Canna~Fangled Abstracts

The therapeutic potential of cannabinoids for movement disorders.

By February 4, 2015No Comments
 pub med big
Logo of nihpa

Mov Disord. Author manuscript; available in PMC 2016 Mar 1.
Published in final edited form as:

Published online 2015 Feb 4. doi:  10.1002/mds.26142

PMCID: PMC4357541
NIHMSID: NIHMS649683
Benzi Kluger, M.D., M.S.,1 Piera Triolo,1 Wallace Jones,1 and  Joseph Jankovic, M.D.2

Abstract

Background

There is growing interest in the therapeutic potential of marijuana (cannabis) and cannabinoid-based chemicals within the medical community and particularly for neurologic conditions. This interest is driven both by changes in the legal status of cannabis in many areas and increasing research into the roles of endocannabinoids within the central nervous system and their potential as symptomatic and/or neuroprotective therapies. We review basic science, preclinical and clinical studies on the therapeutic potential of cannabinoids specifically as it relates to movement disorders.

Results

The pharmacology of cannabis is complex with over 60 neuroactive chemicals identified to date. The endocannabinoid system modulates neurotransmission involved in motor function, particularly within the basal ganglia. Preclinical research in animal models of several movement disorders have shown variable evidence for symptomatic benefits but more consistently suggest potential neuroprotective effects in several animal models of Parkinson’s (PD) and Huntington’s disease (HD). Clinical observations and clinical trials of cannabinoid-based therapies suggests a possible benefit of cannabinoids for tics and probably no benefit for tremor in multiple sclerosis or dyskinesias or motor symptoms in PD. Data are insufficient to draw conclusions regarding HD, dystonia or ataxia and nonexistent for myoclonus or restless legs syndrome.

Conclusions

Despite the widespread publicity about the medical benefits of cannabinoids, further preclinical and clinical research is needed to better characterize the pharmacological, physiological and therapeutic effects of this class of drugs in movement disorders.

Keywords: cannabinoids, cannabis, movement disorders, Parkinson’s disease, Huntington’s disease

Introduction

PM 1aCannabis (marijuana) has long been used for medicinal purposes in many cultures as well as for spiritual and recreational purposes due to its psychoactive properties. Over 60 pharmacologically active compounds or phytocannabinoids have been isolated from cannabis including Δ9-tetrahydrocannabinol (THC), the primary psychotropic compound, and cannabidiol (CBD), a nonpsychoactive chemical with potential therapeutic properties.1 Nearly all cannabis strains are derived from two cannabis species, sativa and indica. Sativa strains have higher THC concentrations and produce more euphoria, whereas indica strains have more CBD and have more sedating, anti-emetic, and analgesic properties.

Over the past three decades, cannabinoid receptors and endogenously produced cannabinoids (eCBs) have been discovered in a wide range of tissues including peripheral nerves and the central nervous system (CNS). The endocannabinoid system (ECS) has been implicated in a broad range of physiological functions, including cognition, mood, motor control, feeding behaviors, and pain.25Consequently, cannabinoid-based therapies have been studied for a variety of illnesses.6 Cannabinoid-based medicines, such as nabilone, dronabinol, and Sativex®, are now approved for clinical indications, including pain, anorexia, spasticity, and chemotherapy-induced nausea and Epidiolex™recently obtained orphan drug status for Dravet syndrome. 67

Preclinical research suggests that cannabinoids have symptomatic and neuroprotective potential for a variety of neurologic conditions, including movement disorders. The American Academy of Neurology (AAN) Guideline Development Subcommittee systematically evaluated the published clinical evidence and concluded that oral cannabis extract is effective in treating multiple sclerosis (MS) related spasticity and central pain or painful spasms, and that cannabinoid-based therapies are probably ineffective in treating levodopa-induced dyskinesias (LID) in Parkinson’s disease (PD) or tremor and are of unknown efficacy for Huntington’s disease (HD), tics or dystonia.8 Our objective is to provide a more in-depth review of preclinical and clinical studies related to the therapeutic potential of cannabinoids for movement disorders.

PRECLINICAL RESEARCH

Endocannabinoids and the Basal Ganglia

The primary cannabinoid receptor subtypes are cannabinoid receptors type 1 (CB1) and type 2 (CB2). CB1 receptors are highly expressed in the CNS, especially the basal ganglia, and also identified in almost all peripheral tissues and cell types.9 CB2 receptors are expressed primarily in the immune system, where they modulate inflammation, but are also expressed in the CNS, particularly in neurons within the dorsal vagal motor nucleus, the nucleus ambiguous, the spinal trigeminal nucleus, and microglia.1011 Recently, CB2 receptors were found in the basal ganglia and studies suggest that impairment of these receptors may be associated with dyskinesias 12 While most actions of cannabinoids are related to CB1 and CB2 receptors, other receptor types have been described, including the transient receptor potential vanilloid type 1 (TRPV1) cation channel,13 the GTP-binding protein-coupled receptor GPR55,14 the abnormal-CBD receptor,15 and the peroxisome-proliferator-activated receptor (PPAR).16

eCBs are lipophilic compounds that demonstrate varying degrees of affinity for G-protein coupled cannabinoid receptors and include anandamide and 2-arachidonoglycerol (2-AG; see Table 1). eCBs primarily function through retrograde signaling, wherein post-synaptic activity leads to eCB production and release with backward transmission across the synapse to depress presynaptic neurotransmitter release.5 The ECS may also support synapse formation and neurogenesis.5 Within the basal ganglia, eCBs and CB1 receptors tend to increase GABAergic and inhibit glutamatergic transmission.9 eCBs also tend to inhibit dopamine release through GABAergic mechanisms.17 eCBs are not stored and are quickly degraded after exerting a transient and localized effect. Removal of eCBs from the extracellular space occurs through cellular uptake and metabolism with anandamide degraded primarily by fatty acid amide hydrolysis (FAAH) and 2-AG degraded by monoacylglycerol lipase.18

Table 1

Summary of compounds mentioned in this review

Neuroprotective Potential of Cannabinoids

Several studies in animal models of both PD and HD suggest that cannabinoid-based therapies may attenuate neurodegeneration (Table 2). Indeed, on October 7, 2003, U.S. Health and Human Services was granted U.S. Patent 6630507, which lists the use of cannabinoids found within the cannabis sativa plant as useful in certain neurodegenerative diseases such as PD, Alzheimer’s disease and dementia caused by human immunodeficiency virus.141 Cannabinoids may offer neuroprotection through both receptor-mediated and receptor-independent mechanisms. Cannabinoids are capable of reducing oxidative damage by acting as scavengers of reactive oxygen species (ROS) and enhancing endogenous antioxidant defenses.19 This property appears to be independent of CB1 and CB2 receptor modulation and restricted to certain cannabinoids, including CBD, THC, cannabinol, CP55,940, and the anandamide analogue AM404.2022 CB2 agonists exert anti-inflammatory effects by inhibiting reactive microglia and cytokine release.202325 Lastly, CB1 agonists reduce excitotoxicity by suppressing glutamatergic activity, subsequent calcium ion influx, and nitric oxide production.2627However, in one study, both a CB1 agonist, THC, and a selective CB1 antagonist, rimonabant, exacerbated malonate-induced striatal lesions.28

Table 2

Preclinical Neuroprotective Studies of Cannabinoids

Parkinson’s disease

Experimental models of PD show increased ECS activity in the basal ganglia, including increased CB1 mRNA levels, CB1 activity, anandamide levels, and decreased cannabinoid clearance.2933These changes appear to be associated with movement suppression and may be reversed by chronic levodopa treatment.2935 Importantly, many cannabinoids demonstrate neuroprotective effects in several models of PD (Table 2). These effects appear to be mediated by both CB receptor dependent and independent mechanisms including antioxidant effects, reduced microglia activation, and modulation of glial-neuron interactions.20,24,67,121

