Whitney U
statistics. (b)Themean±SEM
activity on an accelerating rotorod
(Al-Izki et al. 2012) measured on
day 27 and day 47 during the
second remission (RM2).
*P<0.05, ** P<0.01 compared
to vehicle treated animals, using
Students ttest
Neuroprotective Effect of Cannabis-Plant Based
Cannabinoids in EAE
In contrast to the limited immunosuppressive action of
cannabis-based cannabinoids that inhibit the development of
paralytic EAE (Maresz et al. 2007; Baker et al. 2011), we and
others have shown that CB
1
R agonists, including Δ9-THC and
endocannabinoids, can induce a neuroprotective effect (Pryce
et al.2003; Croxford et al. 2008;Webbetal.2008;Hasseldam
and Johansen 2010; Hernández-Torres et al. 2014;Bernal-
Chico et al. 2015). In EAE, this is seen by a better functional
recovery and a reduced accumulation of disability following
paralytic attack (Croxford et al. 2008; Baker et al. 2011;Al-Izki
et al. 2014). As such, Δ9-THC facilitated a dose-dependent
enhanced recovery from the effects of the inflammatory pen-
umbra (Al-Izki et al.2014) developing during paralytic, relaps-
ing EAE (Figs. 1and 2) at a stage that allows us to dissociate
neuroprotective and immunosuppressive effects (Baker et al.
2011; Al-Izki et al. 2012). This was seen by significantly
(P<0.01) less accumulation of neurological disability, as
assessed using neurological score (Figs. 1a and 2a)andless
loss of motor co-ordination as assessed using rotorod activity
(Figs. 1c and 2b), which is consistent with neuroprotection
observed in previous studies (Pryce et al. 2003). This is prob-
ably mediated by multiple mechanisms including; alterations in
neural excitotoxicity, oxidative stress and changes in the glial
neuroimmune responses (Pryce et al. 2003; Docagne et al.
2007;Rossietal.2011a; Ribeiro et al. 2013; Musella et al.
2014). Whilst we have not found any symptomatic benefit or
immunosuppressive action of CBD in acute (Baker et al.2000;
Maresz et al. 2007) and relapsing EAE (Fig. 1b), interestingly
there was a significantly (P<0.05) better clinical recovery,
indicative of a neuroprotective effect (Figs. 1b and 2a) follow-
ing administration of 5 mg/kg and particularly 10 mg/kg CBD
i.p. (Fig. 2a) when administered shortly before relapse. This
clinical effect was also reflective of better rotorod activity
(Figs. 1c and 2b). For technical reasons, it was not possible to
measure spinal nerve content in these experiments to defini-
tively demonstrate an effect on nerve survival. However, based
on the clinical score and notably the rotorod score, which has a
very strong positive correlation with spinal nerve content (Al-
Izki et al. 2012,2014), it appeared evident that both Δ9-THC
and CBD alone had neuroprotective potential.
A neuroprotective effect of Δ9-THC was not surprising as
CB
1
R agonists, including Δ9-THC, have previously been
shown to have neuroprotective effects as shown histologically,
in other stages of EAE (Pryce et al.2003; Croxford et al. 2008).
Likewise, this is consistent with the observation that CB
1
R-
deficient mice have been shown to accumulate more neurode-
generation and disability as a consequence of neuroimmune
attack than wild type animals (Pryce et al. 2003; Rossi et al.
2011a).Hereitmaybeassociatedwiththeinhibitionofexces-
sive glutamatergic signals that can lead to downstream,
excitotoxic nerve damage, via neural CB
1
R-dependent mecha-
nisms (Pryce et al. 2003; Docagne et al. 2007; Musella et al.
2014). In addition, Δ9-THC also exhibits anti-oxidant proper-
ties (Hampson et al. 1998) and inhibits calcium and sodium ion
channels (Howlett et al. 2002) that can also limit nerve cell
death due to toxic ion concentrations within the disease CNS.
In addition, there may be actions through the inhibition of
microglial cell activity, which could be via CB
2
R(Howlett
et al. 2002; Docagne et al. 2007;Correaetal.2009). Likewise,
CBD has been found to offer neuroprotective potential in a
variety of different experimental paradigms (Mecha et al. 2012,
2013). Although a number of studies indicate that CBD has
neuroprotective anti-oxidant effects (El-Remessy et al. 2003;
Hayakawa et al. 2007), we show here that CBD also appears to
inhibit Na
+
ion channel activity (Fig. 3).
We used veratrine-evoked uptake of [
14
C]-guanidine flux
assay and inhibition of scorpion venom facilitated [
12
H]-
batrachotoxin-B (a sodium channel ligand; BTX-B) binding
using rat cerebral cortex synaptosomes to measure sodium
channel activity (Fig. 3.Garthwaite et al. 2002). In this assay,
veratrine holds the Na
+
channels in an open state and the influx
of [
14
C] guanidine through the channels and into the synapto-
somes is measured (Garthwaite et al. 2002). YC-1, a soluble
guanylylcyclase activator and Na
+
channel inhibitor (Garthwaite
et al. 2002) served as a positive control. This had an IC
50
of
16.2 μM in veratrine-evoked uptake of [
14
C]-guanidine flux
assay (Fig. 3a). This is comparable to the response reported
previously for YC-1 and showed a similar activity to Sipatrigine
(BW619C89. IC
50
=14.8 μM). Lamotrigine (IC
50
=186.2 μM),
which is a clinical sodium channel inhibitor used as an anti-
convulsive agent to treat epilepsy, had a lower inhibitory capac-
ity (Garthwaite et al. 2002). Cannabidiol, surprisingly, was over
ten times more active than YC-1, in this veratrine-evoked uptake
of [
14
C]-guanidine flux assay with an IC
50
of 0.9 μM(Fig.3a).
In contrast, nabilone, a potent CB
1
R/CB
2
R receptor agonist,
used to treat emesis in humans was inactive (>100 μM) in the
assay (Fig. 3a). Likewise nabilone was relatively inactive in
blockage of titrated BTX-B binding, whereas cannabidiol po-
tently (IC
50
of 34μM) inhibited binding of BTX-B to synap-
tosomes (Fig. 3b) and was again about ten times more potent
than YC-1 (IC
50
of 345.7 μM) and compared well with
Sipatrigine (IC
50
of 14.8 μM) and Lamotrigine (IC
50
of
159.6 μM Garthwaite et al. 2002). Our studies on sodium
channel blockage are supported and extended by other recent
studies that have also shown activity of CBD on sodium chan-
nels including Na
v
1.1, Na
v
1.2 and others. (Hill et al. 2014).
Sodium channel inhibitors can exhibit marked neuroprotective
effects in induced-relapsing EAE through effects on nerves and
microglia (Waxman 2002; Al-Izki et al. 2014; Morsali et al.
2013). Sodium channel blockers used to treat epilepsy may also
have some neuroprotective potential in MS (Gnanapavan et al.
2013). This is perhaps consistent with the mechanism of action
of CBD, although clinically CBD does not appear to have the
side-effect potential of some clinical sodium channel blockers,
which limited drug compliance and the perceived success of
clinical trials of this compound class in MS (Kapoor et al. 2010;
Gnanapavan et al. 2013). However, CBD is reported to have
additional ionic effects within mitochondria, and inhibits pro-
inflammatory activities of microglia and other modes of action
(Ryan et al. 2009; Mecha et al. 2012; Espejo-Porras et al. 2013;
Iannotti et al. 2014), which could also add to a neuroprotective
potential and in addition CBD has been reported to limit oligo-
dendrocyte damage (Mecha et al. 2012).
It was interesting however that there appeared to be no
additive effect following co-administration of both CBD with
Δ9-THC in 2:14:1 CBD:Δ9-THC ratios (Fig. 2). Further-
more, the neuroprotective effect of Δ9-THC appeared to be
abrogated by the presence of CBD (Fig. 2). It has been reported
that the presence of CBD limits the sedative and psychoactive
effect of Δ9-THC (Dalton et al. 1976; Russo and Guy 2006).
This may be compatible with the report that CBD has some
CB
1
R antagonist potential, which could alter some behavioural
effects of Δ9-THC (Thomas et al. 2007;Vannetal.2008). As
Log Concentration (M)
-9 -8 -7 -6 -5 -4
[
12
H] Batrachotoxin-B Binding(CPM)
0
500
1000
1500
2000
2500
3000
3500
YC-1
Cannabidiol
Nabilone
B
Log Concentration (M)
-9 -8 -7 -6 -5 -4
[
14
C]-Guanidine Flux (CPM)
0
1000
2000
3000
4000
5000
6000
7000
YC-1
Cannabidiol
Nabilone
A
Fig. 3 Sodium ion channel
Inhibitory activity of cannabidiol.
