16,916
Views
8
CrossRef citations to date
0
Altmetric
Brief Report

ALSUntangled No. 16: Cannabis

Pages 400-404 | Published online: 28 May 2012

Introduction

In a widely viewed series of Internet videos called “Surviving ALS,” Cathy Jordan reports that regularly smoking cannabis has dramatically slowed her ALS progression, and improved her mood and appetite (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 (Citation1–9). Moreover, cannabis could potentially be useful in managing the symptomatology in ALS (Citation10–13). 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 (Citation14–22). Approximately 70 are chemically unique and classified as plant cannabinoids (Citation14–17). Cannabinoids are lipophilic 21 carbon terpenes, biosynthesized predominantly via a recently discovered deoxyxylulose phosphate pathway (Citation14). Delta-9 tetrahydrocannabinol (THC) and delta-8 THC appear to produce the majority of the psychoactive effects of cannabis (Citation18,Citation19). 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 ingredients and may produce most of its effects at moderate, 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 (Citation16). The amount varies according to plant. Some varieties of Cannabis sativa have been found to have no cannabidiol (Citation16). Cannabidiol appears to modulate and reduce untoward effects of THC (Citation18). Cannabidiol breaks down to cannabinol as the plant matures. Much less is known about cannabinol, although it appears to have distinct pharmacological properties from cannabidiol. Cannabinol has anticonvulsant, sedative, and other pharmacological activities likely to interact with the effects of THC (Citation19–22). Cannabinol may induce sleep and may provide some protection against seizures for epileptics (Citation22).

Cannabis Receptors and Endogenous Ligands

Recent advances have increased understanding of the receptors and endogenous ligands composing the cannabinoid system (Citation23–36). Two major cannabinoid receptor subtypes exist: CB1 is predominantly expressed in the brain, and CB2 is primarily found on the cells of the immune system (Citation23,Citation37–39). Both receptors are G protein-coupled, 7-segment transmembrane proteins, similar to the receptors of dopamine, serotonin, and norepinephrine (Citation39,Citation40). Dense cannabinoid receptor concentrations are found in the cerebellum, basal ganglia, and hippocampus, likely accounting for the effect of exogenously administered cannabinoids on motor tone and coordination as well as mood state (Citation41–43). Low concentrations are found in the brainstem, perhaps accounting for the low potential for lethal overdose with cannabinoid-based medicines (Citation44–47).

The discovery of endocannabinoids, (i.e. endogenous metabolites capable of activating the cannabinoid receptors), and an improved understanding of the molecular mechanisms leading to their biosynthesis, release, and inactivation, have inspired research on the pharmaceutical applications of cannabinoid-based medicines (Citation40). A growing number of strategies 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 endocannabinoid uptake or metabolism (Citation48). Distinct varieties of cannabis contain different combinations of partial cannabinoid agonists and antagonists, which could be utilized in designing synthetic cannabinoid 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 inflammatory responses (Citation49–52). Since the pathophysiology of ALS may involve neuroinflammation (Citation53,Citation54), agents such as cannabinoids that modulate this process could potentially be useful.

Alternatively, or in addition, cannabinoids might act similarly to tamoxifen, a Food and Drug Administration (FDA)-approved drug used to treat breast cancer (Citation55–57). Both cannabinoids and tamoxifen are terpenes, organic, lipid soluble compounds that readily penetrate the CNS (Citation55). In a 60 patient pilot study, PALS taking tamoxifen reportedly had improved survival and no significant side effects (Citation56); unfortunately this study has yet to be published 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 inflammation, or by altering glutamate uptake (Citation55). Endocannabinoids can also modulate glutamatergic neurotransmission indirectly via NMDA receptors (Citation33,Citation34,Citation36).

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 symptomatic G93A-SOD1 mice (Citation9). 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 paralleling disease progression (Citation2). The sensitivity of CB1 receptors in controlling both glutamate and GABA transmission is potentiated in the striatum of symptomatic G93A-SOD1 ALS mice (Citation5).

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 (Citation58). Daily injections of the selective CB2 agonist AM-1241, initiated at symptom onset in G93A-SOD1 ALS mice, increased survival after disease onset by 56% (Citation2). 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 (Citation6). 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 (Citation6). Ablation of the CB1 receptor had no effect on disease onset in G93A-SOD1 ALS mice but significantly extended life span. These animal studies all have significant methodological flaws, including small sample size, lack of randomization and lack of blinding.

What Are the Efficacy, 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. (Citation59). While this was well-tolerated, there was no effect on cramp frequency or intensity, or on secondary 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 (Citation11). However Dronabinol 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 (Citation60–62). Most patients find dronabinol too sedating and associated with too many psychoactive effects (Citation63–66). 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. Benefits described included improved speech, swallowing, secretions, fasciculations, appetite, sleep and mood. Side effects included dry mouth, clumsiness, dizziness, 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. Obtaining 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. 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 (Citation67). Although THC is not the ideal target compound per se, it is a relative indicator of potency and quality. For comparison, the average THC content of cannabis at randomly surveyed medical cooperatives in California is approximately 15 to 20% (Citation68). Thus, an independent source of cannabis would be needed to ensure a consistently high cannabinoid content that may be strong enough to possibly 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 (Citation69,Citation70). Nonetheless, it is possible, with coordinated effort, to effectively do double-blind, randomized, 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 (Citation71,Citation72).

