ArticlePDF Available

ALSUntangled No. 16: Cannabis

Authors:
Amyotrophic Lateral Sclerosis, 2012; 13: 400 404
ISSN 1748-296 8 print /I SSN 1471-180X online © 2012 In form a Health care
DOI : 10. 3109 /17482 968 .2 012.6 8726 4
about cannabinol, although it appears to have distinct
pharmacological properties from cannabidiol. Can-
nabinol has anticonvulsant, sedative, and other phar-
macological activities likely to interact with the effects
of THC (19–22). Cannabinol may induce sleep and
may provide some protection against seizures for
epileptics (22).
Cannabis Receptors and Endogenous Ligands
Recent advances have increased understanding of
the receptors and endogenous ligands composing
the cannabinoid system (23–36). Two major can-
nabinoid receptor subtypes exist: CB1 is predomi-
nantly expressed in the brain, and CB2 is primarily
found on the cells of the immune system (23,37–39).
Both receptors are G protein-coupled, 7-segment
transmembrane proteins, similar to the receptors of
dopamine, serotonin, and norepinephrine (39,40).
Dense cannabinoid receptor concentrations are
found in the cerebellum, basal ganglia, and hip-
pocampus, likely accounting for the effect of exog-
enously administered cannabinoids on motor tone
and coordination as well as mood state (41–43). Low
concentrations are found in the brainstem, perhaps
accounting for the low potential for lethal overdose
with cannabinoid-based medicines (44–47).
The discovery of endocannabinoids, (i.e. endog-
enous metabolites capable of activating the cannab-
inoid receptors), and an improved understanding of
the molecular mechanisms leading to their biosynthe-
sis, release, and inactivation, have inspired research
on the pharmaceutical applications of cannabinoid-
based medicines (40). A growing number of strate-
gies for separating sought after therapeutic effects of
cannabinoid receptor agonists from the unwanted
consequences of CB1 receptor activation are emerging.
Ligands have been developed that are potent and
selective agonists for CB1 and CB2 receptors, potent
CB1 selective antagonists, and inhibitors of endo-
cannabinoid uptake or metabolism (48). Distinct
varieties of cannabis contain different combinations
of partial cannabinoid agonists and antagonists, which
could be utilized in designing synthetic cannabinoid
Introduction
In a widely viewed series of Internet videos called
Surviving ALS, Cathy Jordan reports that regu-
larly smoking cannabis has dramatically slowed her
ALS progression, and improved her mood and appe-
tite (see for example http://www.youtube.com/
watch?v -kf8wTBiUDU). Indeed, cannabinoids
and manipulation of the endocannabinoid system
may well have disease-modifying potential in ALS
(1–9). Moreover, cannabis could potentially be use-
ful in managing the symptomatology in ALS (10–13).
Here, on behalf of PALS who are asking about it, we
critically review the evidence for cannabis in ALS.
What Is Cannabis?
Cannabis is a remarkably complex plant. There are
several existing phenotypes, with each containing over
400 distinct chemical moieties (14–22). Approximately
70 are chemically unique and classifi ed as plant
cannabinoids (14–17). Cannabinoids are lipophilic
21 carbon terpenes, biosynthesized predominantly via
a recently discovered deoxyxylulose phosphate path-
way (14). Delta-9 tetrahydrocannabinol (THC) and
delta-8 THC appear to produce the majority of the
psychoactive effects of cannabis (18,19). Delta-9
THC, the active ingredient in dronabinol (Marinol)
is the most abundant cannabinoid in the plant and
this has led researchers to hypothesize that it is the
main source of the drug s impact. However, other
major plant cannabinoids, including cannabidiol and
cannabinol, may modify the pharmacology of THC
and have distinct effects of their own. Cannabidiol is
the second most prevalent of cannabis s active ingre-
dients and may produce most of its effects at moder-
ate, mid range doses. Cannabidiol becomes THC as
the plant matures and THC over time breaks down
into cannabinol. Up to 40% of the cannabis resin in
some strains is cannabidiol (16). The amount varies
according to plant. Some varieties of Cannabis sativa
have been found to have no cannabidiol (16). Can-
nabidiol appears to modulate and reduce untoward
effects of THC (18). Cannabidiol breaks down to
cannabinol as the plant matures. Much less is known
ALSUntangled No. 16: Cannabis
The ALSUntangled Group
Amyotroph Lateral Scler Downloaded from informahealthcare.com by Dr. Josep Gamez on 08/23/12
For personal use only.
ALSUntangled Update 16 401
agonists and antagonists as well as cannabis strains
with high therapeutic potential.
Why Might Cannabis Work in ALS?
The fact that CB2 receptors have been found on
immune cells suggests that cannabinoids play a role in
the regulation of the immune system. Indeed, recent
studies show that cannabinoids can down regulate
cytokine and chemokine production, which in turn
suppresses infl ammatory responses (49–52). Since the
pathophysiology of ALS may involve neuroinfl amma-
tion (53,54), agents such as cannabinoids that modu-
late this process could potentially be useful.