Various concentrations of CBD,
nabilone or YC-1 were incubated
with rat cerebral cortex
synaptosomes in the presence of
(a)veratrineand[
14
C]-guanidine
or (b) Scorpion venon and [
12
H]-
Batrachotoxin-B (Garthwaite
et al. 2002). Uptake of guanidine
or inhibition of binding of
batrachotoxin-B was assessed
using liquid scintillation
spectroscopy (Garthwaite et al.
2002). The results of CBD
affinities were repeated with
comparable results
such this could counteract the CB
1
R-mediated beneficial ef-
fects of Δ9-THC. Whether this impacts dosing of medical
cannabis is difficult to properly address in the absence of
human data. Although it been suggested that the influence of
CBD on CB
1
R-mediated effects of Δ9-THC are of marginal
significance at the concentrations of CBD in typical US of
smoked marijuana (Varvel et al. 2006), it may be relevant that
long-term follow-up in symptom-control trials suggested that
oral Δ9-THC but not oral 1:1 CBD:Δ9-THC cannabis extracts
(Cannador) containing comparable levels of Δ9-THC, could
limit the accumulation of disabilities in MS (Zajicek et al.
2005). This would be consistent with a neuroprotective effect
of Δ9-THC (Carroll et al. 2012), which was formally investi-
gated in a recent trial in MS (Zajicek et al. 2013).
Cannabinoids for the Control of Progression in Multiple
Sclerosis
Genetic depletion of CB
1
R in mice is associated with the
development of neurodegeneration (Pryce et al. 2003;Rossi
et al. 2011a). Furthermore, antagonism of CB
1
Rinhumans
Time (Weeks)
0 12 24 36 48 60 72 84 96 108 120 132 144 156
Proportion of Subjects with Stable EDSS (%)
0
10
20
30
40
50
60
70
80
90
100
Dronabinol (THC)
Placebo
P<0.01
Time (Weeks)
0 12 24 36 48 60 72 84 96 108 120 132 144 156
Proportion of Subjects with Stable EDSS (%)
0
10
20
30
40
50
60
70
80
90
100
A
Dronabinol (THC)
Placebo
Cohort of 5.5 EDSS or Less
Total Trial Cohort (EDSS 4.0-6.5)
B
Fig. 4 Inhibition of progressive
multiple sclerosis by oral
tetrahydrocannabinol. People
with progressive MS were
enrolled into a placebo-controlled
double blind clinical trial to assess
efficacy of twice daily oral
tetrahydrocannabinol (Grey)toa
maximum of 28 mg/day verses
vegetable oil placebo (Black)
capsules (Zajicek et al. 2013). The
probability of progression of
EDSS was performed for the (a)
total population (n=320Δ9-THC
and n=162 placebo) or (b)
participants with a baseline EDSS
score of 5 or lower (n=76 Δ9-
THC and n=34 placebo). The
results represent Kaplan-Meier
estimates of the proportion of
people with stable EDSS over
time weekly. The plot shows
timings of first events of
progression. Figures have been
presented and analysed in Zajicek
J. et al. Lancet Neurol
2013:12:857865+online
supplement. Reproduced with
permission from Elsevier
augments glutamatergic excitability (Oliviero et al. 2012),
which is known in excess to cause excitotoxicity in nerves.
It is of interest therefore that a non-coding; genetic variant of
the CB
1
R gene has been be associated with more rapid pro-
gression and neurodegeneration during inflammatory attack in
MS (Rossi et al. 2011b,2013). Endocannabinoid stimulation
of cannabinoid receptors has also been reported to be neuro-
protective (Eljaschewitsch et al. 2006). This suggests that
agonism of the CB
1
R by exogenous delivered cannabinoids
should have neuroprotective potential. However, unfortunate-
ly, it was reported that daily treatment with oral Δ9-THC had
no overall effect on the progression of MS in the progressive
phase of MS (Zajicek et al. 2013.Fig.4a). This may question
previous suggestions of a neuroprotective effect in MS
(Zajicek et al. 2005) and the accumulated experimental biol-
ogy (Baker et al. 2012). However, similar to the failure of Δ9-
THC in this progression trial, likewise, it was originally re-
ported that oral Δ9-THC and cannabis extract (Cannador) had
no effect on spasticity in a symptom-control trial (Zajicek et al.
2003). Likewise, other cannabis extracts (Sativex) failed to
alter the Ashworth scale as a measure of spasticity in symptom
control trials in MS (Wade et al. 2003). However, through
adapting clinical trial design, duration and the outcome mea-
sures, it has been found that cannabis can indeed control
symptoms of MS (Zajicek et al. 2005,2012; Novotna et al.
2011; Corey-Bloom et al. 2012). This indicates that trial
design is critical in the detection of therapeutic effects and
the translation of animal studies into human benefit (Baker
and Amor 2014). In the trial of Δ9-THC in progressive MS,
lower than expected progression rates occurred and so may
have affected the ability to detect clinical change, which
would require longer trials (Zajicek et al. 2013). Clinical
progression in MS is assessed using the Expanded Disability
Status Score (EDSS). This is a motor score ranging from
health (EDSS Score 0); walking without aid or rest for
500 m (EDSS Score 4), walking with an aid/cane (EDSS
score 6); essentially restricted to a wheel chair (EDSS 7); to
death (EDSS 10). However, this neurological rating system is
not linear and PwMS can progress at variable rates (Leray
et al. 2010). However, progression between from EDSS 3 to
EDSS 6 is more consistent and potentially more rapid (Leray
et al. 2010). Therefore, trials enriched for this subset of PwMS
may have a greater chance of detecting differences. Whilst it
was clear that daily Δ9-THC, at the doses tested, did not slow
progression (Fig. 4a), it is of immense interest that analysis of
a subset of people with MS with an EDSS 5.5 demonstrated
that Δ9-THC significantly (P<0.01) slowed disease progres-
sion (Fig. 4b; Zajicek et al. 2013). This would be consistent
with the accumulating biological knowledge and supportive
experimental evidence in animal models (Baker et al. 2000,
2012) and would strongly suggest that cannabinoids indeed
have the potential to control neuroimmune processes that lead
to neurodegeneration. Furthermore, the presence of Δ9-THC
in the blood, presumably due to recreational cannabis use, was
associated with a better prognosis following traumatic brain
injury (Nguyen et al. 2014). This possibly suggests a neuro-
protective effect from cannabinoid use in humans.
Conclusions
Despite the potential promise of cannabis to control progres-
sion in MS, it remains to be established whether similar trials
of Δ9-THC in progressive MS, using a revised trial design,
will be repeated to deliver licensed treatments. This may be
difficult for investigator-led academic studies due to the per-
ceived failure of the original trial (Zajicek et al. 2013), making
it difficult to raise the significant funds required to undertake
similar studies. This failure in humans probably also stifles
support for further basic science research in this area. Follow-
ing the initial failure of academic led-trials in spasticity
(Zajicek et al. 2003), interest was maintained because of
commercial development of alternative products. However,
in contrast to the short symptom control trials of a few weeks
duration (Novotna et al. 2011; Corey-Bloom et al. 2012),
phase III trials in progressive MS will probably require a
further 56 years to do a 3 year trial that recruits sufficiently
large numbers of people for the trials (estimated to be n=375
with 90 % power to detect an 18 % treatment effect. Zajicek
et al. 2013). Such a study would suffer from competitive
recruitment to other pharmaceutical company and academic-
investigator led trials in progressive MS, where a perceived
failure has not yet occurred. Importantly, unless pharmaceuti-
cal companies are involved, it will be difficult to perform
further studies, typically two phase III trials, to a level actually
required for regulatory approval and licensing (Giovannoni
et al. 2015). However, because of poor patent protection of
oral Δ9-THC as a potential medicine for MS, coupled with
MS drug-pricing structures, where symptom control drugs are
significantly cheaper than current DMT, it means that there
will probably belittle major pharmaceutical interest in funding
and undertaking these studies using a symptom control drug.
The proliferation of outlets supplying relatively cheap,
legalised, medical marijuana that is occurring, notably in the
USA, which is the major commercial market in MS, under-
mines both the chance of commercial development in this area
and the chances of recruiting to placebo-controlled trials, in
regions where medical cannabis is readily available. Whether
retrospective analysis of large numbers of long-term cannabis
users and non-users for symptom control can detect effects on
progression remains to be determined. However, development
of patent-protected formulations of cannabinoids or non-
cannabis pharmaceuticals such as endocannabinoid modula-
tors may be one way to develop commercial interest that could
exploit the cannabinoid biology to help deliver a treatment of
progression in MS.