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)(Citation73). 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 psychoactive, 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 financial information. GW Pharma openly acknowledges that it needs a large patient base to be financially 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 immunomodulation 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 progression, 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 controlled 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 Bedlack, 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 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.

Disclosures: ALSUntangled is sponsored by the Packard Center and the Motor Neurone Disease Association.

References

  • Kim K, Moore DH, Makriyannis A, Abood ME. AM1241, a cannabinoid CB2 receptor selective compound, delays disease 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. 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, Abood ME. Amyotrophic lateral sclerosis: delayed disease progression in mice by treatment with a cannabinoid. Amyotroph Lateral Scler Other Motor Neuron Disord 2004; 5(1):33–9.
  • Weydt P, Hong S, Witting A, Möller T, Stella N, Kliot M. Cannabinol delays symptom onset in SOD1 (G93A) transgenic mice without affecting survival. Amyotroph Lateral Scler Other Motor Neuron Disord 2005; 6(3):182–4.
  • Rossi S, De Chiara V, Musella A, Cozzolino M, Bernardi G, 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, 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 amyotrophic lateral sclerosis. Curr Pharm Des 2008; 14(23): 2306–16.
  • Zhao P, Ignacio S, Beattie EC, Abood ME. Altered presymptomatic AMPA and cannabinoid receptor trafficking 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. Endocannabinoids accumulate in spinal cord of SOD1 transgenic mice. J Neurochem. 2004;89:1555–7.
  • Carter GT, BS Rosen. Marijuana in the management of amyotrophic lateral sclerosis. Am J Hosp Palliat Care 2001; 18(4):264–70.
  • Gelinas D, Miller RG, Abood ME. A pilot study of safety 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. 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 promise for neurological disorders. Curr Opin Investig Drugs 2002; 3(3):437–440
  • Gerra G, Zaimovic A, Gerra ML, Ciccocioppo R, Cippitelli 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 qualitative 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. 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. Phytocannabinoids as novel therapeutic agents in CNS disorders. Pharmacol Ther 2012;133:79–97.
  • Akinshola BE; Chakrabarti A; Onaivi ES. In vitro and in vivo action of cannabinoids. Neurochem Res 1999; 24(10): 1233–40.
  • Agurell S, Halldin M, Lindgren JE, Ohlsson A, Widman M, Gillespie H, and Hollister, L. Pharmacokinetics and metabolism 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, Maiello F, Guadagno F, Petrosino S, Capasso F, Di Marzo V, Izzo AA. Cannabidiol, a safe and non-psychotropic ingredient 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, Guimarães FS, Joca SR. 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, Whalley 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. Handb Exp Pharmacol 2005; 168:81–115.
  • McAllister SD, Hurst DP, Barnett-Norris J, Lynch D, Reggio 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, Moore DH, Aldape K, Yount G, Cannabinoids selectively inhibit proliferation and induce death of cultured human glioblastoma multiforme cells. J Neurooncol 2005; 74(1): 31–40
  • Kapur A, Samaniego P, Thakur GA, Makriyannis A, 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, Wentland M, Abood ME, Hough LB. Significance of cannabinoid CB1 receptors in improgan antinociception. J Pain 2007; 8(11):850–60.
  • Anavi-Goffer S, Fleischer D, Hurst DP, Lynch DL, Barnett-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 mechanisms. J Biol Chem 2007; 282(34):25100–13.
  • McAllister SD, Rizvi G, Anavi-Goffer S, Hurst DP, Barnett-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 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 Pharmacol 2002; 63(12):2121–36.
  • Abood ME, Rizvi G, Sallapudi N, McAllister SD, Activation 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, Breuer A, Mechoulam R, Shohami E. An endogenous cannabinoid (2 AG) is neuroprotective after brain injury. Nature 2001; 4;413(6855):527–31.
  • Chen Y; Buck J. Cannabinoids protect cells from oxidative 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 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 survival/death decision by cannabinoids. J Mol Med 2001; 78:613–625.
  • Maenpaa H, Mannerstrom M, Toimela T, Salminen L, Saransaari P, Tahti H. Glutamate uptake is inhibited by tamoxifen and toremifene in cultured retinal pigment epithelial cells. Pharmacol Toxicol 2002; 91(3):116–122