Alternatively, or in addition, cannabinoids might
act similarly to tamoxifen, a Food and Drug Admin-
istration (FDA)-approved drug used to treat breast
cancer (55–57). Both cannabinoids and tamoxifen
are terpenes, organic, lipid soluble compounds that
readily penetrate the CNS (55). In a 60 patient pilot
study, PALS taking tamoxifen reportedly had
improved survival and no signifi cant side effects
(56); unfortunately this study has yet to be pub-
lished so it cannot be peer reviewed. A follow up
study is underway (http://www.clinicaltrials.gov/ct2/
show/NCT01257581? term tamoxifen als&rank
1). Tamoxifen may affect ALS by modulating
infl ammation, or by altering glutamate uptake (55).
Endocannabinoids can also modulate glutamatergic
neurotransmission indirectly via NMDA receptors
(33,34,36).
What Relevant Animal Data Exists in ALS?
Beyond the theoretical, observations in mice support
the idea that the endocannabinoid system might be
involved in the pathophysiology of ALS. Endogenous
cannabinoids are elevated in spinal cords of symptom-
atic G93A-SOD1 mice (9). mRNA levels, receptor
binding, and function of CB2, but not CB1, receptors
are dramatically and selectively up-regulated in spinal
cords of G93A-SOD1 mice in a temporal pattern par-
alleling disease progression (2). The sensitivity of CB1
receptors in controlling both glutamate and GABA
transmission is potentiated in the striatum of symp-
tomatic G93A-SOD1 ALS mice (5).
Other animal studies suggest that treatment
with cannabinoids could be useful. Treatment with
Delta(9)-THC at onset of tremors delayed motor
impairment and prolonged survival in G93A-SOD1
mice (58). Daily injections of the selective CB2 ago-
nist AM-1241, initiated at symptom onset in G93A-
SOD1 ALS mice, increased survival after disease
onset by 56% (2). Treatment of post symptomatic,
90-day-old G93A-SOD1 mice with a synthetic
cannabinoid, WIN55,212–2, improved motoneuron
survival and muscle force at 120d although this did
not improve overall survival (6). Genetic ablation of
the (Fatty Acid Amide Hydrolase) FAAH enzyme,
which results in raised levels of the endocannabinoid
anandamide by preventing its breakdown, delayed
disease onset in G93A-SOD1 ALS mice but did not
affect survival (6). Ablation of the CB1 receptor had
no effect on disease onset in G93A-SOD1 ALS mice
but signifi cantly extended life span. These animal
studies all have signifi cant methodological fl aws,
including small sample size, lack of randomization
and lack of blinding.
What Are the Effi cacy, Safety and Costs of
Cannabis in Human ALS?
A small randomized, double-blind placebo-controlled
crossover trial of oral THC at 5mg twice daily was
conducted in PALS, with the goal of improving
cramps. (59). While this was well-tolerated, there was
no effect on cramp frequency or intensity, or on sec-
ondary outcome measures including fasciculation
frequency, quality of life, sleep or depression. This
27 patient trial may well have been underpowered for
some of these outcomes. A small trial of dronabinol
in PALS was previously published as an abstract and
indicated good tolerability (11). However Dronabi-
nol is 100% Delta-9 THC, the most psychoactive
ingredient in cannabis. Natural cannabis contains, at
best, 20% THC. There are varying physiological
effects when the other cannabinoid forms are present,
as is the case with natural cannabis plant material.
Moreover, while glutamate toxicity is reduced by
both CBD (cannabadiol - nonpsychoactive), and
THC, the neuroprotection observed with CBD
appears greater than THC (60–62). Most patients
nd dronabinol too sedating and associated with too
many psychoactive effects (63–66). Dronabinol is not
an appropriate substitute for cannabis in this setting.
Within the PatientsLikeMe online community,
48 members with ALS reported taking cannabis in
a variety of forms, durations and dosages. Benefi ts
described included improved speech, swallowing,
secretions, fasciculations, appetite, sleep and mood.
Side effects included dry mouth, clumsiness, dizzi-
ness, pneumothorax and sore throat.
What Would be Needed to Test Cannabis in
Human ALS Clinical Trials?
Doing multi-center clinical research trials for PALS
using cannabis would pose many unique barriers.
First of all there is no commercial manufacturer of
cannabis, thus these studies would have to be funded
either by the federal government or privately, as it
is not likely there would be industry funding. Obtain-
ing the trial drug would require the investigators to
gain access to a large, reliable supply of cannabis
that is legal for medical research. At present, the
only source of cannabis that can be legally used in
research in the United States is through the National
Institute on Drug Abuse (NIDA). Unfortunately
NIDA provides low-potency material, and makes
the cannabis available only to projects it approves.
Amyotroph Lateral Scler Downloaded from informahealthcare.com by Dr. Josep Gamez on 08/23/12
For personal use only.
