Open access peer-reviewed chapter - ONLINE FIRST

The Endocannabinoid System as a Potential Therapeutic Target for Amyotrophic Lateral Sclerosis

Written By

Kamila Saramak and Natalia Szejko

Submitted: 03 July 2023 Reviewed: 29 February 2024 Published: 27 March 2024

DOI: 10.5772/intechopen.114388

Motor Neurons - New Insights IntechOpen
Motor Neurons - New Insights Edited by Natalia Szejko

From the Edited Volume

Motor Neurons - New Insights [Working Title]

M.D. Natalia Szejko and M.D. Kamila Saramak

Chapter metrics overview

14 Chapter Downloads

View Full Metrics

Abstract

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by a selective loss of motor neurons from the spinal cord, brainstem and motor cortex. With a prevalence of about 5.5–9.9 per 100,000 persons, ALS is the most common form of motor neuron disease (MND). Although the mechanisms underlying the pathophysiology of this condition are not yet fully understood, it is believed that excitotoxicity, inflammation and oxidative stress play an important role in selective motor neuron death. Despite intensive research, up to this point no cure for ALS has been identified. There is increasing evidence that cannabinoids, due to their anti-glutamatergic and anti-inflammatory actions, may show neuroprotective effects in ALS patients and slow the progression of the disease. Furthermore, cannabis-based medicine may be useful in managing symptoms like pain, spasticity or weight loss. The aim of this chapter is to summarize the current state of research regarding the efficacy and safety of medical cannabis in the treatment of ALS.

Keywords

  • motor neuron disease
  • amyotrophic lateral sclerosis
  • neuroprotection
  • endocannabinoids
  • cannabis

1. Introduction

Amyotrophic lateral sclerosis (ALS) is a multisystem neurodegenerative disease leading to the progressive degradation of both upper (UMN) and lower motor neurons (LMN). With its prevalence of about 5.5–9.9 per 100,000 persons, ALS is the most common form of motor neuron disease (MND) [1]. The condition affects primarily the pyramidal motor system, including the motor cortex, cranial nerve motor nuclei and spinal cord motor neurons. The mean age of onset of sporadic ALS is about 60 years with a higher incidence among males [2]. The majority of ALS patients present with limb onset of the disease, resulting in focal muscle weakness and atrophy. Over time, due to the damage of the upper motor neurons, spasticity develops. On the other hand, patients with bulbar onset of ALS initially report dysarthria and dysphagia. The limb symptoms may occur almost simultaneously with bulbar symptoms, and in most cases develop within 1–2 years [3]. Subsequently, the disease involves various body regions, in particular respiratory muscles, causing death within 2–3 years for bulbar onset cases and 3–5 years for limb onset cases [4]. The neurodegeneration progresses in the neighboring cortical regions, including the prefrontal cortex, ventral and medial frontal cortical areas, and eventually involves portions of the parietal and temporal lobes and the deep gray structures. This results in non-motor symptoms of the disease such as impairment of executive functions, behavioral changes and language disorders in up to 50% of cases [5]. The cognitive decline in 10–15% of ALS patients is consistent with the diagnosis of frontotemporal dementia (FTD) [6]. To date, the only therapy widely approved for ALS is an anti-glutamate agent riluzole. It has been proven that riluzole slows the progression of ALS, extends survival and delays the time to tracheostomy. The benefit is, however, very limited and riluzole can extend the average survival time by only 3 months [7, 8]. Another drug, edaravone, was first approved in Japan, followed by South Korea, U.S., Canada, Switzerland, and China. Edaravone is a free radical scavenger that can reduce oxidative stress. Its beneficial clinical effects have been initially proven in Japan [9]. The subsequent study in the USA has showed that an intravenous treatment with edaravone prolonged the survival for 6 months [10]. These findings, however, have not been confirmed in the European cohorts [11, 12]. The third drug, AMX0035 has been approved for the treatment of ALS in the USA and Canada. AMX0035 is a combination of two compounds –tauroursodeoxycholic acid (TUDCA) and sodium phenylbutyrate (PB). It is thought to increase the threshold for cell death by blocking key cell death pathways and reducing the stress on the endoplasmic reticulum (ER) simultaneously [13]. The first randomized, placebo-controlled, phase 2 trial of AMX0035 in ALS (CENTAUR) has shown both functional and survival benefits in ALS patients [14]. Moreover, there are many experimental therapies in development, most of them showing anti-inflammatory and anti- excitotoxic mechanisms of action [15].

