Skip to main content
Canna~Fangled Abstracts

What Do We Know About Medical Cannabis in Neurological Disorders and What Are the Next Steps?

By April 27, 2022May 16th, 2022No Comments


Journal List > Front Pharmacol > PMC9091192

Abstract

Medical use of cannabis has been receiving growing attention over the last few decades in modern medicine. As we know that the endocannabinoid system is largely involved in neurological disorders, we focused on the scientific rationale of medical cannabis in three neurological disorders: amyotrophic lateral sclerosis, Parkinson’s disease, and Alzheimer’s disease through pharmacological plausibility, clinical studies, and patients’ view. Clinical studies (randomized controlled trials, open-label studies, cohorts, and case reports) exploring medical cannabis in these disorders show different results depending on the methods and outcomes. Some show benefits on motor symptoms and others on non-motor symptoms and quality of life. Concerning patients’ view, several web surveys were collected, highlighting the real use of cannabis to relieve symptoms of neurological disorders, mostly outside a medical pathway. This anarchic use keeps questioning particularly in terms of risks: consumption of street cannabis, drug–drug interactions with usual medical treatment, consideration of medical history, and adverse reactions (psychiatric, respiratory, cardiovascular disorders, etc.), underlining the importance of a medical supervision. To date, most scientific data support the therapeutic potential of cannabis in neurological disorders. As far as patients and patients’ associations are calling for it, there is an urgent need to manage clinical studies to provide stronger evidence and secure medical cannabis use.

Keywords: medical cannabis, neurological disorders, amyotrophic lateral sclerosis, Parkinson, Alzheimer, pharmacology, scientific research

Introduction

Medical use of cannabis has been receiving growing attention over the last few decades in modern medicine. As cannabis is a complex plant containing hundreds of cannabinoids, we keep questioning about its therapeutic benefits, justified by its pleiotropic pharmacological activity. As a result, it has been reported that changes in endocannabinoid levels may be related to neurological diseases such as Parkinson’s disease, Huntington’s disease, Alzheimer’s disease, and multiple sclerosis (). As we know that the endocannabinoid system (ECS) is largely involved in neurological disorders, we here chose to focus on the scientific rationale of medical cannabis through a narrative review in three neurological disorders: amyotrophic lateral sclerosis (ALS), Parkinson’s disease (PD), and Alzheimer’s disease (AD) through pharmacological plausibility, clinical studies, and patients’ view. Developing medical cannabis could be an important issue to better control neurodegeneration.

Pharmacological Plausibility

ECS is largely expressed in the cerebellum, basal ganglia, and hippocampus and is thus an area of choice for molecular targets. Characterization of the ECS and detection of widespread cannabinoid receptors in the brain and peripheral tissues have opened the door to a vast field of research. The ECS is formed by cannabinoid receptors 1 and 2 (CB1 and CB2), the two endocannabinoids anandamide and 2-arachidonoylglycerol, and endocannabinoid anabolic and catabolic enzymes. Manipulation of the ECS may have beneficial disease-modifying potential in neurological disorders. Exogenous cannabinoids play a pleiotropic activity mostly through two cannabinoid receptors: CB1 is predominantly expressed in the brain, and CB2 is primarily found in the cells of the immune system (). Since the pathophysiology of motor neuron degeneration in ALS may involve mitochondrial dysfunction, excessive glutamate activity, oxidative stress, neuroinflammation, and growth factor deficiency, cannabis could be effective in modulating these processes (). To support these hypotheses, a recent meta-analysis of preclinical studies in murine ALS models conducted by Urbi and colleagues suggests that cannabinoid receptor agonists may improve survival time ().

PD mostly involves dopaminergic and cholinergic systems. The interactions between cannabinoids and dopamine in the basal ganglia may involve both the modulation of other neurotransmitters (GABA, glutamate) and the activation of CB1 and CB2 (). Preclinical studies in the animal model of PD have shown various influences of cannabis on motor and non-motor behaviors: reducing motor fluctuations and levodopa-induced dyskinesias (). Activation of CB2 has shown a reduction in dopamine depletion in PD rats (). In a preclinical study investigating the role of a CB2 receptor agonist on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity in a mouse model of PD managed in 2017, the use of a CB2 agonist reversed the depletion of CB2 and thus increased the levels of dopamine and improved the behavior of PD mice (). Cannabinoids seem to be protective by binding to the CB1 receptor, inhibiting the dopamine beta hydroxylase activity and decreasing glutamate levels or by binding to CB2, reducing neuroinflammation (). All these considerations suggest therapeutic benefits of cannabis in PD.

AD is characterized by extracellular deposits of β-amyloid plaques and neurofibrillary tangles of tau protein (). Cannabis promotes neuroprotection through different signal pathways mediated indirectly by CB receptors by reducing the β-amyloid peptide action and tau phosphorylation, as well as modulating oxidative stress and inflammation (). CB1 and CB2 agonists ameliorated memory and cognitive impairment in mice that have received intracerebral injection of β-amyloid peptide (). CB2 activation also reduced levels of neurotoxic factors and pro-inflammatory mediators produced by reactive astrocytes and microglial cells, stimulated microglial proliferation and migration, and decreased β-amyloid peptide levels (). To resume, cannabis improved immune function, amyloidogenesis, and reduced behavioral symptoms and pain but also stimulated appetite and inhibited acetylcholinesterase in animal models of AD ().

These pharmacological considerations concerning cannabis in neurological disorders suggest mechanism-based therapeutic targets for future clinical studies.

Clinical Studies

We managed a literature search on Medline using the keywords “medical cannabis” and “neurological disorders,” “medical cannabis” and “amyotrophic lateral sclerosis,” “medical cannabis” and “Parkinson,” and “medical cannabis” and “Alzheimer”. The articles were thoroughly screened by reviewing each article with titles, abstracts, and content of the full articles. We only included the studies published between 1986 and 2021 and human studies (clinical trials, case reports, and published protocols) in the English language, including adults of 18 years of age and older, and we excluded review articles and position studies (Figure 1). An additional search on clinicaltrials.gov was also performed using “ALS” and “cannabis,” “Parkinson” and “cannabis,” and “Alzheimer” and “cannabis”.