402 The ALSUntangled Group
NIDA supplies cannabis with a THC content, by
weight, of 2–4% typically, although it has supplied
cannabis with an 8% by weight THC content on
occasion (67). Although THC is not the ideal target
compound per se, it is a relative indicator of potency
and quality. For comparison, the average THC con-
tent of cannabis at randomly surveyed medical
cooperatives in California is approximately 15 to
20% (68). Thus, an independent source of cannabis
would be needed to ensure a consistently high can-
nabinoid content that may be strong enough to pos-
sibly alter the disease progression. An independent
cannabis source would also allow investigators to
avoid NIDA s arbitrary and lengthy review process
that it mandates before providing any cannabis for
research. Historically NIDA has derailed clinical
trial plans by refusing to supply cannabis, even after
the research protocols were approved by the FDA
(69,70). Nonetheless, it is possible, with coordi-
nated effort, to effectively do double-blind, random-
ized, placebo controlled clinical trials with cannabis.
To properly evaluate both subjective and objective
effects, cannabinoid blood levels should be followed
as well, to further ensure adequate data for a
dose-response curve. Mode of drug delivery could
be via vaporization, which would allow for dosing
standardization (71,72).
Another interim option would be clinical trials
with Sativex ® , a product from GW Pharmaceutical
company in the UK. Sativex is a natural cannabinoid
pharmaceutical product, administered as an oral
spray absorbed by the patient s mouth. The drug is
obtained from natural cannabis and standardized by
weight to be 50% THC and 50% cannababidiol
(CBD)(73). This makes it a much better choice than
Marinol (dronabinol). Yet this is not as desirable as
natural cannabis, which contains a multitude of other
therapeutic cannabinoids, many of which are not psy-
choactive, such as cannabinol (CBN). One of the
ALSUntangled team (GTC) has tried to get GW to
pursue this but has not had success to date, with the
company decision being based on fi nancial informa-
tion. GW Pharma openly acknowledges that it needs
a large patient base to be fi nancially viable and is thus
targeting multiple sclerosis (MS). Finally Sativex is
not available as of yet in the United States.
Conclusions
Cannabis has biological properties including immu-
nomodulation and effects on excitototoxicity that
suggest it could be useful in ALS. Evidence from
small, non-randomized, unblinded animal studies
suggest that it could potentially slow ALS progres-
sion, and anecdotal reports suggest that it could
ameliorate troubling ALS symptoms. Given all this,
ALSUntangled supports further careful study of
cannabis and cannabinoids, the active ingredients
contained therein. Natural cannabis, as a single
agent, provides advantages similar to a multiple drug
trial given its numerous mechanisms of action. A
possible next step would be a small case series of
well-characterized PALS using cannabis at con-
trolled dosages that could potentially be monitored
by blood levels of cannabinoids, compared to
matched controls, performed in a geographic area
where it would be legal.
The ALSUntangled Group currently consists of
the following members: Gregory Carter, Richard Bed-
lack, Orla Hardiman, Lyle Ostrow, Edor Kabashi,
Tulio Bertorini, Tahseen Mozaffar, Peter Andersen,
Jeff Dietz, Josep Gamez, Mazen Dimachkie, Yunxia
Wang, Paul Wicks, James Heywood, Steven Novella,
LP Rowland, Erik Pioro, Lisa Kinsley, Kathy Mitch-
ell, Jonathan Glass, Sith Sathornsumetee, Hubert
Kwiecinski, Jon Baker, Nazem Atassi, Dallas Forshew,
John Ravits, Robin Conwit, Carlayne Jackson, Alex
Sherman, Kate Dalton, Katherine Tindall, Ginna
Gonzalez, Janice Robertson, Larry Phillips, Michael
Benatar, Eric Sorenson, Christen Shoesmith, Steven
Nash, Nicholas Marigakis, Dan Moore, James
Caress, Kevin Boylan, Carmel Armon, Megan
Grosso, Bonnie Gerecke, Jim Wymer, Bjorn Oskars-
son, Robert Bowser, Vivian Drory, Jeremy Shefner,
Terry Heiman-Patterson, Noah Lechtzin, Melanie
Leitner, Robert Miller, Hiroshi Mitsumoto, Todd
Levine, James Russell, Khema Sharma, David Saper-
stein, Leo McClusky, Daniel MacGowan, Jonathan
Licht, Ashok Verma, Michael Strong, Catherine
Lomen-Hoerth, Rup Tandan, Michael Rivner, Steve
Kolb, Meraida Polak, Stacy Rudnicki, Pamela
Kittrell, Muddasir Quereshi, George Sachs, Gary
Pattee, Michael Weiss, John Kissel, Jonathan Gold-
stein, Jeffrey Rothstein, Dan Pastula. Note: this paper
represents a consensus of those weighing in. The
opinions expressed in this paper are not necessarily
shared by every investigator in this group.
Disclosures: ALSUntangled is sponsored by the
Packard Center and the Motor Neurone Disease
Association.
References
Kim K, Moore DH, Makriyannis A, Abood ME. AM1241, 1.
a cannabinoid CB2 receptor selective compound, delays dis-
ease progression in a mouse model of amyotrophic lateral
sclerosis. Eur J Pharmacol 2006; 542(1–3):100–5.
Shoemaker JL, Seely KA, Reed RL, Crow JP, Prather PL. 2.