Advertisement

2. Possible role of the endocannabinoid system in the motor neuron disease

In recent decades, the endocannabinoid system has been gaining increasing attention as a potential therapeutic target in a number of neurological disorders [16, 17]. Thus far, two cannabinoid receptors (CB) have been identified: the CB receptor type 1 (CB1) and CB receptor type 2 (CB2), both belonging to the G protein-coupled receptors (GPCRs) family. The CB1 is expressed primarily in the central nervous system (CNS), especially in the neocortex, hippocampus, basal ganglia, cerebellum, and brainstem [18]. Apart from the CNS, CB1 has also been identified in numerous peripheral tissues and cell types [19]. On the other hand, the CB2 receptor is abundantly present in the immune system and in smaller quantities in the CNS, especially in human microglia [20].

Although a wide range of dysfunctional cellular processes related to neurodegeneration in ALS has been described, the exact mechanisms underlying its pathogenesis remain largely unknown. Mutations in more than 30 different genes associated with diverse molecular functions has been linked to ALS, which explains approximately 20% cases of the disease [21]. A process of excessive activation of glutamate receptors, resulting in neuronal dysfunction and death called “excitotoxicity” is believed to play an important role in the pathogenesis of many neurological disorders, including ALS [22]. Elevated extracellular glutamate levels activate postsynaptic glutamate receptors, causing an increased influx of calcium into the postsynaptic neurons. Subsequently, excessive postsynaptic calcium activates neurotoxic cascades, resulting in neuronal death. The activation of N-methyl-D-aspartate (NMDA) receptors may lead to cellular death more rapidly than the activation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic (AMPA) and kainite receptors [23]. Studies in vitro and in vivo have shown that activation of cannabinoid receptors may inhibit neuronal hyperexcitability associated with activation of both NMDA and non-NMDA subtypes of glutamate receptors [24, 25, 26, 27]. Similarly, the potent anti-inflammatory properties of cannabinoids may be used to reduce chronic neuroinflammation and thus prevent motor neuron toxicity. Both CB1 and CB2 receptors were found to modulate release of endogenous interleukin-1 receptor antagonist (IL-1ra) from primary cultured glial cells, exerting neuroprotective effects [28]. Moreover, recent studies proved the anti-oxidative properties of cannabis, in particular inhibition of oxidative and nitrosative stress as well as reduced production of reactive oxygen species (ROS) [29]. CBD has been also found to regulate mitochondrial calcium concentrations as well as mitochondria-mediated intrinsic apoptosis [30, 31]. Additionally, CBD treatment may exert beneficial effects on skeletal muscle by reducing inflammation and improving muscle recovery [32].