Animal studies further suggest that cannabinoids may improve motor symptoms of PD and LID, but results are variable (Table 3). CB1 agonists inhibit basal ganglia dopamine release and are there for expected to be ineffective in alleviating PD motor symptoms. Indeed, CB1 agonists have been shown to exacerbate bradykinesia in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned primates.38 However, CB1 agonists have also been reported to improve motor impairments, possibly through nondopaminergic mechanisms including interactions with adenosine A2A receptors.3944Studies of CB1 antagonists are more consistent in improving motor symptoms without increasing dyskinesias.4549 These effects appear to involve nondopaminergic mechanisms including enhanced striatal glutamate release and may be greater in animals with more severe striatonigral degeneration.294647 Potential explanations for the therapeutic variability of CB-based compounds include differences in lesion severity, trial design, dose, and gender.464849 Different CB1modulators may also exhibit functional selectivity for different G proteins or subpopulations of CB1receptors, a phenomenon referred to as biased agonism.5051

Table 3

Preclinical studies assessing therapeutic symptomatic efficacy of cannabinoids for movement disorders

While CB1 and CB2 receptors are decreased in the basal ganglia of dyskinetic animals, it is not known whether this is compensatory or causal.12 Animal studies suggest cannabinoid-based therapies may improve LID without worsening motor control. Interestingly, these effects are reported for both CB1agonists52 and antagonists45, although these effects are not seen in all studies and higher doses of CB1 agonists may impair motor function suggesting that CB1 agonist effects on LID are related to general motor suppressant effects.5355 Other CB receptors may also be involved in LID as URB597 (a FAAH inhibitor which increases anandamide levels) did not affect LID as monotherapy but did improve LID when co-administered with a TRPV1 antagonist, suggesting that TRPV1 and CB1 may have opposing effects.54

Huntington’s Disease

Experimental animal models indicate that HD is associated with early and widespread reductions in the ECS, particularly CB1 receptors in the striatum.62,63 CB1 receptors mediate brain-derived neurotrophic factor expression and CB1 receptor loss is associated with exacerbation of symptoms, neuropathology, and molecular pathology in the striatum. Moreover, cannabinoid-based therapies generally show neuroprotection in several animal models through both CB receptor mediated and independent effects (Table 2).145,124,127 Caution is warranted as several studies using identical cannabinoids and models showed no benefit or even exacerbation of neurotoxicity.67,28,126Therapeutic studies of cannabinoid-based agents in HD animal models suggest that CB1 and endovanilloid receptor agonists2866 and anandamide reuptake inhibitors67 are capable of alleviating hyperkinesia (Table 3). This therapeutic potential is likely to be realized in early phases of HD because of progressive loss of CB1 receptors in advanced stages.68

Dystonia and Tremor

It has been hypothesized that CB1 agonists reduce overactivity of the globus pallidus interna (GPi) and improve dystonia by reducing GABA reuptake.73 In support of this idea, the CB1 and CB2agonist WIN55,212-2 produces antidystonic effects in a mutant hamster model of dystonia, increases the antidystonic efficacy of benzodiazepines and is reversed by rimonabant, a selective CB1antagonist.7475 Animal models suggest that cannabinoids may reduce MS-related tremor, an effect that appears to be selectively mediated by CB1 receptors.92

CLINICAL RESEARCH

Parkinson’s Disease

Compared to control subject, humans with PD show elevated cerebrospinal fluid levels of anandamide and autopsied brains of PD patients show decreased CB1 mRNA expression in the basal ganglia.3436It is not clear whether these discrepancies reflect medication effects, down-regulation from increased agonist activity or differences in disease severity. Regarding non-motor symptoms, one study reported CB1 receptor gene (CNR1) polymorphisms may influence depression risk in PD.37

Observational and uncontrolled studies suggest cannabinoids may improve PD motor symptoms.5658In a survey of PD patients (N = 339) in the Czech Republic, 25% of respondents reported using cannabis and 46% of these described some benefit; 31% reported improvement of rest tremor, 45% of bradykinesia, and 14% of LID.57 Notably, there was a 54% response rate to this survey suggesting a potential for respondent bias. A small (N=22) open-label study assessing motor exam 30 minutes after smoking cannabis also reported improvements in tremor, rigidity, bradykinesia, pain and sleep.58Regarding nonmotor symptoms, a small (N=6) 4-week open-label study of CBD for psychosis in PD found improvements on the Brief Psychiatric Rating Scale and Parkinson Psychosis Questionnaire and another case series (N=4) reported benefits for rapid eye movement sleep behavior disorder.56,139However, a case series of 5 patients found no benefit for tremor following a single administration of smoked cannabis.138

In contrast, 4 controlled clinical studies of cannabinoids reported no benefit for motor symptoms, mixed results regarding LID and quality of life.5961140 (Table 4) A small (N=5) randomized, double-blind, placebo-controlled crossover trial, assessed the efficacy of 0.03 mg/kg nabilone, a CB1and CB2 agonist for LID given in a split dose 12 and 1 hour prior to a levodopa challenge.61 They found a significant reduction in LID versus placebo on the Rush Dyskinesia Disability Scale and total LID time. Nabilone did not diminish the antiparkinsonian actions of levodopa or improve parkinsonian symptoms although 2 patients reported improvements in painful off-period dystonia. A larger (N=17) 4-week randomized double-blind crossover study of twice daily Cannador, an oral cannabis extract containing 1.25 mg CBD and 2.5 mg THC, titrated Cannador up to 0.25 mg/kg THC for LID.59 Although the blinding seemed to be compromised (71% correct identification of treatment), Cannador failed to improve LID on multiple outcomes, including a non-significant worsening on their primary outcome (UPDRS dyskinesia items) and the Rush Dyskinesia scale. There were no significant changes for other secondary outcomes including motor symptoms (Part 3 UPDRS), quality of life (PDQ-39) or sleep. Another small (N=8) 16-day randomized placebo-controlled trial assessing the efficacy of 20 mg daily oral rimonabant (CB1 antagonist) showed no effect on parkinsonian motor symptoms or LID as measured by the UPDRS and a standardized videotape procedure.60 Examination of data suggested a trend towards worsening in motor scores but the small sample size and other methodological issues prevent any meaningful conclusions. Most recently, 21 PD patients were randomized to placebo, CBD 75mg/day or CBD 300 mg/day for a 6-week trial.140 No significant changes were found for the total UPDRS or any subscales or measures of neuroprotection (serum brain derived neurotrophic factor levels or putamen magnetic resonance spectroscopy). Improvements were noted for the total PDQ-39 score and stigma and activities of daily living subscores for the CBD 300 mg/day group. Despite the low sample size and quality of these studies, the data suggest cannabinoid agonists and antagonists are probably ineffective for both LID and motor symptoms although further study using different doses, formulations or target symptoms (e.g. dystonia, psychosis, sleep) may be justified. While there were no serious adverse events reported, side effects included hypotension, vertigo, visual hallucinations, dizziness and somnolence.

Table 4

Clinical studies of cannabinoids for movement disorders

Huntington’s Disease

Post-mortem human studies and PET imaging studies using a CB1 ligand support experimental HD models in demonstrating early and marked decreases in both subcortical and cortical CB1 receptors.64,65 Clinical research of cannabinoids for HD symptoms are inconclusive. (Table 4) Case reports using nabilone, a CB1 agonist, reported worsening of chorea severity in one patient and benefits for chorea and irritability in another.6970 A small (N=15) randomized double-blind placebo-controlled crossover trial assessed CBD capsules (10 mg/kg divided in twice daily doses) given for 6-weeks for chorea as measured by the Marsden and Quinn chorea severity scale.71 This study found small and statistically insignificant differences between groups on primary and secondary outcomes, including patient global impressions. A larger (N=37) 5-week double-blind placebo-controlled randomized cross-over trial assessed nabilone titrated to 1 or 2 mg given twice daily for 5 weeks on the total motor score of the Unified Huntington’s disease rating scale (UHDRS).72 Change in total UHDRS did not differ between groups. However, statistically significant improvements were noted for the UHDRS chorea scale and the neuropsychiatric inventory with a trend for improvement on the UHDRS behavior score. There were no statistical differences reported between the 1 and 2 mg. Notably, one patient withdrew due to severe sedation. These results are encouraging and suggest a need for larger controlled trials. A phase II (N=25) double-blind placebo-controlled randomized clinical trial is examining the safety and neuroprotective efficacy of nabiximols, a combination of CBD and THC, using clinical and biomarker (cerebrospinal brain-derived neurotrophic factor) outcomes, but results are not yet available (NCT Identifier: NCT01502046).