Acknowledgments The authors thank the support of the National MS
Society (USA) and the MS Society (UK). We thank Prof. John Zajicek
and Susan Ball, Plymouth, UK for providing access to data from the
CUPID trial.
Conflicts of Interest None.
REFERENCES
Al-Izki S, Pryce G, ONeill JK, Butter C, Giovannoni G, Amor S, Baker
D (2012) Practical guide to the induction of relapsing progressive
experimental autoimmune encephalomyelitis in the Biozzi ABH
mouse. Mult Scler Rel Dis 1:2938
Al-Izki S, Pryce G, Hankey DJ, Lidster K, von Kutzleben SM, Browne L,
Clutterbuck L, Posada C, Edith Chan AW, Amor S, Perkins V,
Gerritsen WH, Ummenthum K, Peferoen-Baert R, van der Valk P,
Montoya A, Joel SP, Garthwaite J, Giovannoni G, Selwood DL, Baker
D (2014) Lesional-targeting of neuroprotection to the inflammatory
penumbra in experimental multiple sclerosis. Brain 137:92108
Baker D, Amor S (2012) Publication guidelines for refereeing and
reporting on animal use in experimental autoimmune encephalomy-
elitis. J Neuroimmunol 242:7883
Baker D, Amor S (2014) Experimental autoimmune encephalomyelitis is
a good model of multiple sclerosis if used wisely. Mult Scler Rel Dis
3:555564
Baker D, Pryce G, Croxford JL, Brown P, Pertwee RG, Huffman JW,
Layward L (2000) Cannabinoids control spasticity and tremor in a
multiple sclerosis model. Nature 404:8487
Baker D, Gerritsen W, Rundle J, Amor S (2011) Critical appraisal of
animal models of multiple sclerosis. Mult Scler 17:647657
Baker D, Pryce G, Jackson SJ, Bolton C, Giovannoni G (2012) The
biology that underpins the therapeutic potential of cannabis-based
medicines for the control of spasticity in multiple sclerosis. Mult
Scler Rel Dis 1:6475
Bernal-Chico A, Canedo M, Manterola A, Victoria Sánchez-Gómez M,
Pérez-Samartín A, Rodríguez-Puertas R, Matute C, Mato S (2015)
Blockade of monoacylglycerol lipase inhibits oligodendrocyte
excitotoxicity and prevents demyelination in vivo. Glia 63:163176
Bolton C, ONeill JK, Allen SJ, Baker D (1997) Regulation of chronic
relapsing experimental allergic encephalomyelitis by endogenous
and exogenous glucocorticoids. Int Arch Allergy Immunol 114:
7480
Burgdorf JR, Kilmer B, Pacula RL (2011) Heterogeneity in the compo-
sition of marijuana seized in California. Drug Alcohol Depend 117:
5961
Carroll CB, Zeissler ML, Hanemann CO, Zajicek JP (2012) Δ9-
tetrahydrocannabinol (Δ9-THC) exerts a direct neuroprotective ef-
fect in a human cell culture model of Parkinsonsdisease.
Neuropathol Appl Neurobiol 38:535547
Chong MS, Wolff K, Wise K, Tanton C, Winstock A, Silber E (2006)
Cannabis use in patients with multiple sclerosis. Mult Scler 12:646
651
Clark AJ, Ware MA, Yazer E, Murray TJ, Lynch ME (2004) Patterns of
cannabis use among patients with multiple sclerosis. Neurology 62:
20982100
Compston A, Coles A (2002) Multiple sclerosis. Lancet 359:12211231
Compston A, Coles A (2008) Multiple sclerosis. Lancet 372:15021517
Consroe P, Musty R, Rein J, Tillery W, Pertwee R (1997) The perceived
effects of smoked cannabis on patients with multiple sclerosis. Eur
Neurol 38:4448
Corey-Bloom J, Wolfson T, Gamst A, Jin S, Marcotte TD, Bentley H,
Gouaux B (2012) Smoked cannabis for spasticity in multiple scle-
rosis: a randomized, placebo-controlled trial. CMAJ 184:11431150
Correa F, Docagne F, Mestre L, Clemente D, Hernangómez M, Loría F,
Guaza C (2009) A role for CB2 receptors in anandamide signalling
pathways involved in the regulation of IL-12 and IL-23 in microglial
cells. Biochem Pharmacol 77:86100
Croxford JL, Pryce G, Jackson SJ, Ledent C, Giovannoni G, Pertwee RG,
Yamamura T, Baker D (2008) Cannabinoid-mediated neuroprotec-
tion, not immunosuppression, may be more relevant to multiple
sclerosis. J Neuroimmunol 193:120129
Dalton WS, Martz R, Lemberger L, Rodda BE, Forney RB (1976)
Influence of cannabidiol on delta-9-tetrahydrocannabinol effects.
Clin Pharmacol Ther 19:300309
de Lago E, Moreno-Martet M, Cabranes A, Ramos JA, Fernández-Ruiz J
(2012) Cannabinoids ameliorate disease progression in a model of
multiple sclerosis in mice, acting preferentially through CB1
receptor-mediated anti-inflammatory effects. Neuropharmacology
62:22992308
DocagneF,MuñetónV,ClementeD,AliC,LoríaF,CorreaF,
Hernangómez M, Mestre L, Vivien D, Guaza C (2007)
Excitotoxicity in a chronic model of multiple sclerosis:
Neuroprotective effects of cannabinoidsthrough CB1 and CB2 re-
ceptor activation. Mol Cell Neurosci 34:551561
Eljaschewitsch E, Witting A, Mawrin C, Lee T, Schmidt PM, Wolf S,
Hoertnagl H, Raine CS, Schneider-Stock R, Nitsch R, Ullrich O
(2006) The endocannabinoid anandamide protects neurons during
CNS inflammation by induction of MKP-1 in microglial cells.
Neuron 49:6779
El-Remessy AB, Khalil IE, Matragoon S, Abou-Mohamed G, Tsai NJ,
Roon P, Caldwell RB, Caldwell RW, Green K, Liou GI (2003)
Neuroprotective effect of ()Delta9-tetrahydrocannabinol and
cannabidiol in N-methyl-D-aspartate-induced retinal neurotoxicity:
involvement of peroxynitrite. Am J Pathol 163:19972008
ElSohly MA, Ross SA, Mehmedic Z, Arafat R, Yi B, Banahan BF (2000)
Potency trends of delta9-Δ9-THC and other cannabinoids in con-
fiscated marijuana from 19801997. J Forensic Sci 45:2430
Espejo-Porras F, Fernández-Ruiz J, Pertwee RG, Mechoulam R, García C
(2013) Motor effects of the non-psychotropic
phytocannabinoidcannabidiol that are mediated by 5-HT1A recep-
tors. Neuropharmacology 75:155163
European Monitoring Centre for Drugs and Drug Addiction (2008) A
cannabis reader: global issues and local experiences, Monograph
series 8, vol 1. European Monitoring Centre for Drugs and Drug
Addiction, Lisbon
Garthwaite G, Goodwin DA, Neale S, Riddall D, Garthwaite J (2002)
Soluble guanylylcyclase activator YC-1 protects white matter axons
from nitric oxide toxicity and metabolic stress, probably through
Na(+) channel inhibition. Mol Pharmacol 61:97104
Giovannoni G, Baker D, Schmierer K (2015) The problems with
repurposing: is there really an alternative to Big Pharmafor devel-
oping new drugs for MS? Mult Scler Rel Dis. doi:10.1016/j.msard.
2014.11.005
Gnanapavan S, Grant D, Morant S, Furby J, Hayton T, Teunissen CE,
Leoni V, Marta M, Brenner R, Palace J, Miller DH, Kapoor R,
Giovannoni G (2013) Biomarker report from the phase II
lamotrigine trial in secondary progressive MS - neurofilament as a
surrogate of disease progression. PLoS One 8:e70019
Gunnarsson M, Malmeström C, Axelsson M, Sundström P, Dahle C,
Vrethem M, Olsson T, Piehl F, Norgren N, Rosengren L,
Svenningsson A, Lycke J (2011) Axonal damage in relapsing multiple
sclerosis is markedly reduced by natalizumab. Ann Neurol 69:8389
Hampson AJ, Grimaldi M, Axelrod J, Wink D (1998) Cannabidiol and
()Delta9-tetrahydrocannabinol are neuroprotective antioxidants.