Advertisement

3. Neuroprotective effects of cannabinoids in animal models of ALS

The most commonly used animal model to explore ALS is the SOD (G93A) transgenic mouse. The mouse is genetically engineered to express a mutation in the superoxide dismutase-1 gene (SOD1-G93A) and thus shows a phenotype similar to ALS in humans [33]. Raman et al. have demonstrated, that treatment with delta-9-tetrahydrocannabinol (Δ9-THC) in ALS SOD1 mice improved motor deficits and increased survival by 5%, most probably due to its anti-glutamatergic and anti-oxidant mechanisms of action [34]. Weydt et al. have shown, that cannabinol (CBN), a nonpsychotropic cannabinoid, delayed symptom onset in SOD1 transgenic mice without improving the survival [35]. In the subsequent study, Bilsland et al. have investigated the postsymptomatic treatment with an exogenous synthetic cannabinoid as well as genetic augmentation of endocannabinoids. The group has proven, that a potent CB1 and CB2 receptor agonist, WIN 55, 212-2, delayed the disease progression without affecting survival [36]. On the contrary, Shoemaker et al. have demonstrated, that a selective CB2 agonist, AM-1241, extended the lifespan of SOD1 mice by 4%, while WIN 55, 212-2 extended it by even 11% [37]. Zhao et al. have strengthened the evidence for AM-1241, showing its beneficial effect on disease progression [38]. Consequently, Moreno et al. evaluated the efficacy of nabiximols (trade name Sativex®) in SOD1 mice for the first time. Sativex® is a combination of 2.5 mg of CBD and 2.7 mg of Δ9-THC in the form of an oromucosal spray. In this study, Sativex® proved to be effective both in slowing the disease progression and improving survival [39]. Pasquarelli et al. have examined the neuroprotective and anti-inflammatory properties of 2-arachidonoylglycerol (2-AG). 2-AG is an endogenous CB1 and CB2 receptor agonist, which is present at relatively high levels in the central nervous system and is degraded by monoacylglycerol lipase (MAGL). In this study, the MAGL inhibitor KML29 was applied in order to increase the concentration of the 2-AG in the CNS of the SOD1 transgenic mice. This led to a reduction of proinflammatory cytokines and an increase in brain-derived neurotrophic factor (BDNF) expression levels in the spinal cord, the major site of neurodegeneration in ALS. Moreover, the oral KML29 treatment delayed the disease onset and extended life span in SOD1 mice up to 24 days [40]. Espejo- Porras et al. used another animal model, TDP-43 transgenic mice. The mis-metabolism of the RNA/DNA-binding protein TDP-43 (ALS-TDP), in particular, the presence of cytosolic aggregates of the protein is found in the spinal cords of more than 95% of ALS patients. Both the non-selective agonist WIN55,212-2 alone as well as in combination with the selective CB2 agonist HU-308 were shown to delay disease progression in TDP-43 mice [41]. Rodriguez-Cueto explored the anti-inflammatory, anti-oxidative and neuroprotective properties of VCE-003.2. VCE-003.2 is a novel derivative of the non-psychotrophic phytocannabinoid cannabigerol (CBG), which activates the peroxisome proliferator-activated receptor-γ (PPARγ). The treatment with VCE-003.2 was associated with a strong preservation of spinal motor neurons which may be attributed to normalizing the activation and cell function of the astrocytes [42]. An overview of studies investigating the endocannabinoid system in animal models of ALS is shown in Table 1.

ReferenceModelSubstanceOutcome
Raman et al. [34]SOD1 miceΔ9-THCΔ9-THC delayed disease progression in SOD1 mice and expanded their lifespan by 5%.
Weydt et al. [35]SOD1 micecannabinolCannabinol delayed disease onset in SOD1 mice.
Bilsland et al. [36]SOD1miceWIN 55, 212-2
CB1 and Faah ablation
Delayed disease progression in SOD1 mice.
Expanded lifespan in SOD1 mice by 13%.
Shoemaker et al. [37]SOD1 miceAM-1241
WIN-55,212-2
AM-1241 extended lifespan of SOD1 mice by 4%
WIN-55,212-2 extended lifespan of SOD1 mice by 11%
Zhao et al. [38]SOD1 miceAM-1241Delayed disease progression in SOD1 mice.
Moreno et al. [39]SOD1 micecombination of cannabidiol and
Δ9-THC
(Sativex®)
Delayed disease progression in SOD1 mice and improved survival.
Pasquarelli et al. [40]SOD1 mice2-AGDelayed disease progression in SOD1 mice and improved survival.
Espejo- Porras et al. [41]TDP-43 (A315T) miceWIN55,212-2
HU-308
Delayed disease progression in TDP-43 mice.
Rodriguez-Cueto [42]SOD1 miceVCE-003.2Improved survival of spinal motor neurons. Delayed disease progression in SOD-1 mice.

Table 1.

Summary of studies exploring the endocannabinoid system in animal models of ALS. Studies are presented in chronological order.

SOD 1, superoxide dismutase-1; Δ9-THC, Δ9-tetrahydrocannabinol; FAAH, fatty acid amide hydrolase; CB1, Cannabinoid receptor type 1; 2-AG, 2-arachidonoylglycerol.