Dystonia

While case reports of smoked cannabis for generalized dystonia in Wilson’s disease,76 idiopathic hemidystonia,77 and a case series of 5 patients with dystonia secondary to diverse causes treated with oral CBD (100–600 mg daily)78 suggest cannabinoids may alleviate dystonia, 2 small randomized placebo-controlled clinical trials for dystonia showed no effect (Table 4). Notably, the case series reported exacerbated hypokinesia and tremor in 2 patients and other mild side effects including hypotension, dry mouth, psychomotor slowing, lightheadedness, and sedation.78 A small (N=13) randomized double-blind placebo-controlled cross-over trial tested a single dose of 0.03 mg/kg nabilone or placebo on patients with medication refractory focal or generalized dystonia using the Burke-Fahn-Marsden dystonia scale.73 They reported no significant differences between groups at 60, 120 or 180 minutes, including when separating generalized from segmental patients. 4 patients reported a subjective improvement lasting 3 days after nabilone and 2 patients withdrew secondary to hypotension and sedation. A second small (N=7) randomized placebo-controlled crossover trial assessed 7.5 mg dronabinol, a CB1 and CB2 agonist, given twice daily for 2 weeks in patients with cervical dystonia using the Toronto Western Hospital Spasmodic Torticollis Rating Scale (TWSTRS).79 There were no significant differences noted on TWSTRS (total or subscales) or subjective ratings. One subject withdrew secondary to insomnia and sensation of heart racing, and all but one subject reported mild side effects including lightheadedness, hypotension, vertigo and dry mouth.

Tics and Tourette Syndrome

Case reports of smoked cannabis,80 oral THC,8184 and case series of smoked cannabis (N = 3)85suggest that cannabinoids may be beneficial for tics in patients with Tourette syndrome (TS). Similarly, a survey of 64 TS patients found that 17 (27%) had tried marijuana and 14 of them (82%) found it helpful for tics and behavioral disturbances.86 Although only 2 controlled trials have assessed the efficacy of cannabinoids for tics, the results support these uncontrolled clinical reports. A small (N=12) randomized double-blind placebo-controlled single-dose crossover trial assessed a single dose of 5–10 mg oral THC (dose based on body weight and prior marijuana use) for tics in TS using the Tourette Syndrome Symptom List (TSSL).87 Tics and obsessive compulsive behavior significantly improved on the TSSL with statistically significant improvements or trends towards improvement in other secondary outcomes including the Yale Global Tic Severity Scale. Mild side effects noted by 5 patients included headache, dizziness, nausea and cognitive changes. Another small (N=17) 6-week randomized placebo-controlled parallel group trial of 6-weeks 10 mg daily orally-administered THC assessed tic reduction in TS patients using the Tourette Syndrome Clinical Global Impression scale (TS-CGI).88 This study also found significant improvements in the primary and most secondary outcomes. A related study from the same cohort reported no significant change in neuropsychological performance with THC treatment.89 Limitations of these studies include small sample size, short treatment period, multiple comparisons, fixed or single dose approach, potential blinding issues and possible selection bias. Given these limitations, AAN evidence-based systematic review, as well as a recent Cochran review on the efficacy of cannabinoids in TS, state that there is presently “insufficient evidence to support or refute the clinical use of THC, nabilone, or cannabis for tics”.890 However, in treatment resistant adult patients, THC may have therapeutic effects and is recommended by some experts.91 Positive results of preliminary studies warrant validation of the efficacy and safety of THC for tics in larger randomized clinical trials.

Multiple Sclerosis-related Tremor

A small (N=8) case series of 5–15 mg oral THC given 4 times a day reported objective improvement in tremor in 2 patients and subjective improvement in 5.93 However, subsequent clinical trials assessing the efficacy of cannabinoids for MS-related tremor failed to show benefit. A small (N=13) 6-week randomized double-blind placebo-controlled trial assessed Cannador dosed up to 0.125 mg/kg THC twice daily for patients with MS-related tremor.94 There were no statistically significant effects as measured by a tremor index, the Nine-Hole Peg Test of manual dexterity, spiral drawings or other objective measures although 5 patients reported subjective benefits. Notably, 9 of 13 patients correctly guessed their treatment group and 10 reported mild adverse effects including cognitive changes, drowsiness, lightheadedness and dry mouth. A large (N=337) 8-week double-blind, randomized placebo-controlled trial of up to 24 actuations per day of oral nabiximols spray (65 mg THC, 60 mg CBD), evaluated tremor as a secondary outcome with an index score and patient global impression of change and reported no effect on tremor.95 Another large (N=391) 15-week randomized placebo-controlled trial assessing the efficacy of oral THC (marinol) versus oral cannabis extract (both dosed up to 12.5 mg given twice daily) versus placebo showed no difference in patient ratings of tremor as a secondary outcome.96 Given the consistent lack of response to cannabinoids in patients suffering from MS-related tremor, AAN evidence-based guidelines state THC and oral cannabis extract are probably ineffective for reducing MS-related tremor and nabiximols are possibly ineffective.8 To our knowledge there have been no studies of CB-based treatments for essential tremor or other tremor types.

Other Movement Disorders

There have been no published clinical trials of cannabinoids for ataxia, myoclonus or restless legs syndrome. Two case reports suggest ataxia (in combination with spasticity) in MS may improve following smoked cannabis or oral THC and a survey of 112 MS patients reported some individuals noting improvement in balance.939798 Marijuana may be effective for psychogenic symptoms as illustrated by a report by Sanjay Gupta, called “Weed”, which featured a 19 year old young man who after 7 minutes of “convulsing” with “myoclonus – diaphragmatic flutter”, resembling psychogenic tic or tremor, had a sudden resolution of the movement, respiratory and speech disorder within seconds of smoking marijuana.99,142

Adverse Effects and Risk of Addiction

While cannabinoids appear to be well tolerated when used in moderation, AEs are clearly a major concern. In the systematic review of CB studies conducted by the AAN 6.9% (95% CI 5.7%–8.2%) of participants stopped their medication due to AEs.8 AEs include ataxia, nausea, impaired short-term memory, stroke, cognitive impairment, dry mouth, suicidal ideation, hallucinations, dizziness, fatigue, behavioral or mood changes, impaired motor skills, increased weakness, heart rate and appetite.8100101 Marijuana use is also associated with an increased risk of chronic anxiety, depression and psychosis, though causality has not been established.102 Except for ataxia, there have been no documented cases of movement disorders induced by cannabis use. However, there has been one case report of propriospinal myoclonus possibly induced by cannabis,103 but this form of myoclonus is often of psychogenic origin.104

It is important to note that side effects, as well as therapeutic effects, vary depending on the CB(s), concentration of CB(s), or ratio of CBs in formulations.105 Smoking cannabis has been associated with lung cancer risk so future trials should focus on other methods of ingestion, although oral administration is also problematic due to deposition of cannabinoids into fatty tissue, from which they are released slowly, causing variability in plasma concentrations.106107

There is also an important risk of abuse with marijuana and cannabis-based drugs.108 Studies of marijuana outside of the medical context estimate 9% of persons using cannabis may become addicted and experience symptoms of withdrawal after quitting the drug.109 The mechanism of abuse in marijuana users is not well understood but may involve blunted dopamine reactivity. This is supported by the observation that marijuana abusers, compared to healthy controls, showed markedly blunted dopamine responses when challenged with methylphenidate and that PET scans using [11C] raclopride, a D2 ligand, show dopamine in ventral striatum of marijuana abusers was inversely correlated with addiction severity and craving.110 It is likely that patients using medical formulations may be at lower risk, due to lower doses, use related to a specific indication, and formulations with a lower concentration of THC, the key component responsible for the dependence potential of cannabis.111 Cannabis is generally safe in overdose although illicit synthetic cannabinoids have been associated with severe medical and psychiatric complications.112113 Finally, epidimiological studies suggest that cannabis may be a “gateway drug” as its use is associated with later use of other illicit substances.143 While the causal role and mechanisms of the gateway idea are still debated, it is prudent for future clinical trials to monitor for current and subsequent use of other illicit substances.144