Proc Natl Acad Sci U S A 95:82688273
Hasseldam H, Johansen FF (2010) Neuroprotection without
immunomodulation is not sufficient to reduce first relapse
severity in experimental autoimmune encephalomyelitis.
Neuroimmunomodulation 17:252264
Hayakawa K, Mishima K, Nozako M, Hazekawa M, Irie K, Fujioka M,
Orito K, Abe K, Hasebe N, Egashira N, Iwasaki K, Fujiwara M
(2007) Delayed treatment with cannabidiol has a cerebroprotective
action via a cannabinoid receptor-independent myeloperoxidase-
inhibiting mechanism. J Neurochem 102:14881496
Hernández-Torres G, Cipriano M, Hedén E, Björklund E, Canales A,
Zian D, Feliú A, Mecha M, Guaza C, Fowler CJ, Ortega-Gutiérrez
S, López-Rodríguez ML (2014) A reversible and selective inhibitor
of monoacylglycerol lipase ameliorates multiple sclerosis. Angew
Chem Int Ed Engl 53:1376513770
Hill AJ, Jones NA, Smith I, Hill CL, Williams CM, Stephens GJ, Whalley
BJ (2014) Voltage-gated sodium (NaV) channel blockade by plant
cannabinoids does not confer anticonvulsant effects per se. Neurosci
Lett 566:269274
Howlett AC, Barth F, Bonner TI, Cabral G, Casellas P, Devane WA,
Felder CC, Herkenham M, Mackie K, Martin BR, Mechoulam
R, Pertwee RG (2002) International Union of Pharmacology.
XXVII. Classification of cannabinoid receptors. Pharmacol Rev
54:161202
Iannotti FA, Hill CL, Leo A, Alhusaini A, Soubrane C, Mazzarella E,
Russo E, Whalley BJ, Di Marzo V, Stephens GJ (2014) The non-
psychotropic plant cannabinoids, cannabidivarin (CBDV) and
cannabidiol (CBD), activate and desensitize transient receptor po-
tential vanilloid 1 (TRPV1) channels in vitro: potential for the
treatment of neuronal hyperexcitability. ACS Chem Neurosci 5:
11311141
Kapoor R, Furby J, Hayton T, Smith KJ, Altmann DR, Brenner R,
Chataway J, Hughes RA, Miller DH (2010) Lamotrigine for neuro-
protection in secondary progressive multiple sclerosis: a
randomised, double-blind, placebo-controlled, parallel-group trial.
Lancet Neurol 9:681688
Katona S, Kaminski E, Sanders H, Zajicek J (2005) Cannabinoid influ-
ence on cytokine profile in multiple sclerosis. Clin Exp Immunol
140:580585
Killestein J, Hoogervorst EL, Reif M, Blauw B, Smits M, Uitdehaag BM,
Nagelkerken L, Polman CH (2003) Immunomodulatory effects of
orally administered cannabinoids in multiple sclerosis. J
Neuroimmunol 137:140143
Kozela E, Lev N, Kaushansky N, Eilam R, Rimmerman N, Levy R, Ben-
Nun A, Juknat A, Vogel Z (2011) Cannabidiol inhibits pathogenic T
cells, decreases spinal microglial activation and ameliorates multiple
sclerosis-like disease in C57BL/6 mice. Br J Pharmacol 16:1507
1519
Langford RM, Mares J, Novotna A, Vachova M, Novakova I, Notcutt W,
Ratcliffe S (2013) A double-blind, randomized, placebo-controlled,
parallel-group study of Δ9-THC/CBD oromucosal spray in combi-
nation with the existing treatment regimen, in the relief of central
neuropathic pain in patients with multiple sclerosis. J Neurol 260:
984997
Leray E, Yaouanq J, Le Page E, Coustans M, Laplaud D, Oger J, Edan G
(2010) Evidence for a two-stage disability progression in multiple
sclerosis. Brain 133:19001913
LymanWD, Sonett JR, Brosnan CF, Elkin R, Bornstein MB (1989) Delta
9-tetrahydrocannabinol: a novel treatment for experimental autoim-
mune encephalomyelitis. J Neuroimmunol 23:7381
Malfait AM, Gallily R, Sumariwalla PF, Malik AS, Andreakos E,
Mechoulam R, Feldmann M (2000) The nonpsychoactive can-
nabis constituent cannabidiol is an oral anti-arthritic therapeu-
tic in murine collagen-induced arthritis. Proc Natl Acad Sci U
SA97:95619566
Maresz K, Pryce G, Ponomarev ED, Marsicano G, Croxford JL, Shriver
LP, Ledent C, Cheng X, Carrier EJ, Mann MK, Giovannoni G,
Pertwee RG, Yamamura T, Buckley NE, Hillard CJ, Lutz B, Baker
D, Dittel BN (2007) Direct suppression of CNS autoimmune in-
flammation via the cannabinoid receptor CB1 on neurons and CB2
on autoreactive T cells. Nat Med 13:492497
Marta M, Giovannoni G (2012) Disease modifying drugs in
multiple sclerosis: mechanisms of action and new drugs in
the horizon.CNSNeurolDisord Drug. Targets 11:610623
Masullo L, Papas MA, Cotugna N, Baker S, Mahoney L, Trabulsi J
(2015) Complementary and alternative medicine use and nutrient
intake among individuals with multiple sclerosis in the United
States. J Community Health. Jul 1.2014 [Epub ahead of print]
Mecha M, Torrao AS, Mestre L, Carrillo-Salinas FJ, Mechoulam R,
Guaza C (2012) Cannabidiol protects oligodendrocyte progenitor
cells from inflammation-induced apoptosis by attenuating endoplas-
mic reticulum stress. Cell Death Dis 3:e331
Mecha M, Fel A, Iñigo PM, Mestre L, Carrillo-Salinas FJ, Guaza C
(2013) Cannabidiol provides long-lasting protection against the
deleterious effects of inflammation in a viral model of multiple
sclerosis: a role for A2A receptors. Neurobiol Dis 59:141150
Mechoulam R, Parker LA, Gallily R (2002) Cannabidiol: an overview of
some pharmacological aspects. J Clin Pharmacol 42(11 Suppl):11S
19S
Morsali D, Bechtold D, Lee W, Chauhdry S, Palchaudhuri U, Hassoon P,
Snell DM, Malpass K, Piers T, Pocock J, Roach A, Smith KJ (2013)
Safinamide and flecainide protect axons and reduce microglial acti-
vation in models of multiple sclerosis. Brain 136:10671082
Musella A, Sepman H, Mandolesi G, Gentile A, Fresegna D, Haji N,
Conrad A, Lutz B, Maccarrone M, Centonze D (2014) Pre- and
postsynaptic type-1 cannabinoid receptors control the alterations of
glutamate transmission in experimental autoimmune encephalomy-
elitis. Neuropharmacology 79:567572
Nguyen BM, Kim D, Bricker S, Bongard F, Neville A, Putnam B, Smith
J, Plurad D (2014) Effect of marijuana use on outcomes in traumatic
brain injury. Am Surg 80:979983
Novotna A, Mares J, Ratcliffe S, Novakova I, Vachova M, Zapletalova O,
Gasperini C, Pozzilli C, Cefaro L, Comi G, Rossi P, Ambler Z,
Stelmasiak Z, Erdmann A, Montalban X, Klimek A, Davies P,
Sativex Spasticity Study Group (2011) A randomized, double-blind,
placebo-controlled, parallel-group, enriched-design study of
nabiximols* (Sativex(®)), as add-on therapy, in subjects with refractory
spasticity caused by multiple sclerosis. Eur J Neurol 18:11221131
Oliviero A, Arevalo-Martin A, Rotondi M, García-Ovejero D, Mordillo-
Mateos L, Lozano-Sicilia A, Panyavin I, Chiovato L, Aguilar J,
Foffani G, Di Lazzaro V, Molina-Holgado E (2012) CB1 receptor
antagonism/inverse agonism increases motor system excitability in
humans. Eur Neuropsychopharmacol 22:2735
Pauwels PJ, Leysen JE, Laduron PM (1986) [
3
H]Batrachotoxinin A 20-
alpha-benzoate binding to sodium channels in rat brain: characteriza-
tion and pharmacological significance. Eur J Pharmacol 124:291298
Pryce G, Baker D (2007) Control of spasticity in a multiple sclerosis
model is mediated by CB1, not CB2, cannabinoid receptors. Br J
Pharmacol 150:519525
Pryce G, Ahmed Z, Hankey DJ, Jackson SJ, Croxford JL, Pocock JM,