Advertisement

4. Clinical research with cannabis-based medicine in ALS

In 2004 Amtmann et al. conducted the first survey on marijuana use among 131 patients with ALS. In this group of patients, 13 males reported using cannabis in the previous 12 months. Cannabis smokers reported reduction of depression, appetite loss, pain, spasticity, drooling and weakness [43]. To date, only a very limited number of studies regarding the potential use of CBM in ALS has been conducted. A small pilot study conducted by Gelinas et al. in a group of 20 ALS patients found THC to be effective against muscle cramps, fasciculations, insomnia and lack of appetite [44]. Subsequently, Weber et al. investigated the efficacy of oral THC in the treatment of ALS-related cramps in a group of 27 patients. The participants were randomly assigned to receive 5 mg THC twice daily followed by placebo or vice versa for 2 weeks. The intensity of cramps was assessed using the visual analogue scale (VAS). Unfortunately, no statistically significant improvement of cramp intensity, number of cramps, or fasciculation intensity was observed. No serious adverse events were noted, one of the participants complained about dizziness [45]. Joerger et al. examined the pharmacokinetics (PK) and tolerability of oral THC in ALS patients. The group showed, that adverse events (drowsiness, euphoria, orthostasis, sleepiness, vertigo and weakness) occurred more frequently in patients receiving 10 mg compared to 5 mg THC. No serious adverse events were reported. A marked interindividual variability regarding the absorption and elimination of THC was also observed [46]. Meyer et al. explored the efficacy of nabiximols in the treatment of ALS – related spasticity in a retrospective, mono-centric cohort study with 32 patients. The participants were treated with an oromucosal spray, the mean dose was 5.5 actuations per day. Spasticity was rated using the Numering Rating Scale (NRS) and the patient’s experience was evaluated with the net promoter score (NPS) and treatment satisfaction questionnaire (TSMQ-9). Moderate to severe spasticity was associated with an elevated number of daily THC:CBD actuations and stronger recommendation rate (NPS) [47]. A larger, multicentre, double-blind, randomized, placebo-controlled study (CANALS) was conducted by Riva et al. The group investigated the efficacy of nabiximols (trade name Sativex®) in the treatment of ALS-related spasticity in a group of 59 patients (29 in the nabiximols group and 30 in the placebo group). The participants self-titrated the oromucosal spray during the first 2 weeks of treatment, receiving up to 12 puffs per day, then maintained the dose for 4 weeks. After 6 weeks of treatment a statistically significant reduction of spasticity evaluated with the Modified Asworth Scale (MAS) was observed in the nabiximols group. No serious adverse events occurred in either group. The most common adverse effects in the nabiximols group were asthenia, somnolence, vertigo, and nausea [48]. Only recently, the EMERALD trial, a randomized, double-blind, placebo-controlled study was initiated to investigate the efficacy of cannabis-based medicine (CBME) on slowing down the progression of ALS. The investigational product contains 25 mg of CBD and less than 2 mg of THC (approximately 10%) formulated as a capsule. A total number of 30 patients with probable or definite ALS diagnosis based on the El Escorial criteria, with the symptom duration of <2 years, has been included in the study. The primary objective of the study is to evaluate the efficacy of CMBE on the disease progression measured with ALS Functional Rating Scale-Revised and forced vital capacity (FVC) score after 6 months of treatment [49].

An overview of clinical studies investigating the efficacy and safety of the cannabis – based medicine in ALS patients is shown in Table 2.

ReferenceNumber of patientsSubstanceResultsSafety
Gelinas et al. [44]20oral Δ 9-THCImprovement of effective against muscle cramps, fasciculations, insomnia and lack of appetite.No data.
Weber et al. [45]22oral Δ 9-THCNo significant improvement of cramp intensity, number of cramps, or fasciculation intensity.No study-related SAEs.
AEs: dizziness.
Joerger et al. [46]9oral Δ 9-THCAEs more frequent in patients receiving 10 mg compared to 5 mg THC. A marked interindividual variability regarding the absorption and elimination of THC.No study-related SAEs.
AEs: drowsiness, euphoria, orthostasis, sleepiness, vertigo, and weakness
Meyer et al. [47]32nabiximols (Sativex®)Higher efficacy in subgroups of ALS patients with moderate to severe spasticity.
High treatment satisfaction (TSQM-9).
Not data.
Riva et al. [48]59nabiximols (Sativex®)Significant reduction of ALS-related
spasticity
No SAEs.
AEs: asthenia, somnolence, vertigo, and nausea.