Discussion and Directions for Future Research

Although the number of preclinical studies of cannabinoids for movement disorders has rapidly increased in the last three decades, there are marked gaps in our knowledge about their effects on motor pathways. There are also marked discrepancies and conflicting results in preclinical studies including the precise effects of cannabinoids on neurotransmission and cellular mechanisms of neuroprotection. Furthermore, the paradox of how both CB1 agonists and antagonists exert similar effects on LID and other hyperkinetic movement disorders needs to be explained. Future studies should address not only motor and behavioral effects of cannabinoids but, because of their proven effects on the sensory system, also explore their effects on sensory-motor integration, a disorder which is increasingly recognized as an aspect of movement disorders.114

Several factors may help explain conflicting preclinical results. Given that cannabinoids interact with a wide range of pharmacological targets, discrepancies in data obtained from preclinical studies may partly reflect the multiplicity of cannabinoid actions.115 The complex localization of cannabinoid receptors at different sites in basal ganglia circuits and the broad array of formulations and doses used in preclinical and clinical studies may also help explain contradictory results.850

A change in classification from Schedule I to Schedule IV or V would not only improve access to medical marijuana but could facilitate development and conduct of clinical trials for cannabinoids.116Future clinical trials should be adequately powered, employ appropriate methodology and outcome measures for the specific movement disorder studied and assess blinding adequacy.117 Improved knowledge of cannabinoids and their pharmacology may help identify specific cannabinoids or combinations that provide therapeutic or neuroprotective benefits in patients with movement disorders.

Acknowledgments

Funding sources: This work was not funded.

Dr. Jankovic acknowledges the support of his Centers of Excellence by the National Parkinson Foundation and the Huntington Disease Society of America.

Footnotes

Financial Disclosure/Conflict of Interest: None to report.

Author roles:

BK – Design and conceptualization of the manuscript, review and critique of subsequent drafts.

PT – Literature search and review, analysis and interpretation of data, writing of first draft, design and construction of tables.

WJ – Literature search and review, analysis and interpretation of data, writing and revision of subsequent drafts, design and construction of tables.

JJ – Literature search and review, review and critique of drafts.

Financial Disclosures of all authors:

Dr. Jankovic:

Current Research and Center of Excellence Grants: Allergan, Inc; Auspex Pharmaceuticals, Inc; CHDI Foundation; GE Healthcare; Great Lakes NeuroTechnologies; Huntington’s Disease Society of America; Huntington Study Group; Ipsen Limited; Kyowa Haako Kirin Pharma, Inc; Lundbeck Inc; Medtronic; Merz Pharmaceuticals; Michael J Fox Foundation for Parkinson Research; National Institutes of Health; National Parkinson Foundation; Omeros Corporation; Parkinson Study Group; Pharma Two B; Prothena Biosciences Inc; Psyadon Pharmaceuticals, Inc; St. Jude Medical; Teva Pharmaceutical Industries Ltd; UCB Inc; University of Rochester; Current consultant or an advisory committee member: Allergan, Inc; Auspex Pharmaceuticals, Inc; Ipsen Biopharmaceuticals, Inc.; Lundbeck Inc; Teva Pharmaceutical Industries Ltd

Royalties: Cambridge; Elsevier; Future Science Group; Hodder Arnold; Lippincott Williams and Wilkins; Wiley-Blackwell

During the past two years Dr. Jankovic has served on the following editorial boards and foundation advisory boards:

Editorial boards: Medlink: Neurology; Expert Review of Neurotherapeutics; Neurology in Clinical Practice; The Botulinum Journal; PeerJ; Therapeutic Advances in Neurological Disorders; Neurotherapeutics; Tremor and Other Hyperkinetic Movements; Journal of Parkinson’s Disease; UpToDate.

Speakers Bureau: None

Stock Ownership: None

Dr. Kluger:

Current Research and Recently Completed Research Grants: Colorado Clinical and Translational Sciences Institute, Department of Defense – Army Research Office, Michael J. Fox Foundation, National Institutes of Health (National Institutes of Aging; National Institute of Neurological Disorders and Stroke; National Center for Advancing Translational Sciences), University of Colorado Center for Neuroscience, University of Colorado Hospital Clinical Effectiveness and Safety Program, Veterans Affairs Medical Center Editorial and Foundation Advisory Boards: Frontiers in Movement Disorders, Parkinson’s Association of the Rockies

Royalties: None

Speaker’s Bureau: None

Stock Ownership: None

Financial disclosure related to research covered in this article: None.