An external file that holds a picture, illustration, etc.
Object name is fphar-13-883987-g001.jpg

Flow chart of the included clinical studies.

Only sparse data on the benefits of medical cannabis in neurological disorders are available from clinical studies. As we know that cannabis is a complex plant with hundreds of phytocannabinoids, several components are studied in the following clinical studies. Tetrahydrocannabinol (THC) and cannabidiol (CBD) are the most studied in therapeutic use as they are the two major compounds of the Cannabis sativa L. plant. THC could be synthetized from CBD acid extracted from the plant as dronabinol. Nabilone is completely synthetized and is an analog of THC whereas Sativex®, which is a commercialized medication, contains a mix of THC and CBD directly extracted from the plant of cannabis (nabiximol).

To date, we have only found four clinical studies exploring the use of medical cannabis in ALS (). Data from these studies are summarized in Table 1. The use of dronabinol alone did not demonstrate improvement in cramp intensity, cramp frequency, and fasciculation intensity neither on quality of life, sleep, appetite, and depression in a randomized controlled trial (RCT) of 2010 (). The lack of treatment effect could be due to the short duration treatment (2 weeks). In parallel, an equilibrated mix of tetrahydrocannabinol (THC) and cannabidiol (CBD) in the oromucosal spray Sativex® seems to be effective on ALS-related spasticity and on the patients’ global impression of change in a 6-week RCT () and also in a cohort study (). Noticeably, Sativex® (an equilibrated mix of THC and CBD) is already commercialized and indicated for symptom improvement in adult patients with resistant spasticity due to multiple sclerosis. All these studies reported good tolerability of medical cannabis. Therefore, these modest but encouraging results suggest the need for further studies enrolling a higher number of patients.

TABLE 1

Summary of clinical studies exploring medical cannabis in ALS.

Reference Study design Number and the type of patients Molecule explored and the route of administration Dose/frequency duration Outcomes and efficacy Safety
Randomized, double-blind, placebo-controlled crossover study n = 22 ALS patients suffering from daily cramps (completed) Dronabinol, Marinol®; per os 5 mg of dronabinol twice daily during 2 weeks Wash-out: 2 weeks -No significant effect on cramp intensity (primary outcome) Two AEs non–study-related: one pneumonia and one deep venous thrombosis
-No significant effect on number of cramps per day, number of cramps during daytime and bedtime, fasciculations, quality of life, quality of sleep, appetite, and depression (secondary outcomes)
Randomized, double-blind, placebo-controlled study n = 9 ALS patients suffering from cramps Dronabinol; per os 1) 5 mg (single dose) -PK linear with a doubling of the AUC -Drowsiness, euphoria, orthostasis, sleepiness, vertigo, and weakness: significantly more frequent in patients receiving 10 mg THC as compared to 5 mg THC per day
2) wash-out: 2 weeks -High inter-individual PK variability -No association between drug exposure and the occurrence of AE
3) 10 mg (single dose) -Heart rate peaked approximately together with the plasma concentrations of THC-OH
Observational, retrospective, monocentric, cross-sectional cohort study n = 32 ALS patients suffering from spasticity Mix of THC:CBD (2.7 mg:2.5 mg), Sativex®; oromucosal spray three groups: -Severe spasticity related to high doses of Sativex® No AE reported
>7 sprays (n = 11) -High treatment satisfaction
<7 sprays (n = 16) n = 16 discontinued the treatment during observation period
Infrequent use (n = 5)
Randomized, double-blind, placebo-controlled study n = 59 ALS patients Mix of THC:CBD (2.7 mg:2.5 mg), Sativex®; oromucosal spray 14 days titration, duration 6 weeks -Significant reduction of ALS-related spasticity -three temporarily discontinuations of AEs
Sativex®n = 29 -Significant effect of the patient’s impression of change one nausea and anxiety event, one influenza and accidental fall, and one disease progression event
Placebo, n = 30 -No significant reduction of the global impression of change (caregivers and physicians), pain, spasm frequency, sleep, timed 10-m walk, scores on the amyotrophic lateral sclerosis functional rating scale—revised, forced vital capacity, scores on the barthel activities of daily living index, and body mass index -No SAEs

AE, adverse event; ALS, amyotrophic lateral sclerosis; CBD, cannabidiol; SAE, severe adverse event; THC, tetrahydrocannabinol.

Concerning PD patients, eight clinical studies were published (), as shown in Table 2. Medical cannabis could be effective both on motor symptoms (dystonia, dyskinesia, and fluctuations), and non-motor symptoms (anxiety, sleep quality, hallucinations, and disorientation) (). Two studies (one RCT and one case report of five PD patients) show that there is not any reduction of motor and non-motor symptoms. One open-label study shows that there is an improvement of motor and non-motor symptoms only at the highest dose of CBD (400 mg/day for 4 weeks). Case reports of four PD patients show that there is an improvement of the quality of sleep without nightmares and reduction of agitation. Five studies show improvement of motor and non-motor symptoms and quality of life (three open-label studies and two RCTs). Anyway, all studies demonstrate that there are no serious adverse effects. The main limitations to these findings are short study duration and small sample sizes. Another limitation may be due to the low bioavailability of THC and CBD in oral preparations. This means that there is an obvious need for larger well-conducted studies.

TABLE 2

Summary of clinical studies exploring medical cannabis in PD.