Table 2.

Summary of clinical studies investigating the efficacy and safety of the cannabis – Based medicine in ALS patients. Studies are presented in chronological order.

Δ9-THC, Δ9-tetrahydrocannabinol; AEs, adverse events; SAEs, Serious Adverse Events.

Advertisement

5. Safety profile of cannabis-based medicine in patients with ALS

Thus far, very little is known about the safety and tolerability of CBM in patients with ALS. The only study focusing specifically on the tolerability of oral THC showed, that adverse events (drowsiness, euphoria, orthostasis, sleepiness, vertigo, and weakness) were more frequent in ALS patients receiving 10 mg compared to 5 mg THC [46]. This contrasts with findings in patients with multiple sclerosis and healthy subjects [50], who tolerated single doses even up to 15 mg without significant adverse events [51]. However, the abovementioned study included only 10 participants [46]. The other available preliminary results suggested that the safety profile of CBM in ALS is similar to that in other groups of patients and did not report any serious adverse events [45, 47, 48]. A meta-analysis conducted by Whiting, including diverse populations of patients treated with CBM, showed that the treatment with cannabinoids can be associated with a greater risk of adverse events (AE), including serious adverse events (SAE). The most common short-term AEs encompass dizziness, dry mouth, nausea or vomiting, fatigue, somnolence, euphoria, disorientation, drowsiness, confusion, loss of balance, and hallucinations. Moreover, up to this date no study evaluating the long-term AEs of cannabinoids has been conducted [52].

Advertisement

6. Conclusions

Taking into consideration the above-mentioned reports, there is a valid rationale for the use of cannabis-based medicine in the symptomatic treatment of patients with ALS. The efficacy of cannabis in the management of spasticity, drooling and anorexia has been proven not only in the small RCTs with ALS patients, but also in large RTCs including other groups of patients suffering from similar symptoms in the course of other neurological diseases, most commonly in multiple sclerosis (MS) patients. Moreover, there is increasing evidence that cannabinoid compounds may exert neuroprotective effects and prolong the survival in the animal models of ALS. Therefore, larger RTCs are urgently needed to confirm the therapeutic potential of cannabis in slowing down the disease progression.