Citations

1. Huestis MA. Human cannabinoid pharmacokinetics. Chem Biodivers. 2007;4(8):1770–1804.[PMC free article]  [PubMed]
2. Howlett AC, Barth F, Bonner TI, et al. International Union of Pharmacology. XXVII. Classification of cannabinoid receptors. Pharmacol Rev. 2002;54(2):161–202.  [PubMed]
3. Di Marzo V, Melck D, Bisogno T, De Petrocellis L. Endocannabinoids: endogenous cannabinoid receptor ligands with neuromodulatory action. Trends Neurosci. 1998;21(12):521–528.  [PubMed]
4. Di Marzo V. The endocannabinoid system: its general strategy of action, tools for its pharmacological manipulation and potential therapeutic exploitation. Pharmacol Res. 2009;60(2):77–84.  [PubMed]
5. Castillo PE, Younts TJ, Chavez AE, Hashimotodani Y. Endocannabinoid signaling and synaptic function. Neuron. 2012;76(1):70–81. [PMC free article]  [PubMed]
6. Pacher P, Kunos G. Modulating the endocannabinoid system in human health and disease–successes and failures. Febs J. 2013;280(9):1918–1943. [PMC free article]  [PubMed]
7. Devinsky O, Cilio MR, Cross H, et al. Cannabidiol: pharmacology and potential therapeutic role in epilepsy and other neuropsychiatric disorders. Epilepsia. 2014;55(6):791–802. [PMC free article][PubMed]
8. Koppel BS, Brust JC, Fife T, et al. Systematic review: efficacy and safety of medical marijuana in selected neurologic disorders: report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology. 2014;82(17):1556–1563. [PMC free article]  [PubMed]
9. Benarroch E. Endocannabinoids in basal ganglia circuits: implications for Parkinson disease. Neurology. 2007;69(3):306–309.  [PubMed]
10. Van Sickle MD, Duncan M, Kingsley PJ, et al. Identification and functional characterization of brainstem cannabinoid CB2 receptors. Science. 2005;310(5746):329–332.  [PubMed]
11. Nunez E, Benito C, Tolon RM, Hillard CJ, Griffin WS, Romero J. Glial expression of cannabinoid CB(2) receptors and fatty acid amide hydrolase are beta amyloid-linked events in Down’s syndrome. Neuroscience. 2008;151(1):104–110.  [PubMed]
12. Sierra S, Luquin N, Rico AJ, et al. Detection of cannabinoid receptors CB1 and CB2 within basal ganglia output neurons in macaques: changes following experimental parkinsonism. Brain Struct Funct. 2014  [PubMed]
13. Toth A, Blumberg PM, Boczan J. Anandamide and the vanilloid receptor (TRPV1) Vitam Horm. 2009;81:389–419.  [PubMed]
14. Ross RA. The enigmatic pharmacology of GPR55. Trends Pharmacol Sci. 2009;30(3):156–163.[PubMed]
15. McHugh D, Hu SS, Rimmerman N, et al. N-arachidonoyl glycine, an abundant endogenous lipid, potently drives directed cellular migration through GPR18, the putative abnormal cannabidiol receptor. BMC Neurosci. 2010;11:44. [PMC free article]  [PubMed]
16. O’Sullivan SE. Cannabinoids go nuclear: evidence for activation of peroxisome proliferator-activated receptors. Br J Pharmacol. 2007;152(5):576–582. [PMC free article]  [PubMed]
17. Fitzgerald ML, Shobin E, Pickel VM. Cannabinoid modulation of the dopaminergic circuitry: implications for limbic and striatal output. Prog Neuropsychopharmacol Biol Psychiatry. 2012;38(1):21–29. [PMC free article]  [PubMed]
18. Kendall D, Alexander S, editors. Behavioral Neurobiology of the Endocannabinoid System.Berlin: Spinger-Verlag; 2009. 
19. Sagredo O, Garcia-Arencibia M, de Lago E, Finetti S, Decio A, Fernandez-Ruiz J. Cannabinoids and neuroprotection in basal ganglia disorders. Mol Neurobiol. 2007;36(1):82–91.  [PubMed]
20. Lastres-Becker I, Molina-Holgado F, Ramos JA, Mechoulam R, Fernandez-Ruiz J. Cannabinoids provide neuroprotection against 6-hydroxydopamine toxicity in vivo and in vitro: relevance to Parkinson’s disease. Neurobiol Dis. 2005;19(1–2):96–107.  [PubMed]
21. Garcia-Arencibia M, Gonzalez S, de Lago E, Ramos JA, Mechoulam R, Fernandez-Ruiz J. Evaluation of the neuroprotective effect of cannabinoids in a rat model of Parkinson’s disease: importance of antioxidant and cannabinoid receptor-independent properties. Brain Res. 2007;1134(1):162–170.  [PubMed]
22. Jimenez-Del-Rio M, Daza-Restrepo A, Velez-Pardo C. The cannabinoid CP55,940 prolongs survival and improves locomotor activity in Drosophila melanogaster against paraquat: implications in Parkinson’s disease. Neurosci Res. 2008;61(4):404–411.  [PubMed]
23. Klein TW. Cannabinoid-based drugs as anti-inflammatory therapeutics. Nat Rev Immunol. 2005;5(5):400–411.  [PubMed]
24. Carroll CB, Zeissler ML, Hanemann CO, Zajicek JP. Delta(9)-tetrahydrocannabinol (Delta(9)-THC) exerts a direct neuroprotective effect in a human cell culture model of Parkinson’s disease. Neuropathol Appl Neurobiol. 2012;38(6):535–547.  [PubMed]
25. Sagredo O, Pazos MR, Satta V, Ramos JA, Pertwee RG, Fernandez-Ruiz J. Neuroprotective effects of phytocannabinoid-based medicines in experimental models of Huntington’s disease. J Neurosci Res. 2011;89(9):1509–1518.  [PubMed]
26. Romero J, Orgado JM. Cannabinoids and neurodegenerative diseases. CNS Neurol Disord Drug Targets. 2009;8(6):440–450.  [PubMed]
27. Martinez-Orgado J, Fernandez-Lopez D, Lizasoain I, Romero J. The seek of neuroprotection: introducing cannabinoids. Recent Pat CNS Drug Discov. 2007;2(2):131–139.  [PubMed]
28. Lastres-Becker I, Bizat N, Boyer F, Hantraye P, Brouillet E, Fernandez-Ruiz J. Effects of cannabinoids in the rat model of Huntington’s disease generated by an intrastriatal injection of malonate. Neuroreport. 2003;14(6):813–816.  [PubMed]
29. Di Marzo V, Hill MP, Bisogno T, Crossman AR, Brotchie JM. Enhanced levels of endogenous cannabinoids in the globus pallidus are associated with a reduction in movement in an animal model of Parkinson’s disease. Faseb J. 2000;14(10):1432–1438.  [PubMed]
30. Romero J, Berrendero F, Perez-Rosado A, et al. Unilateral 6-hydroxydopamine lesions of nigrostriatal dopaminergic neurons increased CB1 receptor mRNA levels in the caudate-putamen. Life Sci. 2000;66(6):485–494.  [PubMed]
31. Lastres-Becker I, Cebeira M, de Ceballos ML, et al. Increased cannabinoid CB1 receptor binding and activation of GTP-binding proteins in the basal ganglia of patients with Parkinson’s syndrome and of MPTP-treated marmosets. Eur J Neurosci. 2001;14(11):1827–1832.  [PubMed]
32. Gubellini P, Picconi B, Bari M, et al. Experimental parkinsonism alters endocannabinoid degradation: implications for striatal glutamatergic transmission. J Neurosci. 2002;22(16):6900–6907.[PubMed]
33. Mailleux P, Vanderhaeghen JJ. Dopaminergic regulation of cannabinoid receptor mRNA levels in the rat caudate-putamen: an in situ hybridization study. J Neurochem. 1993;61(5):1705–1712.[PubMed]
34. Hurley MJ, Mash DC, Jenner P. Expression of cannabinoid CB1 receptor mRNA in basal ganglia of normal and parkinsonian human brain. J Neural Transm. 2003;110(11):1279–1288.  [PubMed]
35. Maccarrone M, Gubellini P, Bari M, et al. Levodopa treatment reverses endocannabinoid system abnormalities in experimental parkinsonism. J Neurochem. 2003;85(4):1018–1025.  [PubMed]
36. Pisani A, Fezza F, Galati S, et al. High endogenous cannabinoid levels in the cerebrospinal fluid of untreated Parkinson’s disease patients. Ann Neurol. 2005;57(5):777–779.  [PubMed]
37. Barrero FJ, Ampuero I, Morales B, et al. Depression in Parkinson’s disease is related to a genetic polymorphism of the cannabinoid receptor gene (CNR1) Pharmacogenomics J. 2005;5(2):135–141.[PubMed]
38. Meschler JP, Howlett AC, Madras BK. Cannabinoid receptor agonist and antagonist effects on motor function in normal and 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine (MPTP)-treated non-human primates. Psychopharmacology (Berl) 2001;156(1):79–85.  [PubMed]