  • Klein TW; Lane B; Newton CA; Friedman H. The cannabinoid system and cytokine network. Proc Soc Exp Biol Med 2000;225(1):1 8.
  • Pertwee RG: Cannabinoid receptor ligands: clinical and neuropharmacological 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 action of cannabinoids. Neurochem Res 1999;24(10): 1233–40
  • Di Marzo, Bisogno T, De Petrocellis L. Endocannabinoids: new targets for drug development. Curr Pharm Des 2000; 6(13):1361–80.
  • Hanuš LO, Mechoulam R. Novel Natural and Synthetic Ligands of the Endocannabinoid System. Curr Med Chem 2010; Feb 18. [Epub ahead of print].
  • De Laurentiis A, Fernández Solari J, Mohn C, Zorrilla Zubilete M, Rettori V. Endocannabinoid system participates in neuroendocrine control of homeostasis. Neuroimmunomodulation 2010;17(3):153–6.
  • Nagarkatti P, Pandey R, Rieder SA, Hegde VL, Nagarkatti M. Cannabinoids as novel anti-inflammatory drugs. Future Med Chem 2009;1(7):1333–1349.
  • Mouslech Z, Valla V. Endocannabinoid system: An overview of its potential in current medical practice. Neuro Endocrinol Lett 2009;30(2):153–79.
  • Wegener N, Koch M. Neurobiology and systems physiology of the endocannabinoid system. Pharmacopsychiatry 2009;42 Suppl 1:S79–86.
  • De Petrocellis L, Di Marzo V. An introduction to the endocannabinoid 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 transduction. Vitam Horm. 2009;81:79–110.
  • Pertwee RG. Ligands that target cannabinoid receptors in the brain: from THC to anandamide and beyond. Addict Biol 2008;13(2):147–59.
  • Cencioni MT, Chiurchiù V, Catanzaro G, Borsellino G, Bernardi G, Battistini L, Maccarrone M. Anandamide suppresses 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, Barcaroli D, Oddi S, Federici G, Urbani A, Finazzi Agrò 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. Endocannabinoids and immune regulation. Pharmacol Res> 2009; 60(2):85–92.
  • Parolaro D, Massi P, Rubino T, Monti E. Endocannabinoids 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 S, Re DB. Inflammation 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-motoneuron dialogue in ALS. Acta Myol 2011;30:4–8.
  • Maenpaa H, Mannerstrom M, Toimela T, Salminen L, Saransaari P, Tahti H. Glutamate uptake is inhibited by tamoxifen and toremifene in cultured retinal pigment epithelial cells. Pharmacol Toxicol 2002; 91(3):116–122
  • Traynor BJ, Bruijn L, Conwit R, Beal F, O'Neill G, Fagan SC, Cudkowicz ME. Neuroprotective agents for clinical trials in ALS: a systematic assessment. Neurology 2006; 11; 67(1):20–7.
  • Lee ES, Yin Z, Milatovic D, Jiang H, Aschner M. Estrogen 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, Abood ME. Amyotrophic lateral sclerosis: delayed disease progression in mice by treatment with a cannabinoid. Amyotroph Latearl Scler Other Motor Neuron Disor 2004;5:33–39.
  • Weber M, Goldman B, Truniger S. Tetrahydrocannabinol (THC) for cramps in amyotrophic lateral sclerosis: a randomized, double-blind crossover trial. J Neurol Neurosurg Psychiatry 2010;1135–40.
  • Yiangou Y, Facer P, Durrenberger P, Chessell IP, Naylor A, Bountra C, Banati RR, Anand P. COX-2, CB2 and P2X7-immunoreactivities are increased in activated microglial cells/macrophages of multiple sclerosis and amyotrophic lateral sclerosis spinal cord. BMC Neurol 2006; 6:12.
  • Hampson AJ; Grimaldi M; Axelrod J; Wink D. Cannabidiol 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; 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; 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 saga, science, and sobriquet. Chem Biodivers. 2007;4(8):1614–48.
  • El Sohly MA, Wachtel SR, de Wit H. Cannabis versus THC: response to Russo and McPartland. Psychopharmacology 2003; 165(4):433–434.
  • Carter GT, Flanagan A, Earleywine M, Abrams DI, Aggarwal 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, ZumBrunnen 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 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. Medicinal cannabis: rational guidelines for dosing. IDrugs 2004;7(5):464–70.
  • Aggarwal SK, Kyashna-Tocha M, Carter GT. Dosing Medical Marijuana: Rational Guidelines on Trial in Washington State. MedGenMed 2007; 9(3):52–3.
  • Abrams DI, Vizoso HP, Shade SB, Jay C, Kelly ME, Benowitz NL. Vaporization as a smokeless cannabis delivery system: a pilot study. Clin Pharmacol Ther 2007; 82(5): 572–8.
  • Pomahacova B, Van der Kooy F, Verpoorte R. Cannabis smoke condensate III: The cannabinoid content of vaporised Cannabis sativa. Inhal Toxicol 2009;21(13):1108–12.
  • Kmietowicz Z. Cannabis based drug is licensed for spasticity in patients with MS. BMJ 2010;340:c3363.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.