Reference Study design Number and the type of patients Molecule explored and the route of administration Dose/frequency duration Outcomes and efficacy Safety
Open-label study n = 5 PD patients with dystonia CBD; per os From 100 mg/day to 600 mg/day increased 100 mg/week, duration 6 weeks Improvements in dystonia, dose-related in a 20–50% range -four hypotension events
-three exacerbations of hypokinesia and/or tremor with a higher dose of CBD
-two dry mouth
-two sedation events
-two lightheadedness
Case report n = 5 PD patients resistant to common therapies Cannabis; smoking 1 g; 2,9% THC, duration not reported No effects in reducing the tremor Drowsiness and mild euphoria
Randomized, double-blind, placebo-controlled crossover study n = 9 PD patients with L-Dopa–induced dyskinesia Nabilone; per os 0,03 mg/kg, duration not reported Significant improvement in L-Dopa dyskinesia -Two AEs with withdrawn: one vertigo and one hypotension
-Other AEs (n = 5 patients): mild sedation, “floating sensation”, dizziness, hyperacusis, partial disorientation, and visual hallucinations
Randomized, double-blind, placebo-controlled crossover study n = 19 PD patients with L-Dopa–induced dyskinesia Mix of THC:CBD (2.5 mg:1.25 mg); per os Max 0.25 mg/kg/day THC, administration twice daily, during 4 weeks -No significant effects reported on UPDRS, Rush, Bail, PDQ-39 scales No SAE reported
Wash-out: 2 weeks -No significant effects in improving the quality of life
Open-label study n = 6 PD patients with at least 3-month-old psychosis CBD; per os 150 mg/day to 400 mg/day, duration 4 weeks -Significant improvements in BPRS and psychotic symptoms, in sleep quality, less hallucinations, and disorientations (PPQ) No AE reported
-Significant improvement in UPDRS and CGI-I
Randomized, double-blind, placebo-controlled study n = 21 PD patients without dementia or psychiatric symptoms and no occasional cannabis consumers CBD; per os 75 mg/day or 300 mg/day, duration 6 weeks Significant difference in PDQ39 between placebo and CBD 300 mg/day groups No AE reported
Case reports n = 4 PD patients with sleep behavioral problems CBD; per os 75 mg/day (n = 3); 300 mg/day (n = 1), duration 6 weeks Four patients described an improvement in sleep behavioral disorders No AE reported
Open-label study n = 22 PD patients Cannabis; smoking 0, 5 g, duration not reported -Significant improvements in UPRDS, in tremor, in rigidity, and bradykinesia -Two AEs: one hypoglycaemia and one dizziness
-Significant improvement in sleep and pain scores

BPRS, Brief Psychiatric Rating Scale; CBD, cannabidiol; CGI-I, clinical global impression–improvement; PD, Parkinson’s disease; PDQ-39, Parkinson’s disease questionnaire; THC, tetrahydrocannabinol; UPDRS, Unified Parkinson’s Disease Rating Scale.

To our knowledge, five RCTs and two open-label studies were published in AD regarding medical cannabis effectiveness and safety (). Results of these studies are available in Table 3. Only dronabinol and nabilone were experimented in AD patients. The benefits published in these studies were improving in agitation, nocturnal motor activity, disturbed behavior, anorexia, and the patient’s global impression of change ().

TABLE 3

Summary of clinical studies exploring medical cannabis in AD.

Reference Study design Number and the type of patients Molecule explored and the route of administration Dose/frequency duration Outcomes and efficacy Safety
Randomized, double-blind, placebo-controlled crossover study n = 15 AD patients with behavioral disorders and anorexia Dronabinol; per os 2.5 mg twice daily, duration 6 weeks -Significant improvement in body weight -Nine tiredness and eight somnolence events
-Significant improvement in the severity of behavioral disorders Seven euphoria events
-Significant improvement in the negative affect score -No SAE reported
Open-label study n = 6 patients in the late stages of dementia and suffering from circadian and behavioral disorders Dronabinol; per os 2.5 mg/day, duration 2 weeks -Significant reduction in the nocturnal motor activity No AE reported
-Significant improvement of the NPI score (agitation, aberrant motor, and night-time behaviors)
-Significant reduction in appetite disturbances and irritability
Randomized, placebo-controlled study n = 24 AD patients suffering from agitated behavior Dronabinol; per os 2.5 mg/day, duration 2 weeks -Significant reduction in nocturnal motor activity No AE reported
-Significant improvement of the NPI score
Randomized, double-blind, placebo-controlled crossover study n = 2 AD patients with night time agitation Dronabinol, Marinol®; per os 2.5 mg/day, duration 2 weeks -Reduction in nocturnal motor activity No AE reported
Randomized, double-blind, placebo-controlled crossover study n = 10 patients suffering from dementia THC, Namisol®; per os -Weeks 1–6: 0.75 mg twice daily -Two AEs at 0.75 mg: one dizziness and one fatigue
-Weeks 7–12: 1.5 mg twice daily -Four AEs at 1.5 mg: three agitation and one fatigue
Wash-out period: 4 days -No SAEs
Duration: 12 weeks
Open-label study n = 11 patients with dementia and NPS THC; per os -2.5 mg twice daily -Significant improvement in CGI and NPI scores (delusions, agitation/aggression, irritability, apathy, sleep, and caregiver distress) Three AEs: one fall, one confusion, and one dysphagia
-5 mg twice daily if 2.5 mg ineffective
-7.5 mg twice daily if 5 mg ineffective
Duration: 4 weeks
Randomized, double-blind, placebo-controlled crossover study n = 39 AD patients suffering from agitation Nabilone; per os 1 mg/day; 1,5 mg/day; 2 mg/day according to tolerance, duration 14 weeks -Significant improvement in agitation (CMAI) -36 AEs: 22 sedation, eight falls, one bradycardia, one myoclonic jerk, one elevated urea level, one rash, one NPS increase, and one dizziness
-Significant improvement in NPI (caregiver distress, behavior)
Wash-out: 1 week -Significant improvement in the sMMSE score -Five SAEs: two lethargy, one death, one high INR, and one myocardial infarction
-Significant improvement in the nutritional status without weight gain

AD, Alzheimer’s disease; AE, adverse event; CGI, clinical global impression of change; CMAI, Cohen-Mansfield agitation inventory; sMMSE, standardized mini-mental status examination; NPI, neuropsychiatric inventory; NPS, neuropsychiatric symptoms; SAE, severe adverse event, THC, tetrahydrocannabinol.