39. van Vliet SA, Vanwersch RA, Jongsma MJ, Olivier B, Philippens IH. Therapeutic effects of Delta9-THC and modafinil in a marmoset Parkinson model. Eur Neuropsychopharmacol. 2008;18(5):383–389.  [PubMed]
40. Sanudo-Pena MC, Patrick SL, Khen S, Patrick RL, Tsou K, Walker JM. Cannabinoid effects in basal ganglia in a rat model of Parkinson’s disease. Neurosci Lett. 1998;248(3):171–174.  [PubMed]
41. Brotchie JM. Adjuncts to dopamine replacement: a pragmatic approach to reducing the problem of dyskinesia in Parkinson’s disease. Mov Disord. 1998;13(6):871–876.  [PubMed]
42. Kreitzer AC, Malenka RC. Endocannabinoid-mediated rescue of striatal LTD and motor deficits in Parkinson’s disease models. Nature. 2007;445(7128):643–647.  [PubMed]
43. Segovia G, Mora F, Crossman AR, Brotchie JM. Effects of CB1 cannabinoid receptor modulating compounds on the hyperkinesia induced by high-dose levodopa in the reserpine-treated rat model of Parkinson’s disease. Mov Disord. 2003;18(2):138–149.  [PubMed]
44. Fernandez-Espejo E, Caraballo I, Rodriguez de Fonseca F, et al. Experimental parkinsonism alters anandamide precursor synthesis, and functional deficits are improved by AM404: a modulator of endocannabinoid function. Neuropsychopharmacology. 2004;29(6):1134–1142.  [PubMed]
45. van der Stelt M, Fox SH, Hill M, et al. A role for endocannabinoids in the generation of parkinsonism and levodopa-induced dyskinesia in MPTP-lesioned non-human primate models of Parkinson’s disease. Faseb J. 2005;19(9):1140–1142.  [PubMed]
46. Fernandez-Espejo E, Caraballo I, de Fonseca FR, et al. Cannabinoid CB1 antagonists possess antiparkinsonian efficacy only in rats with very severe nigral lesion in experimental parkinsonism. Neurobiol Dis. 2005;18(3):591–601.  [PubMed]
47. Garcia-Arencibia M, Ferraro L, Tanganelli S, Fernandez-Ruiz J. Enhanced striatal glutamate release after the administration of rimonabant to 6-hydroxydopamine-lesioned rats. Neurosci Lett. 2008;438(1):10–13.  [PubMed]
48. Kelsey JE, Harris O, Cassin J. The CB(1) antagonist rimonabant is adjunctively therapeutic as well as monotherapeutic in an animal model of Parkinson’s disease. Behav Brain Res. 2009;203(2):304–307.  [PubMed]
49. Gonzalez S, Scorticati C, Garcia-Arencibia M, de Miguel R, Ramos JA, Fernandez-Ruiz J. Effects of rimonabant, a selective cannabinoid CB1 receptor antagonist, in a rat model of Parkinson’s disease. Brain Res. 2006;1073–1074:209–219.  [PubMed]
50. Morera-Herreras T, Miguelez C, Aristieta A, Ruiz-Ortega JA, Ugedo L. Endocannabinoid modulation of dopaminergic motor circuits. Front Pharmacol. 2012;3:110. [PMC free article][PubMed]
51. Console-Bram L, Brailoiu E, Brailoiu GC, Sharir H, Abood ME. Activation of GPR18 by cannabinoid compounds: a tale of biased agonism. Br J Pharmacol. 2014;171(16):3908–3917.[PMC free article]  [PubMed]
52. Fox SH, Henry B, Hill M, Crossman A, Brotchie J. Stimulation of cannabinoid receptors reduces levodopa-induced dyskinesia in the MPTP-lesioned nonhuman primate model of Parkinson’s disease. Mov Disord. 2002;17(6):1180–1187.  [PubMed]
53. Cao X, Liang L, Hadcock JR, et al. Blockade of cannabinoid type 1 receptors augments the antiparkinsonian action of levodopa without affecting dyskinesias in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated rhesus monkeys. J Pharmacol Exp Ther. 2007;323(1):318–326.  [PubMed]
54. Morgese MG, Cassano T, Cuomo V, Giuffrida A. Anti-dyskinetic effects of cannabinoids in a rat model of Parkinson’s disease: role of CB(1) and TRPV1 receptors. Experimental neurology. 2007;208(1):110–119. [PMC free article]  [PubMed]
55. Walsh S, Gorman AM, Finn DP, Dowd E. The effects of cannabinoid drugs on abnormal involuntary movements in dyskinetic and non-dyskinetic 6-hydroxydopamine lesioned rats. Brain Res. 2010;1363:40–48.  [PubMed]
56. Zuardi AW, Crippa JA, Hallak JE, et al. Cannabidiol for the treatment of psychosis in Parkinson’s disease. J Psychopharmacol. 2009;23(8):979–983.  [PubMed]
57. Venderova K, Ruzicka E, Vorisek V, Visnovsky P. Survey on cannabis use in Parkinson’s disease: subjective improvement of motor symptoms. Mov Disord. 2004;19(9):1102–1106.  [PubMed]
58. Lotan I, Treves TA, Roditi Y, Djaldetti R. Cannabis (medical marijuana) treatment for motor and non-motor symptoms of Parkinson disease: an open-label observational study. Clin Neuropharmacol. 2014;37(2):41–44.  [PubMed]
59. Carroll CB, Bain PG, Teare L, et al. Cannabis for dyskinesia in Parkinson disease: a randomized double-blind crossover study. Neurology. 2004;63(7):1245–1250.  [PubMed]
60. Mesnage V, Houeto JL, Bonnet AM, et al. Neurokinin B, neurotensin, and cannabinoid receptor antagonists and Parkinson disease. Clin Neuropharmacol. 2004;27(3):108–110.  [PubMed]
61. Sieradzan KA, Fox SH, Hill M, Dick JP, Crossman AR, Brotchie JM. Cannabinoids reduce levodopa-induced dyskinesia in Parkinson’s disease: a pilot study. Neurology. 2001;57(11):2108–2111.  [PubMed]
62. Lastres-Becker I, Berrendero F, Lucas JJ, et al. Loss of mRNA levels, binding and activation of GTP-binding proteins for cannabinoid CB1 receptors in the basal ganglia of a transgenic model of Huntington’s disease. Brain Res. 2002;929(2):236–242.  [PubMed]
63. Lastres-Becker I, Hansen HH, Berrendero F, et al. Alleviation of motor hyperactivity and neurochemical deficits by endocannabinoid uptake inhibition in a rat model of Huntington’s disease. Synapse. 2002;44(1):23–35.  [PubMed]
64. Van Laere K, Casteels C, Dhollander I, et al. Widespread decrease of type 1 cannabinoid receptor availability in Huntington disease in vivo. J Nucl Med. 2010;51(9):1413–1417.  [PubMed]
65. Glass M, Dragunow M, Faull RL. The pattern of neurodegeneration in Huntington’s disease: a comparative study of cannabinoid, dopamine, adenosine and GABA(A) receptor alterations in the human basal ganglia in Huntington’s disease. Neuroscience. 2000;97(3):505–519.  [PubMed]
66. de Lago E, Urbani P, Ramos JA, Di Marzo V, Fernandez-Ruiz J. Arvanil, a hybrid endocannabinoid and vanilloid compound, behaves as an antihyperkinetic agent in a rat model of Huntington’s disease. Brain Res. 2005;1050(1–2):210–216.  [PubMed]
67. de Lago E, Fernandez-Ruiz J, Ortega-Gutierrez S, et al. UCM707, an inhibitor of the anandamide uptake, behaves as a symptom control agent in models of Huntington’s disease and multiple sclerosis, but fails to delay/arrest the progression of different motor-related disorders. Eur Neuropsychopharmacol. 2006;16(1):7–18.  [PubMed]
68. Lastres-Becker I, Fezza F, Cebeira M, et al. Changes in endocannabinoid transmission in the basal ganglia in a rat model of Huntington’s disease. Neuroreport. 2001;12(10):2125–2129.  [PubMed]
69. Muller-Vahl KR, Schneider U, Emrich HM. Nabilone increases choreatic movements in Huntington’s disease. Mov Disord. 1999;14(6):1038–1040.  [PubMed]
70. Curtis A, Rickards H. Nabilone could treat chorea and irritability in Huntington’s disease. J Neuropsychiatry Clin Neurosci. 2006;18(4):553–554.  [PubMed]
71. Consroe P, Laguna J, Allender J, et al. Controlled clinical trial of cannabidiol in Huntington’s disease. Pharmacol Biochem Behav. 1991;40(3):701–708.  [PubMed]
72. Curtis A, Mitchell I, Patel S, Ives N, Rickards H. A pilot study using nabilone for symptomatic treatment in Huntington’s disease. Mov Disord. 2009;24(15):2254–2259.  [PubMed]