References

  1. 1. Mehta P, Raymond J, Punjani R, Han M, Larson T, Kaye W, et al. Prevalence of amyotrophic lateral sclerosis in the United States using established and novel methodologies. Amyotrophic Lateral Sclerosis and Frontotemporal Degeneration. 2017;2022:1-9
  2. 2. Feldman EL, Goutman SA, Petri S, Mazzini L, Savelieff MG, Shaw PJ, et al. Amyotrophic lateral sclerosis. The Lancet. 2022;400:1363-1380
  3. 3. Grossman M. Amyotrophic lateral sclerosis—A multisystem neurodegenerative disorder. Nature Reviews Neurology. 2019;15:5-6
  4. 4. Masrori P, Van Damme P. Amyotrophic lateral sclerosis: A clinical review. European Journal of Neurology. 2020;27:1918-1929
  5. 5. Crockford C, Newton J, Lonergan K, Chiwera T, Booth T, Chandran S, et al. ALS-specific cognitive and behavior changes associated with advancing disease stage in ALS. Neurology. 2018;91:e1370-e1e80
  6. 6. Burrell JR, Halliday GM, Kril JJ, Ittner LM, Götz J, Kiernan MC, et al. The frontotemporal dementia-motor neuron disease continuum. The Lancet. 2016;388:919-931
  7. 7. Lacomblez L, Bensimon G, Leigh PN, Guillet P, Meininger V. Dose-ranging study of riluzole in amyotrophic lateral sclerosis. Amyotrophic lateral sclerosis/Riluzole study group II. Lancet. 1996;347:1425-1431
  8. 8. Bensimon G, Lacomblez L, Meininger V, Group ARS. A controlled trial of riluzole in amyotrophic lateral sclerosis. New England Journal of Medicine. 1994;330:585-591
  9. 9. Abe K, Aoki M, Tsuji S, Itoyama Y, Sobue G, Togo M, et al. Safety and efficacy of edaravone in well defined patients with amyotrophic lateral sclerosis: A randomised, double-blind, placebo-controlled trial. The Lancet Neurology. 2017;16:505-512
  10. 10. Brooks BR, Berry JD, Ciepielewska M, Liu Y, Zambrano GS, Zhang J, et al. Intravenous edaravone treatment in ALS and survival: An exploratory, retrospective, administrative claims analysis. EClinicalMedicine. 2022;52:101590
  11. 11. Witzel S, Maier A, Steinbach R, Grosskreutz J, Koch JC, Sarikidi A, et al. Safety and effectiveness of long-term intravenous administration of edaravone for treatment of patients with amyotrophic lateral sclerosis. JAMA Neurology. 2022;79:121-130
  12. 12. Lunetta C, Moglia C, Lizio A, Caponnetto C, Dubbioso R, Giannini F, et al. The Italian multicenter experience with edaravone in amyotrophic lateral sclerosis. Journal of Neurology. 2020;267:3258-3267
  13. 13. Paganoni S, Macklin EA, Hendrix S, Berry JD, Elliott MA, Maiser S, et al. Trial of sodium phenylbutyrate–taurursodiol for amyotrophic lateral sclerosis. New England Journal of Medicine. 2020;383:919-930
  14. 14. Paganoni S, Hendrix S, Dickson SP, Knowlton N, Macklin EA, Berry JD, et al. Long-term survival of participants in the CENTAUR trial of sodium phenylbutyrate-taurursodiol in amyotrophic lateral sclerosis. Muscle & Nerve. 2021;63:31-39
  15. 15. Jiang J, Wang Y, Deng M. New developments and opportunities in drugs being trialed for amyotrophic lateral sclerosis from 2020 to 2022. Frontiers in Pharmacology. 2022;13:2004025
  16. 16. Saramak K, Szejko N. Endocannabinoid System as a New Therapeutic Avenue for the Treatment of Huntington’s Disease. In: From Pathophysiology to Treatment of Huntington’s Disease. IntechOpen; 2022
  17. 17. Szejko N, Saramak K, Lombroso A, Müller-Vahl K. Cannabis-based medicine in treatment of patients with Gilles de la Tourette syndrome. Neurologia i neurochirurgia polska. 2022;56:28-38
  18. 18. Marsicano G, Kuner R. Anatomical distribution of receptors, ligands and enzymes in the brain and in the spinal cord: Circuitries and neurochemistry. Cannabinoids and the Brain. 2008:161-201
  19. 19. Howlett AC, Blume LC, Dalton GD. CB1 cannabinoid receptors and their associated proteins. Current medicinal Chemistry. 2010;17:1382-1393