To resume, among these 19 clinical studies, nine were randomized double-blind placebo-controlled designed. Open-label design has inherent limitations of a placebo effect and rater bias. Moreover, as the experimental products and the routes of administration used were different (synthetic or natural and mix of cannabinoids or only one cannabinoid; per os or smoked), it adds an additional difficulty to compare results. According to the experimental product, it could also be difficult to perform a placebo-controlled design because of the conspicuous and characteristic smell of a cannabis cigarette, for example. It still underlines that more well-conducted studies would be necessary to further strengthen evidence of effectiveness. Nevertheless, these results are hopeful for patients suffering from these neurological disorders. Moreover, adverse effects reported with the use of medical cannabis do not seem to be limiting for its clinical use. Reported adverse effects were expected ones compared to the knowledge of cannabis use in general population (drowsiness, euphoria, sleepiness, weakness, dizziness, hypotension, and dry mouth). Due to pharmacokinetics variability of medical cannabis (and its numerous metabolites), future studies should apply parallel group study design rather than crossover design.

To date, 13 studies are registered in clinicaltrials.gov; two protocols are already published in Medline (). Urbi et al. published a protocol of a randomized double-blind placebo-controlled study in ALS patients to evaluate the efficacy of a mix oil of CBD:THC (25 mg CBD: <2 mg THC) in slowing the disease progression. Secondary objectives are safety and tolerability. Timler et al. carried a randomized double-blind crossover study experimenting a mix oil of THC:CBD (3:2) in patients with dementia, on behavior symptoms, quality of life, and discomfort by pain.

Overall, clinical studies exploring medical cannabis in neurological disorders show different results depending on the methods and outcomes. Some show benefits on motor symptoms of neurological diseases, some on non-motor symptoms, and others no benefit at all. Therefore, it is becoming essential to conduct more and larger clinical studies in order to scientifically enlighten clinicians and first and foremost patients.

What About Patients’ View?

As cannabis has been presented as a treatment for many medical conditions for few years, patients experiment this plant in many ways to manage their neurological disorders. Nevertheless, very few surveys have been conducted to describe 1) the consumers (medical condition and demographics); 2) the consumption (the cannabinoid type, form, route of administration, frequency, duration, and way of acquisition); 3) the relief symptoms (duration and level of the relief); 4) the adverse effects (type, duration, and frequency). It is unavoidable to understand the motivations and experiences of cannabis use among people living with neurological disorders to better orient clinical trials.

In 2004, Amtmann and colleagues published a worldwide anonymous web survey analyzing the answers of 131 ALS patients (). The mean age was 54 years, and patients were mostly male (75%). Respondents reported a stable family life and a high education level for the majority. The median time since ALS diagnosis was 3 years, and the mean duration was 4 years. About 10% of the respondents (n = 13) reported the use of cannabis to relieve symptoms of ALS in the last 12 months. They mostly consume smoking cannabis. Only three of them reported using medical cannabis (dronabinol). Concerning relieve symptoms, patients reported cannabis as moderately effective in symptoms of appetite loss, depression, pain, spasticity, and drooling and ineffective in reducing difficulties with speech and swallowing and sexual dysfunction. The longest relief was reported for depression. In 2004, Venderova and colleagues also sent an anonymous questionnaire to all patients attending the Prague Movement Disorder Centre (). In total, 339 questionnaires were returned, and 25% of the respondents declared having taken cannabis. The mean age of cannabis users was 63.9 years, and the mean duration of PD was 8.3 years. They mostly reported an oral consumption once a day. Interestingly, none of them reported their doctor. PD patients described alleviations, especially in motor symptoms: 44.7% in bradykinesia, 37.7% in muscle rigidity, 30% in tremor, and 14.1% in L-dopa-induced dyskinesias. Another anonymous web survey managed on PD patients in the United States in 2020 analyzed the answers of 1,064 patients (). The mean age of the respondents was 71.2 years, male accounted for 52.5%, and the mean PD duration was 7.4 years. They were mostly highly educated with 78% of retired, evolving in a stable family life. About 25% of them reported the use of cannabis in the previous 6 months, and 35.6% of them considered themselves as regular users. They most frequently reported spraying or drooping, smoking, and eating as their primary method of cannabis use. The ways of acquisition were medical dispensary (38.7%) and family/friend gift (24.5%). When known, patients reported products with a high THC dosage in 21.2%, to get a better efficacy for both motor and non-motor symptoms. The reported relief symptoms were non-motor symptoms insufficiently controlled by classic medications: anxiety (45.5%), pain (44%), sleep disorders (44%), and specific motor symptoms such as stiffness (43%) or tremor (42%). Only 12.6% of PD cannabis users reported adverse effects (anxiety, impaired coordination, and dizziness). Interestingly, cannabis non-users (75.5%) reported two major reasons for not using cannabis: the lack of evidence (59.9%) and the fear of cannabis adverse effects (34.9%). In 2021, a German nationwide questionnaire survey described the used of medical cannabis in PD patients (). A total of 1,348 questionnaires were analyzed. The mean age of the patients was 71.6 years, and the mean PD duration was 11.6 years. Cannabis use was reported in 8.4% of the questionnaires, with a reduction of pain and muscle cramps in more than 40% of users (respectively, 43.9 and 41.4%). Moreover, more than 20% of them described an improvement in depression (28.1%), stiffness/akinesia (27.3%), sleep disorders (27.1%), freezing (25.0%), tremor (24.3%), anxiety (24.0%), and restless legs syndrome (21.4%). The improvements were related to 54.1% of oral CBD use and to 68.2% inhaling THC-containing cannabis. In the majority of patients (85%), cannabis was well-tolerated. Adverse effects reported were mainly fatigue, dizziness, and ravenous appetite. Another recent survey showed that 95% of movement disorders specialist neurologists reported to be asked to prescribe medical cannabis to their patients ().

Concerning AD patients, a recent Polish anonymous web survey addressed to caregivers identified the attitudes and beliefs of caregivers of individuals with AD toward CBD oil in Poland (). A total of 73 caregivers answered the questionnaire. They reported an effective use of CBD oil in behavioral symptoms of AD, to slow memory loss, agitation, anxiety, and insomnia. Most of the caregivers (84%) answered that CBD oil improved their care recipient’s quality of life. None of them reported adverse effects with the use of CBD oil. It is also interesting to note that only 63% of them informed their physician about this habit. In this survey, people also reported lack of information about the legacy, the medical use of cannabis as far as a lack of scientific data.