Ledent C, Petzold A, Thompson AJ, Giovannoni G, Cuzner ML,
Baker D (2003) Cannabinoids inhibit neurodegeneration in models
of multiple sclerosis. Brain 126:21912202
Pryce G, Visintin C, Ramagopalan SV, Al-Izki S, De Faveri LE, Nuamah
RA, Mein CA, Montpetit A, Hardcastle AJ, Kooij G, de Vries HE,
Amor S, Thomas SA, Ledent C, Marsicano G, Lutz B, Thompson
AJ, Selwood DL, Giovannoni G, Baker D (2014) Control of spas-
ticity in a multiple sclerosis model using central nervous system-
excluded CB1 cannabinoid receptor agonists. FASEB J 28:117130
Ribeiro R, Yu F, Wen J, Vana A, Zhang Y (2013) herapeutic potential ofa
novel cannabinoid agent CB52 in the mouse model of experimental
autoimmune encephalomyelitis. Neuroscience 254:427442
Rossi S, Furlan R, De Chiara V, Muzio L, Musella A, Motta C, Studer V,
Cavasinni F, Bernardi G, Martino G, Cravatt BF, Lutz B,
Maccarrone M, Centonze D (2011a) Cannabinoid CB1 receptors
regulate neuronal TNF-αeffects in experimental autoimmune en-
cephalomyelitis. Brain Behav Immun 25:12421248
Rossi S, Buttari F, Studer V, Motta C, Gravina P, Castelli M, Mantovani
V, De Chiara V, Musella A, Fiore S, Masini S, Bernardi G,
Maccarrone M, Bernardini S, Centonze D (2011b) The (AAT)n
repeat of the cannabinoid CB1 receptor gene influences disease
progression inrelapsing multiple sclerosis. Mult Scler 17:281288
Rossi S, Bozzali M, Bari M, Mori F, Studer V, Motta C, Buttari F,
Cercignani M, Gravina P, Mastrangelo N, Castelli M, Mancino R,
Nucci C, Sottile F, Bernardini S, Maccarrone M, Centonze D (2013)
Association between a genetic variant of type-1 cannabinoid recep-
tor and inflammatory neurodegeneration in multiple sclerosis. PLoS
One 8(12):e82848
Russo E, Guy GW (2006) A tale of two cannabinoids: the therapeutic
rationale for combining tetrahydrocannabinol and cannabidiol. Med
Hypotheses 66:234246
Ryan D, Drysdale AJ, Lafourcade C, Pertwee RG, Platt B (2009)
Cannabidiol targets mitochondria to regulate intracellular Ca2+
levels. J Neurosci 29:20532063
Sexton M, Cudaback E, Abdullah RA, Finnell J, Mischley LK, Rozga M,
Lichtman AH, Stella N (2014) Cannabis use by individuals with
multiple sclerosis: effects on specific immune parameters.
Inflammopharmacology 2014(22):295303
Sisay S, Pryce G, Jackson SJ, Tanner C, Ross RA, Michael GJ,
Selwood DL, Giovannoni G, Baker D (2013) Genetic back-
ground can result in a marked or minimal effect of gene
knockout (GPR55 and CB2 receptor) in experimental auto-
immune encephalomyelitis models of multiple sclerosis. PLoS
One 8(10):e76907
Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG
(2007) Cannabidiol displays unexpectedly high potency as an an-
tagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol
150:613623
Vann RE, Gamage TF, Warner JA, Marshall EM, Taylor NL,
Martin BR, Wiley JL (2008) Divergent effects of cannabidiol
on the discriminative stimulus and place conditioning effects
of Delta(9)-tetrahydrocannabinol. Drug Alcohol Depend 94:
191198
Varvel SA, Wiley JL, Yang R, Bridgen DT, Long K, Lichtman AH,
Martin BR (2006) Interactions between Δ9-THC and cannabidiol
in mouse models of cannabinoid activity. Psychopharmacology 186:
226234
Wade DT, Robson P, House H, Makela P, Aram J (2003) A preliminary
controlled study to determine whether whole-plant cannabisextracts
can improve intractable neurogenic symptoms. Clin Rehabil 17:21
219
Waxman SG (2002) Sodium channels as molecular targets in multiple
sclerosis. J Rehabil Res Dev 39:233242
Webb M, Luo L, Ma JY, Tham CS (2008) Genetic deletion of Fatty Acid
Amide Hydrolase results in improved long-term outcome in chronic
autoimmune encephalitis. Neurosci Lett 439:106110
Wilkinson JD, Whalley BJ, Baker D, Pryce G, Constanti A, Gibbons S,
Williamson EM (2003) Medicinal cannabis: is delta9-
tetrahydrocannabinol necessary for all its effects? J Pharm
Pharmacol 55:16871694
Yadav V, Bever C Jr, Bowen J, Bowling A, Weinstock-Guttman B,
Cameron M, Bourdette D, Gronseth GS, Narayanaswami P (2014)
Summary of evidence-based guideline: complementary and alterna-
tive medicine in multiple sclerosis: report of the guideline develop-
ment subcommittee of the American Academy of Neurology.
Neurology 82:10831092
Zajicek J, Fox P, Sanders H, Wright D, Vickery J, Nunn A, Thompson A,
UK MS Research Group (2003) Cannabinoids for treatment of
spasticity and other symptoms related to multiple sclerosis (CAMS
study): multicentre randomised placebo-controlled trial.Lancet 362:
15171526
Zajicek JP, Sanders HP, Wright DE, Vickery PJ, Ingram WM, Reilly SM,
Nunn AJ, Teare LJ, Fox PJ, Thompson AJ (2005) Cannabinoids in
multiple sclerosis (CAMS) study: safety and efficacy data for 12
months follow up.J NeurolNeurosurg. Psychiatry 76:16641669
Zajicek JP, Hobart JC, Slade A, Barnes D, Mattison PG, MUSEC
Research Group (2012) Multiple sclerosis and extract of cannabis:
results of the MUSEC trial.J NeurolNeurosurg. Psychiatry 83:1125
1132
Zajicek J, Ball S, Wright D, Vickery J, Nunn A, Miller D, Gomez Cano
M, McManus D, Mallik S, Hobart J, CUPID investigator group
(2013) Effect of dronabinol on progression in progressive multiple
sclerosis (CUPID): a randomised, placebo-controlled trial. Lancet
Neurol 12:857865
Materials and Methods
Induction of autoimmune experimental encephalomyelitis protocols con-
sistent with the ARRIVE guidelines have been published previously (Al-
Izki et al. 2012; Baker and Amor 2012). Animal studies were approved
following local ethical and United Kingdom Government, Home Office
review in accordance with the United Kingdom Animals (Scientific
Procedures) Act 1986. Full working protocols of the methods and doses
and use of cannabinoids in animals have been reported previously (Pryce
et al. 2003; Croxford et al. 2008; Al-Izki et al. 2012). Briefly, 68week
adult Biozzi ABH mice (Al-Izki et al. 2012) were injected with spinal
cord homogenate in Freunds adjuvant on day 0 and 7 to induce experi-
mental autoimmune encephalomyelitis with onset around day 1519
post-inoculation (p.i.) and again during remission from paralytic attack
on day 28 p.i. to induce a relapse 78 days later (Al-Izki et al. 2012).
Animals were scored daily: 0= normal; 1=limp tail; 2=impaired righting
reflex; 3=hindlimb paresis; 4=hindlimb paralysis; 5=moribund. Scores
were assessed using Mann Whitney U statistics (Al-Izki et al. 2012). The
motor co-ordination was assessed on an accelerating (040 rpm/
5 min) rotorod and analysed using Students ttest following normality
and equal variance tests (Al-Izki et al.2012). Synthetic Δ9-THC and
CBD were purchased from Δ9-THC Pharm GmbH. Frankfurt, Ger-
many and were diluted in alcohol:cremophor:phosphate buffered
saline (1:1:18). Various doses injected in 0.1 ml intraperitioneally
(i.p). as described previously (Pryce et al. 2003;Croxfordetal.
2008). These were administered shortly before anticipated relapse
(Al-Izki et al. 2012,2014).