The CB2 cannabinoid agonist AM-1241 prolongs survival
in a transgenic mouse model of amyotrophic lateral sclerosis
when initiated at symptom onset. J Neurochem 2007;
101(1):87–98.
Raman C, McAllister SD, Rizvi G, Patel SG, Moore DH, 3.
Abood ME. Amyotrophic lateral sclerosis: delayed disease
progression in mice by treatment with a cannabinoid. Amyo-
troph Lateral Scler Other Motor Neuron Disord 2004;
5(1):33–9.
Weydt P, Hong S, Witting A, M ö ller T, Stella N, Kliot M. 4.
Cannabinol delays symptom onset in SOD1 (G93A) trans-
genic mice without affecting survival. Amyotroph Lateral
Scler Other Motor Neuron Disord 2005; 6(3):182–4.
Amyotroph Lateral Scler Downloaded from informahealthcare.com by Dr. Josep Gamez on 08/23/12
For personal use only.
ALSUntangled Update 16 403
Rossi S, De Chiara V, Musella A, Cozzolino M, Bernardi G, 5.
Maccarrone M, Mercuri NB, Carri MT, Centonze D.
Abnormal sensitivity of cannabinoid CB1 receptors in the
striatum of mice with experimental amyotrophic lateral
sclerosis. Amyotroph Lateral Scler 2009; 19:1–8.
Bilsland LG, Dick JR, Pryce G, Petrosino S, Di Marzo V, 6.
Baker D, Greensmith L. Increasing cannabinoid levels by
pharmacological and genetic manipulation delay disease
progression in SOD1 mice. FASEB J 2006; 20(7):1003–5.
Bilsland LG, Greensmith L. The endocannabinoid system in 7.
amyotrophic lateral sclerosis. Curr Pharm Des 2008; 14(23):
2306–16.
Zhao P, Ignacio S, Beattie EC, Abood ME. Altered pre-8.
symptomatic AMPA and cannabinoid receptor traffi cking
in motor neurons of ALS model mice: implications for
excitotoxicity. Eur J Neurosci 2008; 27(3):572–9.
Witting A, Weydt P, Hong S, Kliot M, Moller T, Stella N. 9.
Endocannabinoids accumulate in spinal cord of SOD1
transgenic mice. J Neurochem. 2004;89:1555 7.
Carter GT, BS Rosen. Marijuana in the management of 10.
amyotrophic lateral sclerosis. Am J Hosp Palliat Care 2001;
18(4):264–70.
Gelinas D, Miller RG, Abood ME. A pilot study of safety 11.
and tolerability of Delta 9-THC (Marinol) treatment for
ALS. Amyotroph Lateral Scler Other Motor Neuron Disord.
2002;3(Suppl. 1):23.
Amtmann D, Weydt P, Johnson KL, Jensen MP, Carter GT. 12.
Survey of cannabis use in patients with amyotrophic lateral
sclerosis. Am J Hosp Palliat Care. 2004;21:95 104.
Carter GT, Weydt P. Cannabis: old medicine with new prom-13.
ise for neurological disorders. Curr Opin Investig Drugs
2002; 3(3):437 440
Gerra G, Zaimovic A, Gerra ML, Ciccocioppo R, Cippitelli 14.
A, Serpelloni G, Somaini L. Pharmacology and toxicology
of Cannabis derivatives and endocannabinoid agonists.
Recent Pat CNS Drug Discov 2010; 5(1):46 52.
Fischedick JT, Glas R, Hazekamp A, Verpoorte R. A qualita-15.
tive and quantitative HPTLC densitometry method for the
analysis of cannabinoids in Cannabis sativa L. Phytochem
Anal 2009; 20(5):421 6.
Izzo AA, Borrelli F, Capasso R, Di Marzo V, Mechoulam R. 16.
Non-psychotropic plant cannabinoids: new therapeutic
opportunities from an ancient herb. Trends Pharmacol Sci
2009; 30(10):515 27.
Hill AJ, Williams CM, Whalley BJ, Stephens GJ. Phytocan-17.
nabinoids as novel therapeutic agents in CNS disorders.
Pharmacol Ther 2012;133:79-97.
Akinshola BE; Chakrabarti A; Onaivi ES. In vitro and in 18.
vivo action of cannabinoids. Neurochem Res 1999; 24(10):
1233–40.
Agurell S, Halldin M, Lindgren JE, Ohlsson A, Widman M, 19.
Gillespie H, and Hollister, L. Phar macokinetics and metab-
olism of delta 1 tetrahydrocannabinol and other cannabinoids
with emphasis on man. Pharmacol Rev 1986; 38(1):21–43.
Borrelli F, Aviello G, Romano B, Orlando P, Capasso R, 20.
Maiello F, Guadagno F, Petrosino S, Capasso F, Di Marzo V,
Izzo AA. Cannabidiol, a safe and non-psychotropic ing redi-
ent of the marijuana plant Cannabis sativa, is protective in a
murine model of colitis. J Mol Med 2009; 87(11):1111 21.
Zanelati TV, Biojone C, Moreira FA, Guimar ã es FS, Joca SR. 21.