  20. 20. Klegeris A, Bissonnette CJ, McGeer PL. Reduction of human monocytic cell neurotoxicity and cytokine secretion by ligands of the cannabinoid-type CB2 receptor. British Journal of Pharmacology. 2003;139:775-786
  21. 21. Turner MR, Al-Chalabi A, Chio A, Hardiman O, Kiernan MC, Rohrer JD, et al. Genetic screening in sporadic ALS and FTD. Journal of Neurology, Neurosurgery & Psychiatry. 2017;88:1042
  22. 22. Van Den Bosch L, Van Damme P, Bogaert E, Robberecht W. The role of excitotoxicity in the pathogenesis of amyotrophic lateral sclerosis. Biochimica et Biophysica Acta -Molecular Basis of Disease. 2006;1762:1068-1082
  23. 23. Choi DW. Excitotoxic cell death. Journal of Neurobiology. 1992;23:1261-1276
  24. 24. Kim SH, Won SJ, Mao XO, Jin K, Greenberg DA. Molecular mechanisms of cannabinoid protection from neuronal excitotoxicity. Molecular Pharmacology. 2006;69:691-696
  25. 25. Abood ME, Rizvi G, Sallapudi N, McAllister SD. Activation of the CB1 cannabinoid receptor protects cultured mouse spinal neurons against excitotoxicity. Neuroscience Letters. 2001;309:197-201
  26. 26. Marsicano G, Goodenough S, Monory K, Hermann H, Eder M, Cannich A, et al. CB1 cannabinoid receptors and on-demand defense against excitotoxicity. Science. 2003;302:84-88
  27. 27. Liu Q , Bhat M, Bowen WD, Cheng J. Signaling pathways from cannabinoid receptor-1 activation to inhibition of N-methyl-D-aspartic acid mediated calcium influx and neurotoxicity in dorsal root ganglion neurons. Journal of Pharmacology and Experimental Therapeutics. 2009;331:1062-1070
  28. 28. Molina-Holgado F, Pinteaux E, Moore JD, Molina-Holgado E, Guaza C, Gibson RM, et al. Endogenous interleukin-1 receptor antagonist mediates anti-inflammatory and neuroprotective actions of cannabinoids in neurons and glia. Journal of Neuroscience. 2003;23:6470-6474
  29. 29. Velayudhan L, Van Diepen E, Marudkar M, Hands O, Suribhatla S, Prettyman R, et al. Therapeutic potential of cannabinoids in neurodegenerative disorders: A selective review. Current Pharmaceutical Design. 2014;20:2218-2230
  30. 30. Ryan D, Drysdale AJ, Lafourcade C, Pertwee RG, Platt B. Cannabidiol targets mitochondria to regulate intracellular Ca2+ levels. Journal of Neuroscience. 2009;29(7):2053-2063
  31. 31. Gross C, Ramirez DA, McGrath S, Gustafson DL. Cannabidiol induces apoptosis and perturbs mitochondrial function in human and canine glioma cells. Frontiers in Pharmacology. 2021;12:725136
  32. 32. Schouten M, Dalle S, Koppo K. Molecular mechanisms through which Cannabidiol may affect skeletal muscle metabolism, inflammation, tissue regeneration, and anabolism: A narrative review. Cannabis & Cannabinoid Research. 2022;7:745-757
  33. 33. Kim BW, Ryu J, Jeong YE, Kim J, Martin LJ. Human motor neurons with SOD1-G93A mutation generated from CRISPR/Cas9 gene-edited iPSCs develop pathological features of amyotrophic lateral sclerosis. Frontiers in Cellular Neuroscience. 2020;14:604171
  34. 34. Raman C, McAllister SD, Rizvi G, Patel SG, Moore DH, Abood ME. Amyotrophic lateral sclerosis: Delayed disease progression in mice by treatment with a cannabinoid. Amyotrophic Lateral Sclerosis Other Motor Neuron Disorders. 2004;5:33-39
  35. 35. Weydt P, Hong S, Witting A, Möller T, Stella N, Kliot M. Cannabinol delays symptom onset in SOD1 (G93A) transgenic mice without affecting survival. Amyotrophic Lateral Sclerosis. 2005;6:182-184
  36. 36. Bilsland LG, Dick J, Pryce G, Petrosino S, Di Marzo V, Baker D, et al. Increasing cannabinoid levels by pharmacological and genetic manipulation delay disease progression in SOD1 mice. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology. 2006;20:1003-1005
  37. 37. Shoemaker JL, Seely KA, Reed RL, Crow JP, Prather PL. The CB2 cannabinoid agonist AM-1241 prolongs survival in a transgenic mouse model of amyotrophic lateral sclerosis when initiated at symptom onset. Journal of Neurochemistry. 2007;101:87-98