Despite being great sources of information, these surveys present several limitations. First, the results are based on small sample sizes compared to the affected population; respondents may not have been representative of the entire ALS, PD, and AD population. Second, internet users’ population constitutes a selection bias because all patients with neurological disorders could not use the internet, and internet users tend to be highly educated. Third, cannabis users may have been more inclined to answer the surveys and inflated the number of users and benefits, leading to a possible answer bias. Another limitation is the country of survey and/or residence because cannabis could be legal or not.

What Are the Main Risks of Cannabis Use in Neurological Disorders?

The main risk is that most of the therapeutic uses of cannabis are performed outside of a medical pathway exposing patients to uncontrolled drugs (street cannabis) and unexpected drug–drug interactions. Published case reports show CBD interactions with antiepileptic drugs (), warfarin (), immunosuppressants such as tacrolimus (), and methadone (). In all these case reports, the consequence of the drug–drug interaction was an increase in plasma concentrations of co-administered medications and potential associated complications. Cannabis could also cause well-known psychiatric adverse effects (psychosis, paranoia, anxiety, disorientation, etc.). Therefore, it is important to supervise and regulate the consumption of medical cannabis. In the majority of clinical studies exploring medical cannabis, exclusion criteria included history of psychiatric disorders (). In addition to the respiratory adverse effects, cardiovascular complications are poorly known but also reported with the use of cannabis. Jouanjus et al. conducted an observational retrospective study in 2011 in patients admitted to a French hospital with a relation of cannabis use. In total, 200 patients were included, and 619 adverse effects were reported with 9.5% of cardiovascular ones (). Serious cardiovascular complications described with the use of cannabis are arrythmia including ventricular tachycardia, acute coronary syndromes, peripheral complications (arteriopathies), and cerebral complications (acute cerebral angiopathy, transient cortical blindness, and spasm of the cerebral artery) (). There is a lack of consensus that likely reflects a general knowledge gap and paucity of data to guide clinical practice. Nevertheless, it is essential to supervise cannabis consumption and consider the medical history and the concomitant medication use in these patients to avoid serious complications.

Discussion/Conclusion

In recent decades, the endocannabinoid system has attracted considerable interest as a potential therapeutic target in numerous pathological conditions. Medical cannabis has clearly demonstrated several benefits on neurological disorders, owing to its pleiotropic pharmacological activity. Preclinical, clinical, and real-life experiences described in ALS, PD, and AD are even more important cues to develop research on medical cannabis. Acceptable safety and tolerability profiles are also strong arguments to be considered in the development of medical cannabis. It is now essential to answer several questions to broadly develop medical cannabis in neurological disorders: 1) which cannabinoids (THC, CBD, mix of THC and CBD, and others)? 2) What dosage? 3) What frequency? 4) Which route of administration? 5) In which symptoms? Answers to these questions will be helpful for patients and clinicians to manage care pathways with medical cannabis treatment.

As all pharmacological substance and a fortiori with its pleiotropic activity, clinicians should be cautious with the use of medical cannabis because of drug–drug interactions and with the medical history of their patients. Only few patients inform their clinicians of cannabis experience, and very few data are available on these interactions.

Nevertheless, there is a call for more clinical studies to secure cannabis consumption in a medical enrollment. As far as patients and patients’ associations are calling for it, there is an urgent need to manage clinical studies to provide stronger evidence and secure medical cannabis use. Therefore, more controlled clinical studies with larger neuropsychiatric populations should be prioritized to bring important cues in the near future and support the translation of research findings to clinical settings.