Veratrine induced flux of [
14
C]guanidine in synaptosomes has been
reported previously (Pauwels et al. 1986; Garthwaite et al. 2002). Briefly
veratrine (100 μg/ml final concentration), and rat cerebral cortex synap-
tosomes (4 mg/ml, wet weight) were incubated in the absence or presence
of compound at 37 °C for 5 min in polypropylene test tubes. Uptake was
initiated by the addition of pre-warmed [
14
C]-guanidine (final concentra-
tion 1 μCi/ml) and stopped 2 min later by the addition of 10 ml of ice-cold
wash medium as described previously (Pauwels et al. 1986). Incubates
were immediately filtered under vacuum through GF/C filters by using a
Brandel harvester. The incubation tubes were rinsed with 5 ml of ice-cold
wash buffer, which was then used to wash the filter. Filters were trans-
ferred to minivials (Beckman Coulter, Fullerton, CA) with the use of a
Brandel deposit/dispense system and subsequently counted by liquid
scintillation spectroscopy with Picofluor40 liquid scintillator (Garthwaite
et al. 2002). Cannabidiol; YC-1 (5-[1-phenylmethyl)-1H-indazol-3-yl]-2-
furanmethanol (Cayman, Chem Ann Arbor, Michigan, USA); Nabilone
(Cambridge Labs; Newcastle, UK); Lamotrigine (6-(2,3-
Dichlorophenyl)-1,2,4-triazine-3,5-diamine. Tocris, Bristol, UK) and
Sipatrigine (BW619C89. 2-(4-Methyl-1-piperazinyl)-5-(2,3,5-
trichlorophenyl)-4-pyrimidinamine. Tocris Ltd) were diluted with medi-
um from 10 mM stock solutions.
Batrachotoxin-B (BTX-B) Binding. This was performed as described
previously (Garthwaite et al. 2002). Binding was initiated by the addition
of synaptosomes (final concentration 10 mg/ml, wet weight) to a mixture
of test compound and 10 nM [
3
H]Batrachotoxin-B in the absence or
presence of scorpion venom (25 μg/ml final concentration). Samples
were mixed and incubated for 90 min at 25 °C. Ice-cold wash medium
(5 ml) was added and then the samples subjected to vacuum filtration
through GF/C filters by using a Brandel harvester. Incubation tubes were
rinsed with 5 ml of ice-cold wash buffer, which was then used to washthe
filter. Radioactivity in the filter was counted as described above.
Randomised, double-blind, placebo-controlled trial of Δ9-THC in
people with progressive MS has been reported previously (Zajicek et al.
2013), with an International Standard Randomised Controlled Trial num-
ber 62942668. Human studies were approved by the South and West
Devon Research Ethics Committee and done in accordance with Good
Clinical Practice guidelines. Eligible patients provided written informed
consent before participation as International Standard Randomised
Controlled Trial (ISRCTN 62942668). Briefly, 1865 year old humans
with primary or secondary progressive MS (Expanded disability status
scale (EDSS) Score4.06.5), not on current disease modifying therapy
(DMT), were enrolled into the study. These were randomised to oral
dronabinol (Δ9-THC) starting at 3.5 mg twice a day escalated to a
maximum of 28 mg/day depending on tolerability (n=329)orvegetable
oil placebo in gelatin capsules (n=164). These were supplied by Insys
Therapeutics (Phoenix, AZ, USA). Analysis of the total population (n=
493) or subgroup analysis on time to progression in those participants
with a baseline EDSS score of 5 or lower (n=110) was performed using
a log-rank test to compare probability of progression between treatment
groups (Zajicek et al. 2013).
... ∆ 9 -THC pleiotropic effects such as analgesic, muscle relaxation, and pain tolerance [1] increases weight and appetite; improves sleep and depression; alters mood, behavior, feeling, and thoughts [72,73] cures neuropathic pain, spasticity, dysphoria, and anxiety disorders [74,75] antiemetic activity, i.e., prevents vomiting in cancer patient during chemotherapy [76] displays antiglaucoma activity and reduces intraocular pressure [76] ∆ 9 -THCA neuroprotective and antitumor activity [77] ∆ 9 -THCV non-psychoactive effect; cures obesity [75] effective against metabolic disorders, pancreatic disease, and hepatosteatosis syndrome [1] CBC non-psychotropic and anti-inflammatory activity [77,78] CBD cures memory loss, obesity, convulsive disorder, and rheumatoid arthritis [79] cures epilepsy; exhibits antipsychotic, antinausea, and antianxiety activity [80] CBDA anti-inflammatory and antihyperalgesia effect [81,82] CBG non-psychotic activity [75] CBC, CBL, and DCA cures HIV and cancer; possess immune boosting activity [83] CBG and amofrutin anti-inflammatory activity [84] CBL anti-inflammatory, anti-microbial, antipsychotic, and antiallergic activity [5] GFA and DCA anti-microbial activity [85] PET increases analgesia, catalepsy, hypolocomotion and hypothermia [28] 6.1. Cannabinoid Receptors in Humans and Their Role CB 1 R (cannabinoid receptor type 1, first cloned in 1990) mRNA is highly expressed in the brain. ...
... ∆ 9 -THC pleiotropic effects such as analgesic, muscle relaxation, and pain tolerance [1] increases weight and appetite; improves sleep and depression; alters mood, behavior, feeling, and thoughts [72,73] cures neuropathic pain, spasticity, dysphoria, and anxiety disorders [74,75] antiemetic activity, i.e., prevents vomiting in cancer patient during chemotherapy [76] displays antiglaucoma activity and reduces intraocular pressure [76] ∆ 9 -THCA neuroprotective and antitumor activity [77] ∆ 9 -THCV non-psychoactive effect; cures obesity [75] effective against metabolic disorders, pancreatic disease, and hepatosteatosis syndrome [1] CBC non-psychotropic and anti-inflammatory activity [77,78] CBD cures memory loss, obesity, convulsive disorder, and rheumatoid arthritis [79] cures epilepsy; exhibits antipsychotic, antinausea, and antianxiety activity [80] CBDA anti-inflammatory and antihyperalgesia effect [81,82] CBG non-psychotic activity [75] CBC, CBL, and DCA cures HIV and cancer; possess immune boosting activity [83] CBG and amofrutin anti-inflammatory activity [84] CBL anti-inflammatory, anti-microbial, antipsychotic, and antiallergic activity [5] GFA and DCA anti-microbial activity [85] PET increases analgesia, catalepsy, hypolocomotion and hypothermia [28] 6.1. Cannabinoid Receptors in Humans and Their Role CB 1 R (cannabinoid receptor type 1, first cloned in 1990) mRNA is highly expressed in the brain. ...
... ∆ 9 -THC pleiotropic effects such as analgesic, muscle relaxation, and pain tolerance [1] increases weight and appetite; improves sleep and depression; alters mood, behavior, feeling, and thoughts [72,73] cures neuropathic pain, spasticity, dysphoria, and anxiety disorders [74,75] antiemetic activity, i.e., prevents vomiting in cancer patient during chemotherapy [76] displays antiglaucoma activity and reduces intraocular pressure [76] ∆ 9 -THCA neuroprotective and antitumor activity [77] ∆ 9 -THCV non-psychoactive effect; cures obesity [75] effective against metabolic disorders, pancreatic disease, and hepatosteatosis syndrome [1] CBC non-psychotropic and anti-inflammatory activity [77,78] CBD cures memory loss, obesity, convulsive disorder, and rheumatoid arthritis [79] cures epilepsy; exhibits antipsychotic, antinausea, and antianxiety activity [80] CBDA anti-inflammatory and antihyperalgesia effect [81,82] CBG non-psychotic activity [75] CBC, CBL, and DCA cures HIV and cancer; possess immune boosting activity [83] CBG and amofrutin anti-inflammatory activity [84] CBL anti-inflammatory, anti-microbial, antipsychotic, and antiallergic activity [5] GFA and DCA anti-microbial activity [85] PET increases analgesia, catalepsy, hypolocomotion and hypothermia [28] 6.1. Cannabinoid Receptors in Humans and Their Role CB 1 R (cannabinoid receptor type 1, first cloned in 1990) mRNA is highly expressed in the brain. ...
Article
Full-text available
Phytocannabinoids are a structurally diverse class of bioactive naturally occurring compounds found in angiosperms, fungi, and liverworts and produced in several plant organs such as the flower and glandular trichrome of Cannabis sativa, the scales in Rhododendron, and oil bodies of liverworts such as Radula species; they show a diverse role in humans and plants. Moreover, phytocannabinoids are prenylated polyketides, i.e., terpenophenolics, which are derived from isoprenoid and fatty acid precursors. Additionally, targeted productions of active phytocannabinoids have beneficial properties via the genes involved and their expression in a heterologous host. Bioactive compounds show a remarkable non-hallucinogenic biological property that is determined by the variable nature of the side chain and prenyl group defined by the enzymes involved in their biosynthesis. Phytocannabinoids possess therapeutic, antibacterial, and antimicrobial properties; thus, they are used in treating several human diseases. This review gives the latest knowledge on their role in the amelioration of abiotic (heat, cold, and radiation) stress in plants. It also aims to provide synthetic and biotechnological approaches based on combinatorial biochemical and protein engineering to synthesize phytocannabinoids with enhanced properties.