Antidepressant-like effects of cannabidiol in mice: possible
involvement of 5-HT1A receptors. Br J Pharmacol 2010;
159(1):122 8.
Jones NA, Hill AJ, Smith I, Bevan SA, Williams CM, Whal-22.
ley BJ, Stephens GJ. Cannabidiol displays antiepileptiform
and antiseizure properties in vitro and in vivo. J Pharmacol
Exp Ther 2010;332(2):569 77.
Abood ME. Molecular biology of cannabinoid receptors. 23.
Handb Exp Pharmacol 2005; 168:81 115.
McAllister SD, Hurst DP, Barnett-Norris J, Lynch D, Reggio 24.
PH, Abood ME, Structural mimicry in class AG protein-
coupled receptor rotamer toggle switches: the importance of
the F3.36(201)/W6.48(357) interaction in cannabinoid CB1
receptor activation. J Biol Chem 2004; 279(46):48024 37.
McAllister SD, Chan C, Taft RJ, Luu T, Abood ME, 25.
Moore DH, Aldape K, Yount G, Cannabinoids selectively
inhibit proliferation and induce death of cultured human glio-
blastoma multiforme cells. J Neurooncol 2005; 74(1):31 40
Kapur A, Samaniego P, Thakur GA, Makriyannis A, 26.
Abood ME. Mapping the structural requirements in the
CB1 cannabinoid receptor transmembrane helix II for signal
transduction. J Pharmacol Exp Ther 2008; 325(1):341 8.
Gehani NC, Nalwalk JW, Razdan RK, Martin BR, Sun X, 27.
Wentland M, Abood ME, Hough LB. Signifi cance of can-
nabinoid CB1 receptors in improgan antinociception. J Pain
2007; 8(11):850 60.
Anavi-Goffer S, Fleischer D, Hurst DP, Lynch DL, Barnett-28.
Norris J, Shi S, Lewis DL, Mukhopadhyay S, Howlett AC,
Reggio PH, Abood ME, Helix 8 Leu in the CB1 cannabinoid
receptor contributes to selective signal transduction mecha-
nisms. J Biol Chem 2007; 282(34):25100 13.
McAllister SD, Rizvi G, Anavi-Goffer S, Hurst DP, Barnett-29.
Norris J, Lynch DL, Reggio PH, Abood ME, An aromatic
microdomain at the cannabinoid CB(1) receptor constitutes
an agonist/inverse agonist binding region. J Med Chem
2003; 46:24:5139 52.
McAllister SD, Tao Q, Barnett-Norris J, Buehner K, Hurst 30.
DP, Guarnieri F, Reggio PH, Nowell Harmon KW, Cabral
GA, Abood ME, A critical role for a tyrosine residue in the
cannabinoid receptors for ligand recognition. Biochem Phar-
macol 2002; 63(12):2121 36.
Abood ME, Rizvi G, Sallapudi N, McAllister SD, Activation 31.
of the CB1 cannabinoid receptor protects cultured mouse
spinal neurons against excitotoxicity. Neurosci Lett 2001;
309(3):197 201.
Panikashvili D, Simeonidou C, Ben Shabat S, Hanus L, 32.
Breuer A, Mechoulam R, Shohami E. An endogenous can-
nabinoid (2 AG) is neuroprotective after brain injury. Nature
2001; 4;413(6855):527–31.
Chen Y; Buck J. Cannabinoids protect cells from oxidative 33.
cell death: a receptor independent mechanism. J Pharmacol
Exp Ther 2000 Jun;293(3):807–12.
Eshhar N; Striem S; Biegon A. HU 211, a non psychotropic 34.
cannabinoid, rescues cortical neurones from excitatory amino
acid toxicity in culture. Neuroreport 1993 13;5(3):237–40.
Guzman M, Sanchez C, Galve-Roperth I. Control of the cell 35.
survival/death decision by cannabinoids. J Mol Med 2001;
78:613 625.
Maenpaa H, Mannerstrom M, Toimela T, Salminen L, 36.
Saransaari P, Tahti H. Glutamate uptake is inhibited by
tamoxifen and toremifene in cultured retinal pigment epi-
thelial cells. Pharmacol Toxicol 2002; 91(3):116 122
Klein TW; Lane B; Newton CA; Friedman H. The can-37.
nabinoid system and cytokine network. Proc Soc Exp Biol
Med 2000;225(1):1 8.
Pertwee RG: Cannabinoid receptor ligands: clinical and neu-38.
ropharmacological considerations, relevant to future drug
discovery and development. Expert Opin Investig Drugs
2000;9(7):1553 71.
Akinshola BE; Chakrabarti A; Onaivi ESIn vitro and in vivo 39.
action of cannabinoids. Neurochem Res 1999;24(10):1233–40
Di Marzo, Bisogno T, De Petrocellis L. Endocannabinoids: 40.
new targets for drug development. Curr Pharm Des 2000;
6(13):1361–80.
Hanu š LO, Mechoulam R. Novel Natural and Synthetic 41.
Ligands of the Endocannabinoid System. Curr Med Chem
2010; Feb 18. [Epub ahead of print].