Author Contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

  • Ahmed A. I., van den Elsen G. A., Colbers A., Kramers C., Burger D. M., van der Marck M. A., et al. (2015). Safety, Pharmacodynamics, and Pharmacokinetics of Multiple Oral Doses of delta-9-tetrahydrocannabinol in Older Persons with DementiaPsychopharmacology (Berl) 232, 2587–2595. 10.1007/s00213-015-3889-y [PMC free article] [PubMed] [CrossRef[]
  • Amtmann D., Weydt P., Johnson K. L., Jensen M. P., Carter G. T. (2004). Survey of Cannabis Use in Patients with Amyotrophic Lateral SclerosisAm. J. Hosp. Palliat. Care 21, 95–104. 10.1177/104990910402100206 [PubMed] [CrossRef[]
  • Anderson L. L., Absalom N. L., Abelev S. V., Low I. K., Doohan P. T., Martin L. J., et al. (2019). Coadministered Cannabidiol and Clobazam: Preclinical Evidence for Both Pharmacodynamic and Pharmacokinetic InteractionsEpilepsia 60, 2224–2234. 10.1111/epi.16355 [PMC free article] [PubMed] [CrossRef[]
  • Appel S. H., Zhao W., Beers D. R., Henkel J. S. (2011). The Microglial-Motoneuron Dialogue in ALSActa Myol 30, 4–8. [PMC free article] [PubMed[]
  • Aso E., Ferrer I. (2014). Cannabinoids for Treatment of Alzheimer’s Disease: Moving toward the ClinicFront. Pharmacol. 5, 37. 10.3389/fphar.2014.00037 [PMC free article] [PubMed] [CrossRef[]
  • Bega D., Simuni T., Okun M. S., Chen X., Schmidt P. (2017). Medicinal Cannabis for Parkinson’s Disease: Practices, Beliefs, and Attitudes Among Providers at National Parkinson Foundation Centers of ExcellenceMov Disord. Clin. Pract. 4, 90–95. 10.1002/mdc3.12359 [PMC free article] [PubMed] [CrossRef[]
  • Bilsland L. G., Greensmith L. (2008). The Endocannabinoid System in Amyotrophic Lateral SclerosisCurr. Pharm. Des. 14, 2306–2316. 10.2174/138161208785740081 [PubMed] [CrossRef[]
  • Carroll C. B., Bain P. G., Teare L., Liu X., Joint C., Wroath C., et al. (2004). Cannabis for Dyskinesia in Parkinson Disease: A Randomized Double-Blind Crossover StudyNeurology 63, 1245–1250. 10.1212/01.WNL.0000140288.48796.8E [PubMed] [CrossRef[]
  • Carter G. T., Abood M. E., Aggarwal S. K., Weiss M. D. (2010). Cannabis and Amyotrophic Lateral Sclerosis: Hypothetical and Practical Applications, and a Call for Clinical TrialsAm. J. Hosp. Palliat. Care 27, 347–356. 10.1177/1049909110369531 [PubMed] [CrossRef[]
  • Chagas M. H., Eckeli A. L., Zuardi A. W., Pena-Pereira M. A., Sobreira-Neto M. A., Sobreira E. T., et al. (2014a). Cannabidiol Can Improve Complex Sleep-Related Behaviours Associated with Rapid Eye Movement Sleep Behaviour Disorder in Parkinson’s Disease Patients: a Case SeriesJ. Clin. Pharm. Ther. 39, 564–566. 10.1111/jcpt.12179 [PubMed] [CrossRef[]
  • Chagas M. H., Zuardi A. W., Tumas V., Pena-Pereira M. A., Sobreira E. T., Bergamaschi M. M., et al. (2014b). Effects of Cannabidiol in the Treatment of Patients with Parkinson’s Disease: an Exploratory Double-Blind TrialJ. Psychopharmacol. 28, 1088–1098. 10.1177/0269881114550355 [PubMed] [CrossRef[]
  • Collin C., Davies P., Mutiboko I. K., Ratcliffe S., and for the Sativex Spasticity in MS Study Group (2007). Randomized Controlled Trial of Cannabis-Based Medicine in Spasticity Caused by Multiple SclerosisEur. J. Neurol. 14, 290–296. 10.1111/j.1468-1331.2006.01639.x [PubMed] [CrossRef[]
  • Consroe P., Sandyk R., Snider S. R. (1986). Open Label Evaluation of Cannabidiol in Dystonic Movement DisordersInt. J. Neurosci. 30, 277–282. 10.3109/00207458608985678 [PubMed] [CrossRef[]
  • Cooray R., Gupta V., Suphioglu C. (2020). Current Aspects of the Endocannabinoid System and Targeted THC and CBD Phytocannabinoids as Potential Therapeutics for Parkinson’s and Alzheimer’s Diseases: a ReviewMol. Neurobiol. 57, 4878–4890. 10.1007/s12035-020-02054-6 [PMC free article] [PubMed] [CrossRef[]
  • Cristino L., Bisogno T., Di Marzo V. (2020). Cannabinoids and the Expanded Endocannabinoid System in Neurological DisordersNat. Rev. Neurol. 16, 9–29. 10.1038/s41582-019-0284-z [PubMed] [CrossRef[]
  • Esposito G., De Filippis D., Carnuccio R., Izzo A. A., Iuvone T. (2006). The Marijuana Component Cannabidiol Inhibits Beta-Amyloid-Induced Tau Protein Hyperphosphorylation through Wnt/beta-Catenin Pathway rescue in PC12 CellsJ. Mol. Med. (Berl) 84, 253–258. 10.1007/s00109-005-0025-1 [PubMed] [CrossRef[]
  • Feeney M. P., Bega D., Kluger B. M., Stoessl A. J., Evers C. M., De Leon R., et al. (2021). Weeding through the Haze: a Survey on Cannabis Use Among People Living with Parkinson’s Disease in the USNpj Parkinsons Dis. 7, 21. 10.1038/s41531-021-00165-y [PMC free article] [PubMed] [CrossRef[]
  • Ferreira C., Almeida C., Tenreiro S., Quintas A. (2020). Neuroprotection or Neurotoxicity of Illicit Drugs on Parkinson’s DiseaseLife (Basel) 10, E86. 10.3390/life10060086 [PMC free article] [PubMed] [CrossRef[]
  • Fraguas-Sánchez A. I., Torres-Suárez A. I. (2018). Medical Use of CannabinoidsDrugs 78, 1665–1703. 10.1007/s40265-018-0996-1 [PubMed] [CrossRef[]
  • Frankel J. P., Hughes A., Lees A. J., Stern G. M. (1990). Marijuana for Parkinsonian TremorJ. Neurol. Neurosurg. Psychiatry 53, 436. 10.1136/jnnp.53.5.436 [CrossRef[]