  38. 38. Zhao P, Ignacio S, Beattie EC, Abood ME. Altered presymptomatic AMPA and cannabinoid receptor trafficking in motor neurons of ALS model mice: Implications for excitotoxicity. European Journal of Neuroscience. 2008;27:572-579
  39. 39. Moreno-Martet M, Espejo-Porras F, Fernández-Ruiz J, de Lago E. Changes in endocannabinoid receptors and enzymes in the spinal cord of SOD 1G93A transgenic mice and evaluation of a Sativex®-like combination of phytocannabinoids: Interest for future therapies in amyotrophic lateral sclerosis. CNS Neuroscience Therapeutics. 2014;20:809-815
  40. 40. Pasquarelli N, Engelskirchen M, Hanselmann J, Endres S, Porazik C, Bayer H, et al. Evaluation of monoacylglycerol lipase as a therapeutic target in a transgenic mouse model of ALS. Neuropharmacology. 2017;124:157-169
  41. 41. Espejo-Porras F, García-Toscano L, Rodríguez-Cueto C, Santos-García I, de Lago E, Fernandez-Ruiz J. Targeting glial cannabinoid CB2 receptors to delay the progression of the pathological phenotype in TDP-43 (A315T) transgenic mice, a model of amyotrophic lateral sclerosis. British Journal of Pharmacology. 2019;176:1585-1600
  42. 42. Rodríguez-Cueto C, Santos-García I, García-Toscano L, Espejo-Porras F, Bellido M, Fernández-Ruiz J, et al. Neuroprotective effects of the cannabigerol quinone derivative VCE-003.2 in SOD1G93A transgenic mice, an experimental model of amyotrophic lateral sclerosis. Biochemical Pharmacology. 2018;157:217-226
  43. 43. Amtmann D, Weydt P, Johnson KL, Jensen MP, Carter GT. Survey of cannabis use in patients with amyotrophic lateral sclerosis. American Journal of Hospice and Palliative Medicine. 2004;21:95-104
  44. 44. Gelinas D, Miller R, Abood M. A pilot study of safety and tolerability of Delta 9-THC (Marinol) treatment for ALS. Amyotrophic Lateral Sclerosis and Other Motor Neuron Disorders. 2002;3:23-24
  45. 45. Weber M, Goldman B, Truniger S. Tetrahydrocannabinol (THC) for cramps in amyotrophic lateral sclerosis: A randomised, double-blind crossover trial. Journal of Neurology, Neurosurgery & Psychiatry. 2010;81:1135-1140
  46. 46. Joerger M, Wilkins J, Fagagnini S, Baldinger R, Brenneisen R, Schneider U, et al. Single-dose pharmacokinetics and tolerability of oral delta-9-tetrahydrocannabinol in patients with amyotrophic lateral sclerosis. Drug Metabolism Letters. 2012;6:102-108
  47. 47. Meyer T, Funke A, Münch C, Kettemann D, Maier A, Walter B, et al. Real world experience of patients with amyotrophic lateral sclerosis (ALS) in the treatment of spasticity using tetrahydrocannabinol: Cannabidiol (THC: CBD). BMC Neurology. 2019;19:1-13
  48. 48. Riva N, Mora G, Sorarù G, Lunetta C, Ferraro OE, Falzone Y, et al. Safety and efficacy of nabiximols on spasticity symptoms in patients with motor neuron disease (CANALS): A multicentre, double-blind, randomised, placebo-controlled, phase 2 trial. The Lancet Neurology. 2019;18:155-164
  49. 49. Urbi B, Broadley S, Bedlack R, Russo E, Sabet A. 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 trial. BMJ Open. 2019;9:e029449
  50. 50. Killestein J, Hoogervorst E, Reif M, Kalkers N, Van Loenen A, Staats P, et al. Safety, tolerability, and efficacy of orally administered cannabinoids in MS. Neurology. 2002;58:1404-1407
  51. 51. Naef M, Curatolo M, Petersen-Felix S, Arendt-Nielsen L, Zbinden A, Brenneisen R. The analgesic effect of oral delta-9-tetrahydrocannabinol (THC), morphine, and a THC-morphine combination in healthy subjects under experimental pain conditions. Pain. 2003;105:79-88
  52. 52. Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, et al. Cannabinoids for medical use: A systematic review and meta-analysis. Journal of the American Medical Association. 2015;313:2456-2473

Written By

Kamila Saramak and Natalia Szejko

Submitted: 03 July 2023 Reviewed: 29 February 2024 Published: 27 March 2024