De Laurentiis A, Fern á ndez Solari J, Mohn C, Zorrilla 42.
Zubilete M, Rettori V. Endocannabinoid system participates
in neuroendocrine control of homeostasis. Neuroimmu-
nomodulation 2010;17(3):153 6.
Nagarkatti P, Pandey R, Rieder SA, Hegde VL, Nagarkatti 43.
M. Cannabinoids as novel anti-infl ammatory drugs. Future
Med Chem 2009;1(7):1333 1349.
Amyotroph Lateral Scler Downloaded from informahealthcare.com by Dr. Josep Gamez on 08/23/12
For personal use only.
404 The ALSUntangled Group
Mouslech Z, Valla V. Endocannabinoid system: An overview 44.
of its potential in current medical practice. Neuro Endocri-
nol Lett 2009;30(2):153 79.
Wegener N, Koch M. Neurobiology and systems physiology 45.
of the endocannabinoid system. Pharmacopsychiatry
2009;42 Suppl 1:S79 86.
De Petrocellis L, Di Marzo V. An introduction to the endo-46.
cannabinoid system: from the early to the latest concepts.
Best Pract Res Clin Endocrinol Metab 2009;23(1):1 15.
Goodfellow CE, Glass M. Anandamide receptor signal 47.
transduction. Vitam Horm. 2009;81:79 110.
Pertwee RG. Ligands that target cannabinoid receptors in 48.
the brain: from THC to anandamide and beyond. Addict
Biol 2008;13(2):147 59.
Cencioni MT, Chiurchi ù V, Catanzaro G, Borsellino G, 49.
Bernardi G, Battistini L, Maccarrone M. Anandamide sup-
presses proliferation and cytokine release from primary
human T-lymphocytes mainly via CB2 receptors. PLoS One
2010; 5(1):e8688.
Pasquariello N, Catanzaro G, Marzano V, Amadio D, Bar-50.
caroli D, Oddi S, Federici G, Urbani A, Finazzi Agr ò A,
Maccarrone M. Characterization of the endocannabinoid
system in human neuronal cells and proteomic analysis of
anandamide-induced apoptosis. J Biol Chem 2009 23;284(43):
29413 26.
Pandey R, Mousawy K, Nagarkatti M, Nagarkatti P. Endo-51.
cannabinoids and immune regulation. Pharmacol Res 2009;
60(2):85 92.
Parolaro D, Massi P, Rubino T, Monti E. Endocannabinoids 52.
in the immune system and cancer. Prostaglandins Leukot
Essent Fatty Acids 2002; 66(2 3):319 32.
Papadimitriou D, Le Verche V, Jacqier A, Ikiz B, Przedborski 53.
S, Re DB. Infl ammation in ALS and SMA: sorting out the
good from the evil. Neurobiol Dise 2010:37:493 502.
Appel SH, Zhao W, Beers DR, Henkel JS. The microglial-54.
motoneuron dialogue in ALS. Acta Myol 2011;30:4 8.
Maenpaa H, Mannerstrom M, Toimela T, Salminen L, 55.
Saransaari P, Tahti H. Glutamate uptake is inhibited by
tamoxifen and toremifene in cultured retinal pigment epi-
thelial cells. Pharmacol Toxicol 2002; 91(3):116 122
Traynor BJ, Bruijn L, Conwit R, Beal F, O’Neill G, Fagan 56.
SC, Cudkowicz ME. Neuroprotective agents for clinical tri-
als in ALS: a systematic assessment. Neurology 2006; 11;
67(1):20 7.
Lee ES, Yin Z, Milatovic D, Jiang H, Aschner M. Estrogen 57.
and tamoxifen protect against Mn-induced toxicity in rat
cortical primary cultures of neurons and astrocytes. Toxicol
Sci 2009;110(1):156 67.
Raman C, McAllister SD, Rizvi G, Patel SG, Moore DH, 58.
Abood ME. Amyotrophic lateral sclerosis: delayed disease
progression in mice by treatment with a cannabinoid. Amyo-
troph Latearl Scler Other Motor Neuron Disor 2004;5:33 39.
Weber M, Goldman B, Truniger S. Tetrahydrocannabinol 59.
(THC) for cramps in amyotrophic lateral sclerosis: a rand-
omized, double-blind crossover trial. J Neurol Neurosurg
Psychiatry 2010;1135 40.
Yiangou Y, Facer P, Durrenberger P, Chessell IP, Naylor A, 60.
Bountra C, Banati RR, Anand P. COX-2, CB2 and P2X7-
immunoreactivities are increased in activated microglial
cells/macrophages of multiple sclerosis and amyotrophic lat-
eral sclerosis spinal cord. BMC Neurol 2006; 6:12.
Hampson AJ; Grimaldi M; Axelrod J; Wink D. Cannabidiol 61.
and( )Delta9 tetrahydrocannabinol are neuroprotective
antioxidants. Proc Natl Acad Sci U S A 95(14):8268 73,
1998.
Hampson AJ; Grimaldi M; Lolic M; Wink D; Rosenthal R; 62.