73. Fox SH, Kellett M, Moore AP, Crossman AR, Brotchie JM. Randomised, double-blind, placebo-controlled trial to assess the potential of cannabinoid receptor stimulation in the treatment of dystonia. Mov Disord. 2002;17(1):145–149.  [PubMed]
74. Richter A, Loscher W. (+)-WIN 55,212-2, a novel cannabinoid receptor agonist, exerts antidystonic effects in mutant dystonic hamsters. Eur J Pharmacol. 1994;264(3):371–377.  [PubMed]
75. Richter A, Loscher W. Effects of pharmacological manipulations of cannabinoid receptors on severity of dystonia in a genetic model of paroxysmal dyskinesia. Eur J Pharmacol. 2002;454(2–3):145–151.  [PubMed]
76. Uribe Roca MC, Micheli F, Viotti R. Cannabis sativa and dystonia secondary to Wilson’s disease. Mov Disord. 2005;20(1):113–115.  [PubMed]
77. Chatterjee A, Almahrezi A, Ware M, Fitzcharles MA. A dramatic response to inhaled cannabis in a woman with central thalamic pain and dystonia. J Pain Symptom Manage. 2002;24(1):4–6.[PubMed]
78. Consroe P, Sandyk R, Snider SR. Open label evaluation of cannabidiol in dystonic movement disorders. Int J Neurosci. 1986;30(4):277–282.  [PubMed]
79. Zadikoff C, Wadia PM, Miyasaki JM, et al. Cannabinoid, CB1 agonists in cervical dystonia: Failure in a phase IIa randomized controlled trial. Basal Ganglia. 2011;1(2):91–95.
80. Hemming M, Yellowlees PM. Effective treatment of Tourette’s syndrome with marijuana. J Psychopharmacol. 1993;7(4):389–391.  [PubMed]
81. Brunnauer A, Segmiller FM, Volkamer T, Laux G, Muller N, Dehning S. Cannabinoids improve driving ability in a Tourette’s patient. Psychiatry Res. 2011;190(2–3):382.  [PubMed]
82. Muller-Vahl KR, Schneider U, Kolbe H, Emrich HM. Treatment of Tourette’s syndrome with delta-9-tetrahydrocannabinol. Am J Psychiatry. 1999;156(3):495.  [PubMed]
83. Müller-Vahl K, Schneider U, Emrich H. Combined treatment of Tourette-Syndrome with delta-9-THC and dopamine receptor antagonists. Journal of Cannabis Therapeutics. 2002;2:145–154.
84. Hasan A, Rothenberger A, Munchau A, Wobrock T, Falkai P, Roessner V. Oral delta 9-tetrahydrocannabinol improved refractory Gilles de la Tourette syndrome in an adolescent by increasing intracortical inhibition: a case report. J Clin Psychopharmacol. 2010;30(2):190–192.[PubMed]
85. Sandyk R, Awerbuch G. Marijuana and Tourette’s syndrome. J Clin Psychopharmacol. 1988;8(6):444–445.  [PubMed]
86. Muller-Vahl KR, Kolbe H, Schneider U, Emrich HM. Cannabinoids: possible role in patho-physiology and therapy of Gilles de la Tourette syndrome. Acta Psychiatr Scand. 1998;98(6):502–506.  [PubMed]
87. Muller-Vahl KR, Schneider U, Koblenz A, et al. Treatment of Tourette’s syndrome with Delta 9-tetrahydrocannabinol (THC): a randomized crossover trial. Pharmacopsychiatry. 2002;35(2):57–61.[PubMed]
88. Muller-Vahl KR, Schneider U, Prevedel H, et al. Delta 9-tetrahydrocannabinol (THC) is effective in the treatment of tics in Tourette syndrome: a 6-week randomized trial. J Clin Psychiatry. 2003;64(4):459–465.  [PubMed]
89. Muller-Vahl KR, Prevedel H, Theloe K, Kolbe H, Emrich HM, Schneider U. Treatment of Tourette syndrome with delta-9-tetrahydrocannabinol (delta 9-THC): no influence on neuropsychological performance. Neuropsychopharmacology. 2003;28(2):384–388.  [PubMed]
90. Curtis A, Clarke CE, Rickards HE. Cannabinoids for Tourette’s Syndrome. Cochrane Database Syst Rev. 2009;(4):CD006565.  [PubMed]
91. Muller-Vahl KR. Treatment of Tourette syndrome with cannabinoids. Behav Neurol. 2013;27(1):119–124.  [PubMed]
92. Baker D, Pryce G, Croxford JL, et al. Cannabinoids control spasticity and tremor in a multiple sclerosis model. Nature. 2000;404(6773):84–87.  [PubMed]
93. Clifford DB. Tetrahydrocannabinol for tremor in multiple sclerosis. Ann Neurol. 1983;13(6):669–671.  [PubMed]
94. Fox P, Bain PG, Glickman S, Carroll C, Zajicek J. The effect of cannabis on tremor in patients with multiple sclerosis. Neurology. 2004;62(7):1105–1109.  [PubMed]
95. Collin C, Ehler E, Waberzinek G, et al. A double-blind, randomized, placebo-controlled, parallel-group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis. Neurol Res. 2010;32(5):451–459.  [PubMed]
96. Zajicek J, Fox P, Sanders H, et al. Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial. Lancet. 2003;362(9395):1517–1526.  [PubMed]
97. Meinck HM, Schonle PW, Conrad B. Effect of cannabinoids on spasticity and ataxia in multiple sclerosis. J Neurol. 1989;236(2):120–122.  [PubMed]
98. Consroe P, Musty R, Rein J, Tillery W, Pertwee R. The perceived effects of smoked cannabis on patients with multiple sclerosis. Eur Neurol. 1997;38(1):44–48.  [PubMed]
99. Baizabal-Carvallo JF, Jankovic J. The clinical features of psychogenic movement disorders resembling tics. Journal of Neurological Neurosurgery Psychiatry. 2014;85(5):573–5.  [PubMed]
100. Moreira FA, Grieb M, Lutz B. Central side-effects of therapies based on CB1 cannabinoid receptor agonists and antagonists: focus on anxiety and depression. Best Pract Res Clin Endocrinol Metab. 2009;23(1):133–144.  [PubMed]
101. Riedel G, Davies SN. Cannabinoid function in learning, memory and plasticity. Handb Exp Pharmacol. 2005;(168):445–477.  [PubMed]
102. Volkow ND, Baler RD, Compton WM, Weiss SR. Adverse health effects of marijuana use. N Engl J Med. 2014;370(23):2219–2227. [PMC free article]  [PubMed]
103. Lozsadi DA, Forster A, Fletcher NA. Cannabis-induced propriospinal myoclonus. Mov Disord. 2004;19(6):708–709.  [PubMed]
104. Esposito M, Erro R, Edwards MJ, et al. The pathophysiology of symptomatic propriospinal myoclonus. Mov Disord. 2014  [PubMed]
105. Borgelt LM, Franson KL, Nussbaum AM, Wang GS. The pharmacologic and clinical effects of medical cannabis. Pharmacotherapy. 2013;33(2):195–209.  [PubMed]
106. Berthiller J, Straif K, Boniol M, et al. Cannabis smoking and risk of lung cancer in men: a pooled analysis of three studies in Maghreb. J Thorac Oncol. 2008;3(12):1398–1403.  [PubMed]
107. Lile JA, Kelly TH, Charnigo RJ, Stinchcomb AL, Hays LR. Pharmacokinetic and pharmacodynamic profile of supratherapeutic oral doses of Delta(9)-THC in cannabis users. J Clin Pharmacol. 2013;53(7):680–690. [PMC free article]  [PubMed]
108. Haberstick BC, Young SE, Zeiger JS, et al. Prevalence and correlates of alcohol and cannabis use disorders in the United States: results from the national longitudinal study of adolescent health. Drug Alcohol Depend. 2014;136:158–161. [PMC free article]  [PubMed]
109. Warner LA, Kessler RC, Hughes M, Anthony JC, Nelson CB. Prevalence and correlates of drug use and dependence in the United States. Results from the National Comorbidity Survey. Arch Gen Psychiatry. 1995;52(3):219–229.  [PubMed]
110. Volkow ND, Wang GJ, Telang F, et al. Decreased dopamine brain reactivity in marijuana abusers is associated with negative emotionality and addiction severity. Proc Natl Acad Sci U S A. 2014;111(30):E3149–3156. [PMC free article]  [PubMed]
111. Robson P. Abuse potential and psychoactive effects of delta-9-tetrahydrocannabinol and cannabidiol oromucosal spray (Sativex), a new cannabinoid medicine. Expert Opin Drug Saf. 2011;10(5):675–685.  [PubMed]
112. Robson P. Therapeutic aspects of cannabis and cannabinoids. Br J Psychiatry. 2001;178:107–115.[PubMed]
113. Berry-Caban CS, Ee J, Ingram V, Berry CE, Kim EH. Synthetic cannabinoid overdose in a 20-year-old male US soldier. Subst Abus. 2013;34(1):70–72.  [PubMed]
114. Patel N, Jankovic J, Hallett M. Sensory aspects of movement disorders. Lancet Neurol. 2014;13(1):100–112. [PMC free article]  [PubMed]