  • García-Arencibia M., González S., de Lago E., Ramos J. A., Mechoulam R., Fernández-Ruiz J. (2007). Evaluation of the Neuroprotective Effect of Cannabinoids in a Rat Model of Parkinson’s Disease: Importance of Antioxidant and Cannabinoid Receptor-independent PropertiesBrain Res. 1134, 162–170. 10.1016/j.brainres.2006.11.063 [PubMed] [CrossRef[]
  • Gilmartin C. G. S., Dowd Z., Parker A. P. J., Harijan P. (2021). Interaction of Cannabidiol with Other Antiseizure Medications: A Narrative ReviewSeizure 86, 189–196. 10.1016/j.seizure.2020.09.010 [PubMed] [CrossRef[]
  • Goyal H., Awad H. H., Ghali J. K. (2017). Role of Cannabis in Cardiovascular DisordersJ. Thorac. Dis. 9, 2079–2092. 10.21037/jtd.2017.06.104 [PMC free article] [PubMed] [CrossRef[]
  • Grayson L., Vines B., Nichol K., Szaflarski J. P. UAB CBD Program (2018). An Interaction between Warfarin and Cannabidiol, a Case ReportEpilepsy Behav. Case Rep. 9, 10–11. 10.1016/j.ebcr.2017.10.001 [PMC free article] [PubMed] [CrossRef[]
  • Herrmann N., Ruthirakuhan M., Gallagher D., Verhoeff N. P. L. G., Kiss A., Black S. E., et al. (2019). Randomized Placebo-Controlled Trial of Nabilone for Agitation in Alzheimer’s DiseaseAm. J. Geriatr. Psychiatry 27, 1161–1173. 10.1016/j.jagp.2019.05.002 [PubMed] [CrossRef[]
  • Joerger M., Wilkins J., Fagagnini S., Baldinger R., Brenneisen R., Schneider U., et al. (2012). Single-dose Pharmacokinetics and Tolerability of Oral delta-9- Tetrahydrocannabinol in Patients with Amyotrophic Lateral SclerosisDrug Metab. Lett. 6, 102–108. 10.2174/1872312811206020102 [PubMed] [CrossRef[]
  • Jouanjus E., Leymarie F., Tubery M., Lapeyre-Mestre M. (2011). Cannabis-related Hospitalizations: Unexpected Serious Events Identified through Hospital DatabasesBr. J. Clin. Pharmacol. 71, 758–765. 10.1111/j.1365-2125.2010.03897.x [PMC free article] [PubMed] [CrossRef[]
  • Jouanjus E., Lapeyre‐Mestre M., Micallef J. The French Association of the Regional Abuse and Dependence Monitoring Centres (CEIP‐A) Working Group on Cannabis Complications (2014). Cannabis Use: Signal of Increasing Risk of Serious Cardiovascular DisordersJ. Am. Heart Assoc. 3, 638. 10.1161/JAHA.113.000638 [CrossRef[]
  • Leino A. D., Emoto C., Fukuda T., Privitera M., Vinks A. A., Alloway R. R. (2019). Evidence of a Clinically Significant Drug-Drug Interaction between Cannabidiol and TacrolimusAm. J. Transpl. 19, 2944–2948. 10.1111/ajt.15398 [CrossRef[]
  • Leszko M., Meenrajan S. (2021). Attitudes, Beliefs, and Changing Trends of Cannabidiol (CBD) Oil Use Among Caregivers of Individuals with Alzheimer’s DiseaseComplement. Ther. Med. 57, 102660. 10.1016/j.ctim.2021.102660 [PubMed] [CrossRef[]
  • Li H., Liu Y., Tian D., Tian L., Ju X., Qi L., et al. (2020). Overview of Cannabidiol (CBD) and its Analogues: Structures, Biological Activities, and Neuroprotective Mechanisms in Epilepsy and Alzheimer’s DiseaseEur. J. Med. Chem. 192, 112163. 10.1016/j.ejmech.2020.112163 [PubMed] [CrossRef[]
  • Lotan I., Treves T. A., Roditi Y., Djaldetti R. (2014). Cannabis (Medical Marijuana) Treatment for Motor and Non-motor Symptoms of Parkinson Disease: an Open-Label Observational StudyClin. Neuropharmacol 37, 41–44. 10.1097/WNF.0000000000000016 [PubMed] [CrossRef[]
  • Lucas C. J., Galettis P., Schneider J. (2018). The Pharmacokinetics and the Pharmacodynamics of CannabinoidsBr. J. Clin. Pharmacol. 84, 2477–2482. 10.1111/bcp.13710 [PMC free article] [PubMed] [CrossRef[]
  • Madden K., Tanco K., Bruera E. (2020). Clinically Significant Drug-Drug Interaction between Methadone and CannabidiolPediatrics 145, e20193256. 10.1542/peds.2019-3256 [PubMed] [CrossRef[]
  • Mahlberg R., Walther S. (2007). Actigraphy in Agitated Patients with Dementia. Monitoring Treatment OutcomesZ. Gerontol. Geriatr. 40, 178–184. 10.1007/s00391-007-0420-z [PubMed] [CrossRef[]
  • Meyer T., Funke A., Münch C., Kettemann D., Maier A., Walter B., et al. (2019). Real World Experience of Patients with Amyotrophic Lateral Sclerosis (ALS) in the Treatment of Spasticity Using Tetrahydrocannabinol:cannabidiol (THC:CBD)BMC Neurol. 19, 222. 10.1186/s12883-019-1443-y [PMC free article] [PubMed] [CrossRef[]
  • Morgese M. G., Cassano T., Cuomo V., Giuffrida A. (2007). Anti-dyskinetic Effects of Cannabinoids in a Rat Model of Parkinson’s Disease: Role of CB(1) and TRPV1 ReceptorsExp. Neurol. 208, 110–119. 10.1016/j.expneurol.2007.07.021 [PMC free article] [PubMed] [CrossRef[]
  • Papadimitriou D., Le Verche V., Jacquier A., Ikiz B., Przedborski S., Re D. B. (2010). Inflammation in ALS and SMA: Sorting Out the Good from the EvilNeurobiol. Dis. 37, 493–502. 10.1016/j.nbd.2009.10.005 [PMC free article] [PubMed] [CrossRef[]
  • Patricio F., Morales-Andrade A. A., Patricio-Martínez A., Limón I. D. (2020). Cannabidiol as a Therapeutic Target: Evidence of its Neuroprotective and Neuromodulatory Function in Parkinson’s DiseaseFront. Pharmacol. 11, 595635. 10.3389/fphar.2020.595635 [PMC free article] [PubMed] [CrossRef[]
  • Ramírez B. G., Blázquez C., Gómez del Pulgar T., Guzmán M., de Ceballos M. L. (2005). Prevention of Alzheimer’s Disease Pathology by Cannabinoids: Neuroprotection Mediated by Blockade of Microglial ActivationJ. Neurosci. 25, 1904–1913. 10.1523/JNEUROSCI.4540-04.2005 [PMC free article] [PubMed] [CrossRef[]