... Accruing evidence supports a neuroprotective and immunomodulatory action of THC in multiple sclerosis animal models (Pryce et al., 2015). Here, we identify CNS remyelination as a key mechanism underlying THC protective actions under demyelinating conditions. ...
... However, although nonsedating doses of THC do not inhibit relapsing autoimmunity in the experimental autoimmune encephalomyelitis model, they dose-dependently inhibit the accumulation of disability (Croxford et al., 2008;Pryce et al., 2015), suggesting a regenerative action of THC in this animal model. The complex immunomodulatory actions of THC in immune-mediated demyelination animal models can therefore mask the regenerative capacity of the compound. ...
Article
Full-text available
Background and purpose: Research on demyelinating disorders aims to find novel molecules able to induce oligodendrocyte precursor cell differentiation to promote CNS remyelination and functional recovery. Δ9 -Tetrahydrocannabinol (THC), the most prominent active constituent of the hemp plant Cannabis sativa, confers neuroprotection in animal models of demyelination. However, the possible effect of THC on myelin repair has never been studied. Experimental approach: By using oligodendroglia-specific reporter mouse lines in combination with 2 models of toxin-induced demyelination, we analyzed the effect of THC on the processes of oligodendrocyte regeneration and functional remyelination. Key results: We show that THC administration enhanced oligodendrocyte regeneration, white matter remyelination, and motor function recovery. THC also promoted axonal remyelination in organotypic cerebellar cultures. THC remyelinating action relied on the induction of oligodendrocyte precursor differentiation upon cell cycle exit and via CB1 cannabinoid receptor activation. Conclusions and implications: Overall, our study identifies THC administration as a promising pharmacological strategy aimed to promote functional CNS remyelination in demyelinating disorders.
... These observations were confirmed by several other groups (12)(13)(14) using various models of EAE. However, we would like to state that the group of Maresz et al. (5) using a model of homogenized spinal cord-induced EAE did not find CBD to be effective in ameliorating the disease symptoms (see also 15). CBD at 5 mg/kg was also shown to reduce the symptoms of another autoimmune disease, a model of rheumatoid arthritis in mice (16). ...
... (À)-8,9-Dihydro-7-hydroxy-CBD (6) Synthesis of (+)-8,9-dihydro-7-hydroxy-CBD (14) and (+)-8,9-dihydro-6-hydroxy-CBD (15) (+)-8,9-Dihydro-CBD (10) Basic aluminum oxide (156 mg) was added to dry dichloromethane (150 mL) (Scheme 2). To this suspension, BF 3 Ádiethyletherate (2.3 mL) was added under nitrogen. ...
... 24 Each patient received 200-300 mg daily of (CBD)25 or placebo along with antiepileptic medications for up to 4 months.26 They found that in the treatment group 7 of 8 responded with fewer seizures, 27 this result corroborates with studies by Pryce (2018) 28 where they point out that cannabis use decreases some symptoms associated with these disorders. 29 Cannabis use decreases pain and spasticity in people with (MS), decreases tremor, stiffness, and pain in people with Parkinson's disease, and improves quality of life in patients with (MS) by improving appetite and decreasing pain and muscle spasticity. ...
Article
Full-text available
Objetivo: apresentar o estado da arte das publicações expressas na literatura cientifica mundial sobre a temática, bem comoidentificar os benefícios terapêuticos da Cannabis medicinal no tratamento dos sintomas das doenças neurodegenerativasespecificamente doenças de Parkinson, esclerose múltipla e Alzheimer. Método: trata-se de uma revisão integrativa da literatura,cuja busca de dados foi realizada nas bibliotecas virtuais. Web of Science, Scopus, Medline, Lilacs, Cochrane Library e Scielo noperíodo de agosto a outubro de 2021. Resultados: foram encontrados 158 artigos. Vinte e tres artigos foram selecionadospara serem lidos na íntegra e 8 atenderam aos critérios desta revisão. Conclusão: as evidências mostram que embora cada vezmais prescritos ou autorizados, a cannabis medicinal ou os Canabinóides para a doenças neurodegenerativas continuam a sercontroversos para muitos médicos
... Their structural complexity and diversity often provide a wide range of bioactivities that can be harnessed as therapeutic agents for various diseases and indications. Cannabinoids are one such class of natural products originally isolated from the plant Cannabis sativa that offers a wide range of bioactivities such as anti-bacterial [2], anti-epileptic [3] and anti-tumor effects [4]. ...
Article
Full-text available
Natural products make up a large proportion of medicine available today. Cannabinoids from the plant Cannabis sativa is one unique class of meroterpenoids that have shown a wide range of bioactivities and recently seen significant developments in their status as therapeutic agents for various indications. Their complex chemical structures make it difficult to chemically synthesize them in efficient yields. Synthetic biology has presented a solution to this through metabolic engineering in heterologous hosts. Through genetic manipulation, rare phytocannabinoids that are produced in low yields in the plant can now be synthesized in larger quantities for therapeutic and commercial use. Additionally, an exciting avenue of exploring new chemical spaces is made available as novel derivatized compounds can be produced and investigated for their bioactivities. In this review, we summarized the biosynthetic pathways of phytocannabinoids and synthetic biology efforts in producing them in heterologous hosts. Detailed mechanistic insights are discussed in each part of the pathway in order to explore strategies for creating novel cannabinoids. Lastly, we discussed studies conducted on biological targets such as CB1, CB2 and orphan receptors along with their affinities to these cannabinoid ligands with a view to inform upstream diversification efforts.
... Activation of CB2R with HU-308 alleviated clinical signs of EAE through activation of autophagy and restoring the reduced LC3-II/LC3-I ratio [127]. Cannabis, which targets CB2R, is used to treat MS and has been reported to reduce disease progression via autophagic-related pathways [153]. Additionally, cAMP-dependent PKA inhibitor H89 was found to induce autophagy through downstream modulation of Akt [126]. ...
Article
Full-text available
Autophagy is a constitutive process that degrades, recycles and clears damaged proteins or organelles, yet, despite activation of this pathway abnormal proteins accumulate in neurons in neurodegenerative diseases, and in oligodendrocytes in white matter disorders. Here, we discuss the role of autophagy in white matter disorders including neurotropic infections, inflammatory diseases such as multiple sclerosis, and in hereditary metabolic disorders and acquired toxic‐metabolic disorders. Once triggered due to cell stress, autophagy can enhance cell survival or cell death that may contribute to oligodendrocyte damage and myelin loss in white matter diseases. For some disorders, the mechanisms leading to myelin loss are clear while the aetiological agent and pathological mechanisms are unknown for other myelin disorders, although emerging studies indicate that a common mechanism underlying these disorders is dysregulation of autophagic pathways. In this review we discuss the alterations in the autophagic process in white matter disorders and the potential use of autophagy modulating agents as therapeutic approaches in these pathological conditions. This article is protected by copyright. All rights reserved.
Article
Full-text available
Cannabis belongs to the family Cannabaceae, and phytocannabinoids are produced by the Cannabis sativa L. plant. A long-standing debate regarding the plant is whether it contains one or more species. Phytocannabinoids are bioactive natural products found in flowers, seeds, and fruits. They can be beneficial for treating human diseases (such as multiple sclerosis, neurodegenerative diseases, epilepsy, and pain), the cellular metabolic process, and regulating biological function systems. In addition, several phytocannabinoids are used in various therapeutic and pharmaceutical applications. This study provides an overview of the different sources of phytocannabinoids; further, the biosynthesis of bioactive compounds involving various pathways is elucidated. The structural classification of phytocannabinoids is based on their decorated resorcinol core and the bioactivities of naturally occurring cannabinoids. Furthermore, phytocannabinoids have been studied in terms of their role in animal models and antimicrobial activity against bacteria and fungi; further, they show potential for therapeutic applications and are used in treating various human diseases. Overall, this review can help deepen the current understanding of the role of biotechnological approaches and the importance of phytocannabinoids in different industrial applications.
Chapter
Multiple sclerosis (MS) is an autoimmune disease typified by overt demyelination and inflammation that develop in selected regions of the central nervous system (CNS). Besides these signs, a diffuse loss of synaptic contacts, axonal pruning and astrocytosis are also observed, that in general correlate with the dysregulation of the glutamatergic system and with the onset of neurological symptoms. Concomitantly to the synaptic derangements, impaired glutamate homeostasis also dysregulates the immunocompetent responses, impairing the functional cross-talk between the immune system and the CNS. The study of the glutamatergic system therefore emerges as an important issue for deciphering the cellular events at the basis of MS as it would permit the proposal of new appropriate pharmacological interventions for the cure of the pathology. The chapter describes recent advances in basic research, preclinical and clinical studies concerning the impact of altered glutamate homeostasis in the course of the disease, as well as in the innovative strategies that would permit the restoration of central glutamatergic transmission.