Axelrod J: Neuroprotective antioxidants from marijuana.
Ann N Y Acad Sci 899:274 82, 2000.
Nagayama T; Sinor AD; Simon RP; Chen J; Graham SH; 63.
Jin K; Greenberg DA. Cannabinoids and neuroprotection in
global and focal cerebral ischemia and in neuronal cultures.
J Neurosci 19(8):2987 95, 1999.
Russo EB. History of cannabis and its preparations in 64.
saga, science, and sobriquet. Chem Biodivers. 2007;4(8):
1614 48.
El Sohly MA, Wachtel SR, de Wit H. Cannabis versus THC: 65.
response to Russo and McPartland. Psychopharmacology
2003; 165(4):433 434.
Carter GT, Flanagan A, Earleywine M, Abrams DI, Aggar-66.
wal SK, Grinspoon L: Cannabis in palliative medicine:
improving care and reducing opioid-related morbidity. Am J
Hosp Palliat Med 2011; 28(5):297 303
Aggarwal SK, Carter GT, Sullivan MD, Morrill R, Zum-67.
Brunnen C, Mayer JD. Characteristics of patients with
chronic pain accessing treatment with medicinal cannabis in
Washington State. J Opioid Manag 2009; 5(5):257 286
Carter GT, Abood ME, Aggarwal SK, Weiss MD. Cannabis 68.
and amyotrophic lateral sclerosis: practical and hypothetical
applications, and a call for clinical trials. Am J Hosp Palliat
Med 2010; 27(5):347 56;
Carter GT, Weydt P, Kyashna-Tocha M, Abrams DI. 69.
Medicinal cannabis: rational guidelines for dosing. IDrugs
2004;7(5):464 70.
Aggarwal SK, Kyashna-Tocha M, Carter GT. Dosing Med-70.
ical Marijuana: Rational Guidelines on Trial in Washington
State. MedGenMed 2007; 9(3):52 3.
Abrams DI, Vizoso HP, Shade SB, Jay C, Kelly ME, 71.
Benowitz NL. Vaporization as a smokeless cannabis delivery
system: a pilot study. Clin Pharmacol Ther 2007; 82(5):
572 8.
Pomahacova B, Van der Kooy F, Verpoorte R. Cannabis 72.
smoke condensate III: The cannabinoid content of vaporised
Cannabis sativa. Inhal Toxicol 2009;21(13):1108 12.
Kmietowicz Z. Cannabis based drug is licensed for spasticity 73.
in patients with MS. BMJ 2010;340:c3363.
The ALSUntangled Group currently consists of the
following members: Richard Bedlack, Orla Hardiman,
Tulio Bertorini, Tahseen Mozaffar, Peter Andersen,
Jeff Dietz, Josep Gamez, Mazen Dimachkie, Yunxia
Wang, Paul Wicks, James Heywood, Steven Novella,
LP Rowland, Erik Pioro, Lisa Kinsley, Kathy
Mitchell, Jonathan Glass, Sith Sathornsumetee,
Hubert Kwiecinski, Jon Baker, Nazem Atassi, Dallas
Forshew, John Ravits, Robin Conwit, Carlayne
Jackson, Alex Sherman, Kate Dalton, Katherine
Tindall, Ginna Gonzalez, Janice Robertson, Larry
Phillips, Michael Benatar, Eric Sorenson, Christen
Shoesmith, Steven Nash, Nicholas Marigakis, Dan
Moore, James Caress, Kevin Boylan, Carmel Armon,
Megan Grosso, Bonnie Gerecke, Jim Wymer, Bjorn
Oskarsson, Robert Bowser, Vivian Drory, Jeremy
Shefner, Terry Heiman-Patterson, Noah Lechtzin,
Melanie Leitner, Robert Miller, Hiroshi Mitsumoto,
Todd Levine, James Russell, Khema Sharma, David
Saperstein, Leo McClusky, Daniel MacGowan,
Jonathan Licht, Ashok Verma, Michael Strong,
Catherine Lomen-Hoerth, Rup Tandan, Michael
Rivner, Steve Kolb, Meraida Polak, Stacy Rudnicki,
Pamela Kittrell, Muddasir Quereshi, George Sachs,
Gary Pattee, Michael Weiss, John Kissel, Jonathan
Goldstein, Jeffrey Rothstein, Dan Pastula. Note: this
paper represents a consensus of those weighing in.
The opinions expressed in this paper are not
necessarily shared by every investigator in this
group.