  • Riva N., Mora G., Sorarù G., Lunetta C., Ferraro O. E., Falzone Y., et al. (2019). Safety and Efficacy of Nabiximols on Spasticity Symptoms in Patients with Motor Neuron Disease (CANALS): a Multicentre, Double-Blind, Randomised, Placebo-Controlled, Phase 2 TrialLancet Neurol. 18, 155–164. 10.1016/S1474-4422(18)30406-X [PubMed] [CrossRef[]
  • Segovia G., Mora F., Crossman A. R., Brotchie J. M. (2003). Effects of CB1 Cannabinoid Receptor Modulating Compounds on the Hyperkinesia Induced by High-Dose Levodopa in the Reserpine-Treated Rat Model of Parkinson’s DiseaseMov Disord. 18, 138–149. 10.1002/mds.10312 [PubMed] [CrossRef[]
  • Selkoe D. J. (2011). Alzheimer’s DiseaseCold Spring Harb Perspect. Biol. 3, a004457. 10.1101/cshperspect.a004457 [PMC free article] [PubMed] [CrossRef[]
  • Shelef A., Barak Y., Berger U., Paleacu D., Tadger S., Plopsky I., et al. (2016). Safety and Efficacy of Medical Cannabis Oil for Behavioral and Psychological Symptoms of Dementia: An-Open Label, Add-On, Pilot StudyJ. Alzheimers Dis. 51, 15–19. 10.3233/JAD-150915 [PubMed] [CrossRef[]
  • Shi J., Cai Q., Zhang J., He X., Liu Y., Zhu R., et al. (2017). AM1241 Alleviates MPTP-Induced Parkinson’s Disease and Promotes the Regeneration of DA Neurons in PD MiceOncotarget 8, 67837–67850. 10.18632/oncotarget.18871 [PMC free article] [PubMed] [CrossRef[]
  • Sieradzan K. A., Fox S. H., Hill M., Dick J. P., Crossman A. R., Brotchie J. M. (2001). Cannabinoids Reduce Levodopa-Induced Dyskinesia in Parkinson’s Disease: a Pilot StudyNeurology 57, 2108–2111. 10.1212/WNL.57.11.2108 [PubMed] [CrossRef[]
  • Song L., Yang X., Ma Y., Wu N., Liu Z. (2014). The CB1 Cannabinoid Receptor Agonist Reduces L-DOPA-Induced Motor Fluctuation and ERK1/2 Phosphorylation in 6-OHDA-Lesioned RatsDrug Des. Devel Ther. 8, 2173–2179. 10.2147/DDDT.S60944 [CrossRef[]
  • Stampanoni Bassi M., Sancesario A., Morace R., Centonze D., Iezzi E. (2017). Cannabinoids in Parkinson’s DiseaseCannabis Cannabinoid Res. 2, 21–29. 10.1089/can.2017.0002 [PMC free article] [PubMed] [CrossRef[]
  • Timler A., Bulsara C., Bulsara M., Vickery A., Smith J., Codde J. (2020). Use of Cannabinoid-Based Medicine Among Older Residential Care Recipients Diagnosed with Dementia: Study Protocol for a Double-Blind Randomised Crossover TrialTrials 21, 188. 10.1186/s13063-020-4085-x [PMC free article] [PubMed] [CrossRef[]
  • Urbi B., Broadley S., Bedlack R., Russo E., Sabet A. (2019a). Study Protocol for a Randomised, Double-Blind, Placebo-Controlled Study Evaluating the Efficacy of Cannabis-Based Medicine Extract in Slowing the Disease pRogression of Amyotrophic Lateral Sclerosis or Motor Neurone Disease: the EMERALD TrialBMJ Open 9, e029449. 10.1136/bmjopen-2019-029449 [CrossRef[]
  • Urbi B., Owusu M. A., Hughes I., Katz M., Broadley S., Sabet A. (2019b). Effects of Cannabinoids in Amyotrophic Lateral Sclerosis (ALS) Murine Models: a Systematic Review and Meta-AnalysisJ. Neurochem. 149, 284–297. 10.1111/jnc.14639 [PubMed] [CrossRef[]
  • van de Donk T., Niesters M., Kowal M. A., Olofsen E., Dahan A., van Velzen M. (2019). An Experimental Randomized Study on the Analgesic Effects of Pharmaceutical-Grade Cannabis in Chronic Pain Patients with FibromyalgiaPain 160, 860–869. 10.1097/j.pain.0000000000001464 [PMC free article] [PubMed] [CrossRef[]
  • Venderová K., Růžička E., Voříšek V., Višňovský P. (2004). Survey on Cannabis Use in Parkinson’s Disease: Subjective Improvement of Motor SymptomsMov. Disord. 19, 1102–1106. 10.1002/mds.20111 [PubMed] [CrossRef[]
  • Volicer L., Stelly M., Morris J., McLaughlin J., Volicer B. J. (1997). Effects of Dronabinol on Anorexia and Disturbed Behavior in Patients with Alzheimer’s DiseaseInt. J. Geriatr. Psychiatry 12, 913–919. 10.1002/(sici)1099-1166(199709)12:9<913::aid-gps663>3.0.co;2-d [PubMed] [CrossRef[]
  • Wallace M. S., Marcotte T. D., Umlauf A., Gouaux B., Atkinson J. H. (2015). Efficacy of Inhaled Cannabis on Painful Diabetic NeuropathyJ. Pain 16, 616–627. 10.1016/j.jpain.2015.03.008 [PMC free article] [PubMed] [CrossRef[]
  • Walther S., Mahlberg R., Eichmann U., Kunz D. (2006). Delta-9-tetrahydrocannabinol for Nighttime Agitation in Severe DementiaPsychopharmacology (Berl) 185, 524–528. 10.1007/s00213-006-0343-1 [PubMed] [CrossRef[]
  • Walther S., Schüpbach B., Seifritz E., Homan P., Strik W. (2011). Randomized, Controlled Crossover Trial of Dronabinol, 2.5 Mg, for Agitation in 2 Patients with DementiaJ. Clin. Psychopharmacol. 31, 256–258. 10.1097/JCP.0b013e31820e861c [PubMed] [CrossRef[]
  • Weber M., Goldman B., Truniger S. (2010). Tetrahydrocannabinol (THC) for Cramps in Amyotrophic Lateral Sclerosis: a Randomised, Double-Blind Crossover TrialJ. Neurol. Neurosurg. Psychiatry 81, 1135–1140. 10.1136/jnnp.2009.200642 [PubMed] [CrossRef[]
  • Yenilmez F., Fründt O., Hidding U., Buhmann C. (2021). Cannabis in Parkinson’s Disease: The Patients’ ViewJ. Parkinsons Dis. 11, 309–321. 10.3233/JPD-202260 [PubMed] [CrossRef[]
  • Zuardi A. W., Crippa J. A., Hallak J. E., Pinto J. P., Chagas M. H., Rodrigues G. G., et al. (2009). Cannabidiol for the Treatment of Psychosis in Parkinson’s DiseaseJ. Psychopharmacol. 23, 979–983. 10.1177/0269881108096519 [PubMed] [CrossRef[]

Articles from Frontiers in Pharmacology are provided here courtesy of Frontiers Media SA

Leave a Reply