Article
Full-text available
Objective: To test the effectiveness and long term safety of cannabinoids in multiple sclerosis (MS), in a follow up to the main Cannabinoids in Multiple Sclerosis (CAMS) study. Methods: In total, 630 patients with stable MS with muscle spasticity from 33 UK centres were randomised to receive oral D9-tetrahydrocannabinol (D9-THC), cannabis extract, or placebo in the main 15 week CAMS study. The primary outcome was change in the Ashworth spasticity scale. Secondary outcomes were the Rivermead Mobility Index, timed 10 metre walk, UK Neurological Disability Score, postal Barthel Index, General Health Questionnaire-30, and a series of nine category rating scales. Following the main study, patients were invited to continue medication, double blinded, for up to12 months in the follow up study reported here. Results: Intention to treat analysis of data from the 80% of patients followed up for 12 months showed evidence of a small treatment effect on muscle spasticity as measured by change in Ashworth score from baseline to 12 months (D9-THC mean reduction 1?82 (n=154, 95% confidence interval (CI) 0.53 to 3.12), cannabis extract 0.10 (n=172, 95% CI 20.99 to 1.19), placebo 20.23 (n=176, 95% CI 21.41 to 0.94); p=0.04 unadjusted for ambulatory status and centre, p=0.01 adjusted). There was suggestive evidence for treatment effects of D9-THC on some aspects of disability. There were no major safety concerns. Overall, patients felt that these drugs were helpful in treating their disease. Conclusions: These data provide limited evidence for a longer term treatment effect of cannabinoids. A long term placebo controlled study is now needed to establish whether cannabinoids may have a role beyond symptom amelioration in MS.
Article
Full-text available
Cannabinoids affect immune responses in ways that may be beneficial for autoimmune diseases. We sought to determine whether chronic Cannabis use differentially modulates a select number of immune parameters in healthy controls and individuals with multiple sclerosis (MS cases). Subjects were enrolled and consented to a single blood draw, matched for age and BMI. We measured monocyte migration isolated from each subject, as well as plasma levels of endocannabinoids and cytokines. Cases met definition of MS by international diagnostic criteria. Monocyte cell migration measured in control subjects and individuals with MS was similarly inhibited by a set ratio of phytocannabinoids. The plasma levels of CCL2 and IL17 were reduced in non-naïve cannabis users irrespective of the cohorts. We detected a significant increase in the endocannabinoid arachidonoylethanolamine (AEA) in serum from individuals with MS compared to control subjects, and no significant difference in levels of other endocannabinoids and signaling lipids irrespective of Cannabis use. Chronic Cannabis use may affect the immune response to similar extent in individuals with MS and control subjects through the ability of phytocannabinoids to reduce both monocyte migration and cytokine levels in serum. From a panel of signaling lipids, only the levels of AEA are increased in individuals with MS, irrespective of Cannabis use or not. Our results suggest that both MS cases and controls respond similarly to chronic Cannabis use with respect to the immune parameters measured in this study.
Article
Laboratory evidence has shown that cannabinoids might have a neuroprotective action. We investigated whether oral dronabinol (Δ(9)-tetrahydrocannabinol) might slow the course of progressive multiple sclerosis.
Article
Multiple sclerosis is primarily an inflammatory disorder of the brain and spinal cord in which focal lymphocytic infiltration leads to damage of myelin and axons. Initially, inflammation is transient and remyelination occurs but is not durable. Hence, the early course of disease is characterised by episodes of neurological dysfunction that usually recover. However, over time the pathological changes become dominated by widespread microglial activation associated with extensive and chronic neurodegeneration, the clinical correlate of which is progressive accumulation of disability. Paraclinical investigations show abnormalities that indicate the distribution of inflammatory lesions and axonal loss (MRI); interference of conduction in previously myelinated pathways (evoked electrophysiological potentials); and intrathecal synthesis of oligoclonal antibody (examination by lumbar puncture of the cerebrospinal fluid). Multiple sclerosis is triggered by environmental factors in individuals with complex genetic-risk profiles. Licensed disease modifying agents reduce the frequency of new episodes but do not reverse fixed deficits and have questionable effects on the long-term accumulation of disability and disease progression. We anticipate that future studies in multiple sclerosis will provide a new taxonomy on the basis of mechanisms rather than clinical empiricism, and so inform strategies for improved treatment at all stages of the disease.
Article
The inhibition of monoacylglycerol lipase (MAGL) the enzyme responsible for the inactivation of the endocannabinoid 2-arachidonoylglycerol, is a strategy for the treatment of multiple sclerosis. In their Communication on page 13765 ff., M. L. López-Rodríguez and co-workers describe a potent, reversible, and selective inhibitor of MAGL that slows the clinical progression of multiple sclerosis without inducing undesirable CB1-mediated side effect.
Article
Traumatic brain injury (TBI) is associated with significant morbidity and mortality. Several studies have demonstrated neuroprotective effects of cannabinoids. The objective of this study was to establish a relationship between the presence of a positive toxicology screen for tetrahydrocannabinol (THC) and mortality after TBI. A 3-year retrospective review of registry data at a Level I center of patients sustaining TBI having a toxicology screen was performed. Pediatric patients (younger than 15 years) and patients with a suspected nonsurvivable injury were excluded. The THC(+) group was compared with the THC(-) group with respect to injury mechanism, severity, disposition, and mortality. Logistic regression was used to determine independent associations with mortality. There were 446 cases meeting all inclusion criteria. The incidence of a positive THC screen was 18.4 per cent (82). Overall mortality was 9.9 per cent (44); however, mortality in the THC(+) group (2.4% [two]) was significantly decreased compared with the THC(-) group (11.5% [42]; P = 0.012). After adjusting for differences between the study cohorts on logistic regression, a THC(+) screen was independently associated with survival after TBI (odds ratio, 0.224; 95% confidence interval, 0.051 to 0.991; P = 0.049). A positive THC screen is associated with decreased mortality in adult patients sustaining TBI.
Article
Monoacylglycerol lipase (MAGL) is the enzyme responsible for the inactivation of the endocannabinoid 2-arachidonoylglycerol (2-AG). MAGL inhibitors show analgesic and tissue-protecting effects in several disease models. However, the few efficient and selective MAGL inhibitors described to date block the enzyme irreversibly, and this can lead to pharmacological tolerance. Hence, additional classes of MAGL inhibitors are needed to validate this enzyme as a therapeutic target. Here we report a potent, selective, and reversible MAGL inhibitor (IC50=0.18 μM) which is active in vivo and ameliorates the clinical progression of a multiple sclerosis (MS) mouse model without inducing undesirable CB1-mediated side effects. These results support the interest in MAGL as a target for the treatment of MS.
Article
The endocannabinoids 2-araquidonoylglycerol (2-AG) and anandamide (AEA) are bioactive lipids crucially involved in the regulation of brain function in basal and pathological conditions. Blockade of endocannabinoid metabolism has emerged as a promising therapeutic strategy for inflammatory diseases of the central nervous system, including myelin disorders such as multiple sclerosis. Nevertheless, the biological actions of endocannabinoid degradation inhibitors in oligodendrocytes and white matter tracts are still ill defined. Here we show that the selective monoacylglycerol lipase (MAGL) inhibitor JZL184 suppressed cell death by mild activation of AMPA receptors in oligodendrocytes in vitro, an effect that was mimicked by MAGL substrate 2-AG and by the second major endocannabinoid AEA, in a concentration-dependent manner, whereas inhibition of the AEA metabolizing enzyme fatty acid amide hydrolase with URB597 was devoid of effect. Pharmacological experiments suggested that oligodendrocyte protection from excitotoxicity resulting from MAGL blockade involved the activation of cannabinoid CB1 receptors and the reduction of AMPA-induced cytosolic calcium overload, mitochondrial membrane depolarization, and production of reactive oxygen species. Administration of JZL184 under a therapeutic regimen decreased clinical severity, prevented demyelination, and reduced inflammation in chronic experimental autoimmune encephalomyelitis. Furthermore, MAGL inactivation robustly preserved myelin integrity and suppressed microglial activation in the cuprizone-induced model of T-cell-independent demyelination. These findings suggest that MAGL blockade may be a useful strategy for the treatment of immune-dependent and -independent damage to the white matter. GLIA 2015;63:163–176