115. Gowran A, Noonan J, Campbell VA. The multiplicity of action of cannabinoids: implications for treating neurodegeneration. CNS Neurosci Ther. 2011;17(6):637–644.  [PubMed]
116. Wolk L, Kuehn BM. Colorado tackles medical implications of marijuana. Jama. 2014;311(20):2053–2054.  [PubMed]
117. Galpern WR, Coffey CS, Albanese A, et al. Designing clinical trials for dystonia. Neurotherapeutics. 2014;11(1):117–127. [PMC free article]  [PubMed]
118. Garcia C, Palomo-Garo C, Garcia-Arencibia M, et al. Symptom-relieving and neuroprotective effects of the phytocannabinoid 9-THCV in animal models of Parkinson’s disease. British Journal of Pharmacology. 2011;163:1495–1506. [PMC free article]  [PubMed]
119. Gilgun-Sherki Y, Melamed E, Mechoulam R, Daniel O. The CB1 cannabinoid receptor agonist, HU-210, reduces levodopa-induced rotations in 6-hydroxydopamine-lesioned rats. Pharmacology & Toxicology. 2003;93:66–70.  [PubMed]
120. Jeon P, Yang S, Jeong H, Kim H. Cannabinoid receptor agonist protects cultured dopaminergic neurons from the death by the proteasomal dysfunction. Anatomy and Cell Biology. 2011:135–142.[PMC free article]  [PubMed]
121. Price D, Martinez A, Seillier A, et al. WIN55,212-2, a cannabinoid receptor agonist, protects against nigrostriatal cell loss in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. European Journal of Neuroscience. 2009;29:2177–2186. [PMC free article][PubMed]
122. Lastres-Becker I, Bizat N, Boyer F, et al. Potential involvement of cannabinoid receptors in 3-nitropropionic acid toxicity in vivo. Neuroreport. 2004;15(25):2375–2379.  [PubMed]
123. Palazuelos J, Aguado T, Pazos M, et al. Microglial CB2 cannabinoid receptors are neuroprotective in Huntington’s disease excitotoxicity. Brain. 2009;132:1352–3164.  [PubMed]
124. Pintor A, Tebano MT, Martire A, et al. The cannabinoid receptor agonist WIN 55, 212-2 attenuates the effects induced by quinolinic acid in the rat striatum. Neuropharmacology. 2006;51:1004–1012.  [PubMed]
125. Sagredo O, Gonzalez S, Aroyo I, et al. Cannabinoid CB2 receptor agonists protect the striatum against malonate toxicity: Relevance for Huntington’s disease. GLIA. 2009;57:1154–1167.[PMC free article]  [PubMed]
126. Scotter EL, Goodfellow CE, Graham ES, Dragunow M, Glass M. Neuroprotective potential of CB1 receptor agonists in an in vitro model of Huntington’s disease. British Journal of Pharmacology. 2010;160:747–761. [PMC free article]  [PubMed]
127. Valdeolivas S, Satta V, Pertwee R, et al. Sativex-like combination of phytocannabinoids is neuroprotective in malonate-lesioned rats, an inflammatory model of Huntington’s disease: Role of CB1 and CB2 receptors. ACS Chemical Neuroscience. 2012:400–406. [PMC free article]  [PubMed]
128. Gutierrez-Valdez AL, Garcia-Ruiz R, Anaya-Martinez V, et al. The combination of oral L-DOPA/rimonabant for effective dyskinesia treatment and cytological preservation in a rat model of Parkinson’s disease and L-DOPA-induced dyskinesia. Behavioural Pharmacology. 2013;24(8):640–652.  [PubMed]
129. Johnston T, Huot P, Fox S, et al. Fatty acid amide hydrolase (FAAH) inhibition reduces L-3,4-dihydroxyphenylalanine-induced hyperactivity in the 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned non-human primate model of Parkinson’s disease. The Journal of Pharmacology and Experimental Therapies. 2011;336(2):423–430.  [PubMed]
130. Madsen MV, Peacock LP, Werge T, et al. Effects of cannabinoid CB1 receptor agonism and antagonism on SKF81297-induced dyskinesia and haloperidol-induced dystonia in Cebus apella monkeys. Neuropharmacology. 2011;60:418–422.  [PubMed]
131. Martinez A, Macheda T, Morgese M, Trabace L, Giuffrida A. The cannabinoid agonist WIN55212-2 decreases l-DOPA-induced PKA activation and dyskinetic behavior in 6-OHDA-treated rats. Neuroscience Research. 2012;72:236–242. [PMC free article]  [PubMed]
132. Moss DE, McMaster B, Rogers J. Tetrahydrocannabinol potentiates reserpine-induced hypokinesia. Pharmacology Biochemistry & Behavior. 1981;15:779–783.  [PubMed]
133. Van Vilet S, Vanwersch R, Jongsma M, Olivier B, Philippens I. Therapeutic effects of Δ9-THC and modafinil in a marmoset Parkinson model. European Neuropsychopharmacology. 2008;18:383–389.  [PubMed]
134. de Lago E, Miguel R, Lastres-Becker I, Ramos J, Fernandez-Ruiz J. Involvement of vanilloid-like receptors in the effects of anandamide on motor behavior and nigrostriatal dopaminergic activity: in vivo and in vitro evidence. Brain Research. 2004;1007:152–159.  [PubMed]
135. Dowie MJ, Howard ML, Nicholson LFB, Faull RLM, Hannan AJ, Glass M. Behavioural and molecular consequences of chronic cannabinoid treatment in Huntington’s disease transgenic mice. Neuroscience. 2010;170:324–336.  [PubMed]
136. Cabranes A, Pryce G, Baker D, Fernandez-Ruiz J. Changes in CB1 receptors in motor-related brain structures of chronic relapsing experimental allergic encephalomyelitis mice. Brain Research. 2006;1107:199–205.  [PubMed]
137. Feigenbaum JJ, Bergman F, Richmond SA, et al. Nonpsychotropic cannabinoid acts as a functional N-methyl-D-aspartate receptor blocker. Proceedings of the National Academy of Sciences. 1989;86:9584–9587. [PMC free article]  [PubMed]
138. Frankel JP, Hughes A, Lees AJ, Stern GM. Marijuana for Pakinsonian tremor. Journal of Neurology, Neurosurgery, and Psychiatry. 1990;53:436–442. [PMC free article]  [PubMed]
139. Chagas MH, Eckeli AL, Zuardi AW, Pena-Pereira MA, Sobreira-Neto MA, Sobreira ET, Camilo MR, Bergamaschi MM, Schenck CH, Hallak JE, Tumas V, Crippa JA. Cannabidiol can improve complex sleep-related behaviours associated with rapid eye movement sleep behaviour disorder in Parkinson’s disease patients: a case series. Journal of Clinical Pharmacy and Therapeutics. 2014;39(5):564–6.  [PubMed]
140. Chagas MH, Zuardi AW, Tumas V, Pena-Pereira MA, Sobreira ET, Bergamaschi MM, Dos Santos AC, Teixeira AL, Hallak JE, Crippa JA. Effects of cannabidiol in the treatment of patients with Parkinson’s disease: An exploratory double-blind trial. Journal of Psychopharacology. 2014;28(11):1088–98.  [PubMed]
141. Martínez-Orgado J, Fernández-López D, Lizasoain I, Romero J. The seek of neuroprotection: introducing cannabinoids. Recent Patents on CNS Drug Discovery. 2007;2(2):131–9.  [PubMed]
142. Thenganatt MA, Jankovic J. Psychogenic tremor: a video guide to its distinguishing features. Tremor Other Hyperkinet Mov (N Y) 2014 Aug 27;4:253. [PMC free article]  [PubMed]
143. Secades-Villa R, Garcia-Rodriguez O, Jin CJ, Wang S, Blanco C. Probability and predictors of the cannabis gateway effect: A national study. International Journal of Drug Policy. 2014 epub ahead of print. [PMC free article]  [PubMed]
144. Anthony JC. Steppingstone and gateway ideas: a discussion of origins, research challenges, and promising lines of research for the future. Drug and Alcohol Dependence. 2012;123(Suppl 1):S99–S104. [PMC free article]  [PubMed]
145. Sagredo O, Ramos JA, Decio A, Mechoulam R, Fernández-Ruiz J. Cannabidiol reduced the striatal atrophy caused 3-nitropropionic acid in vivo by mechanisms independent of the activation of cannabinoid, vanilloid TRPV1 and adenosine A2A receptors. European Journal of Neuroscience. 2007;26(4):843–51.  [PubMed]
146. Lastres-Becker I, de Miguel R, De Petrocellis L, Makriyannis A, Di Marzo V, Fernández-Ruiz J. Compounds acting at the endocannabinoid and/or endovanilloid systems reduce hyperkinesia in a rat model of Huntington’s disease. Journal of Neurochemistry. 2003;84(5):1097–109.  [PubMed]
twin memes II