Amyotroph Lateral Scler Downloaded from informahealthcare.com by Dr. Josep Gamez on 08/23/12
For personal use only.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The endocannabinoid system (ECS) is a lipid signalling system, comprising of the endogenous cannabis-like ligands (endocannabinoids) anandamide (AEA) and 2-arachidonoylglycerol (2-AG), which derive from arachidonic acid. These bind to a family of G-protein-coupled receptors, called CB1 and CB2. The cannabinoid receptor 1 (CB1R) is distributed in brain areas associated with motor control, emotional responses, motivated behaviour and energy homeostasis. In the periphery, the same receptor is expressed in the adipose tissue, pancreas, liver, GI tract, skeletal muscles, heart and the reproduction system. The CB2R is mainly expressed in the immune system regulating its functions. Endo cannabinoids are synthesized and released upon demand in a receptor-dependent way. They act as retrograde signalling messengers in GABAergic and glutamatergic synapses and as modulators of postsynaptic transmission, interacting with other neurotransmitters. Endocannabinoids are transported into cells by a specific uptake system and degraded by the enzymes fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL). The ECS is involved in various pathophysiological conditions in central and peripheral tissues. It is implicated in the hormonal regulation of food intake, cardiovascular, gastrointestinal, immune, behavioral, antiproliferative and mammalian reproduction functions. Recent advances have correlated the ECS with drug addiction and alcoholism. The growing number of preclinical and clinical data on ECS modulators is bound to result in novel therapeutic approaches for a number of diseases currently treated inadequately. The ECS dysregulation has been correlated to obesity and metabolic syndrome pathogenesis. Rimonabant is the first CB1 blocker launched to treat cardiometabolic risk factors in obese and overweight patients. Phase III clinical trials showed the drug's ability to regulate intra-abdominal fat tissue levels, lipidemic, glycemic and inflammatory parameters. However, safety conerns have led to its withdrawal. The role of endocannabinoids in mammalian reproduction is an emerging research area given their implication in fertilization, preimplantation embryo and spermatogenesis. The relevant preclinical data on endocannabinoid signalling open up new perspectives as a target to improve infertility and reproductive health in humans.
Article
There is now convincing evidence for the existence of at least two types of cannabinoid receptor, CB1 and CB2, both coupled to G proteins. CB1 receptors are present in the central nervous system and in certain peripheral tissues where at least some are located at autonomic nerve terminals. CB2 receptors are found only outside the brain, mainly in cells of the immune system. The existence of endogenous ligands for cannabinoid receptors, both centrally and peripherally, is also generally accepted. These recent discoveries have prompted the development of cannabinoid receptor antagonists and of selective cannabinoid CB1 and CB2 receptor agonists. These compounds are important experimental tools that will help to establish the physiological roles of cannabinoid receptors and their endogenous ligands. The availability of such compounds should also facilitate the discovery of novel therapeutic uses for cannabinoid receptor agonists and antagonists.
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.
Article
Cannabidiol and other cannabinoids were examined as neuroprotectants in rat cortical neuron cultures exposed to toxic levels of the neurotransmitter, glutamate. The psychotropic cannabinoid receptor agonist Δ9-tetrahydrocannabinol (THC) and cannabidiol, (a non-psychoactive constituent of marijuana), both reduced NMDA, AMPA and kainate receptor mediated neurotoxicities. Neuroprotection was not affected by cannabinoid receptor antagonist, indicating a (cannabinoid) receptor-independent mechanism of action. Glutamate toxicity can be reduced by antioxidants. Using cyclic voltametry and a fenton reaction based system, it was demonstrated that Cannabidiol, THC and other cannabinoids are potent antioxidants. As evidence that cannabinoids can act as an antioxidants in neuronal cultures, cannabidiol was demonstrated to reduce hydroperoxide toxicity in neurons. In a head to head trial of the abilities of various antioxidants to prevent glutamate toxicity, cannabidiol was superior to both a-tocopherol and ascorbate in protective capacity. Recent preliminary studies in a rat model of focal cerebral ischemia suggest that cannabidiol may be at least as effective in vivo as seen in these in vitro studies.
Chapter
To date, two cannabinoid receptors have been isolated by molecular cloning. The CB1 and CB2 cannabinoid receptors are members of the G protein-coupled receptor family. There is also evidence for additional cannabinoid receptor subtypes. The CB1 and CB2 receptors recognize endogenous and exogenous cannabinoid compounds, which fall into five structurally diverse classes. Mutagenesis and molecular modeling studies have identified several key amino acid residues involved in the selective recognition of these ligands. Numerous residues involved in receptor activation have been elucidated. Regions of the CB1 receptor mediating desensitization and internalization have also been discovered. The known genetic structures of the CB1 and CB2 receptors indicate polymorphisms and multiple exons that may be involved in tissue and species-specific regulation of these genes. The cannabinoid receptors are regulated during chronic agonist exposure, and gene expression is altered in disease states. There is a complex molecular architecture of the cannabinoid receptors that allows a single receptor to recognize multiple classes of compounds and produce an array of distinct downstream effects.
Article
Significant advances are being made towards understanding the genetic basis for spinal neurodegenerative diseases, however, effective pharmacotherapy remains elusive. One of the primary theories underlying neuron vulnerability is susceptibility to excitotoxicity. We present for the first time evidence that the activation of the CB1 cannabinoid receptor effectively modulates kainate toxicity in primary neuronal cultures prepared from mouse spinal cord. Addition of Δ9-tetrahydrocannabinol to the culture medium attenuated the toxicity produced by kainate. The CB1 receptors were localized to spinal neurons and astrocytes. The neuroprotective effect was blocked with the CB1 receptor antagonist, SR141716A, indicating a receptor-mediated effect.