Next Article in Journal
3D Printed Pharmaceutical Systems for Personalized Treatment in Metabolic Syndrome
Previous Article in Journal
A Bird’s Eye View of Various Cell-Based Biomimetic Nanomedicines for the Treatment of Arthritis
Previous Article in Special Issue
Therapeutic Potential of Natural Compounds in Neurodegenerative Diseases: Insights from Clinical Trials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cannabinoids and Multiple Sclerosis: A Critical Analysis of Therapeutic Potentials and Safety Concerns

1
Department of Chemistry, School of Sciences and Engineering, The American University in Cairo, New Cairo 11835, Egypt
2
Biochemistry Department, Faculty of Science, Suez University, P.O. Box 43518, Suez 43533, Egypt
3
Institute of Global Health and Human Ecology, School of Sciences and Engineering, The American University in Cairo, P.O. Box 74, New Cairo 11835, Egypt
*
Author to whom correspondence should be addressed.
Pharmaceutics 2023, 15(4), 1151; https://doi.org/10.3390/pharmaceutics15041151
Submission received: 7 January 2023 / Revised: 19 March 2023 / Accepted: 21 March 2023 / Published: 5 April 2023
(This article belongs to the Special Issue Drug Targeting for CNS Disease)

Abstract

:
Multiple sclerosis (MS) is a complicated condition in which the immune system attacks myelinated axons in the central nervous system (CNS), destroying both myelin and axons to varying degrees. Several environmental, genetic, and epigenetic factors influence the risk of developing the disease and how well it responds to treatment. Cannabinoids have recently sparked renewed interest in their therapeutic applications, with growing evidence for their role in symptom control in MS. Cannabinoids exert their roles through the endogenous cannabinoid (ECB) system, with some reports shedding light on the molecular biology of this system and lending credence to some anecdotal medical claims. The double nature of cannabinoids, which cause both positive and negative effects, comes from their actions on the same receptor. Several mechanisms have been adopted to evade this effect. However, there are still numerous limitations to using cannabinoids to treat MS patients. In this review, we will explore and discuss the molecular effect of cannabinoids on the ECB system, the various factors that affect the response to cannabinoids in the body, including the role of gene polymorphism and its relation to dosage, assessing the positive over the adverse effects of cannabinoids in MS, and finally, exploring the possible functional mechanism of cannabinoids in MS and the current and future progress of cannabinoid therapeutics.

1. Introduction

Multiple sclerosis (MS) is a neurodegenerative condition that can cause paralysis, demyelination, and harm to neuronal axons [1]. Inflammation and myelin sheath degeneration are the hallmarks of MS that lead to lesions, which have been found in the white matter of the brain stem, optic nerve, and spinal cord [2]. The myelin sheath is destroyed via the immune system, which begins to perceive its constituent parts as foreign components. Infiltration of the immune cells is thought to cause plaque formation, which initiates disease symptoms through releasing cytokines and inflammatory mediators that cause inflammation, myelin damage, oligodendrocyte loss, neuronal function loss, and eventually axonal degeneration [3,4]. MS’s signs and symptoms depend on where the lesions are in the brain or spinal cord [5]. MS has three stages: pre-clinical, relapsing/remitting, and progressive clinical. Depending on the severity of the disease, the symptoms can differ from person to person. They can result in short-term, long-term, or even permanent losses due to disrupted signal transmission. The complete remission and the treatment of progressive forms of MS remain controversial and a medical challenge, even though numerous medications have been developed for the disease [6]. The drugs used to manage MS are classified into two main groups: disease-modifying agents and symptomatic treatment [7]. Symptomatic treatment aims to decrease the symptoms, but it is limited by its toxicity [8]. Recently, the therapeutic uses of cannabinoids as a symptomatic treatment has been gaining popularity, with many trials and patients, who believe that it may help with the management and control of symptoms in MS. Despite the overwhelming evidence supporting the use of cannabinoids in the treatment of MS, there is still a lack of knowledge regarding the precise effects of cannabinoids due to patient variability. In this review, we will explore and discuss the molecular effect of cannabinoids on the ECB system, the various factors that affect the response to cannabinoids in the body, including the role of gene polymorphism and its relation to dosage, assessing the positive over the adverse effects of cannabinoids in MS, and finally, exploring the possible functional mechanism of cannabinoids in MS and the current and future progress of cannabinoid therapeutics.

2. Cannabinoids and the Endocannabinoid System

Cannabis sativa is the main species of the complex plant known as Cannabis, a member of the Cannabacea family [9]. The Cannabis sativa plant, known as hemp, has been known as a psychoactive substance for over 4000 years due to its hallucinogenic characteristics. Cannabis is also known as marijuana, among other local names [10].
More than sixty physiologically active chemical substances, known as cannabinoids, can be created either naturally (phytocannabinoids), by animals (endocannabinoids), or artificially (synthetic cannabinoids) [11]. The cannabis plant contains more than 100 phytocannabinoids, including the 2 most significant ones, which are Δ⁹-tetrahydrocannabinol (THC) and cannabidiol (CBD). Δ⁹-THC, commonly referred to as THC unless stated otherwise, is believed to be the main psychoactive compound found in cannabis [12]. Unlike the phytocannabinoids found in cannabis, all endocannabinoids are derived from Arachidonic acid and, therefore, have different chemical structures. At present, there are five known endocannabinoids, including N-Arachidonoyl Ethanolamide (also called Anandamide), 2-Arachidonoyl Glycerol, 2-Arachidonoyl Glyceryl Ether (Noladin ether), O-Arachidonoyl Ethanolamine (Virodhamine), and N-Arachidonoyl Dopamine (NADA). Among these endocannabinoids, Anandamide and 2-Arachidonoyl glycerol are considered the most significant [13]. The Chinese pharmacopeia has referred to cannabinoids as a therapeutic agent since 400 AD [14]. Since 2015, there has been an upsurge in cannabinoid products and delivery systems, and an increase in the number of nations legalizing cannabinoids, possibly due to greater public awareness of the drug’s potential for medical use [15].
The endocannabinoid (ECB) system is a highly intricate signaling system composed of neuronal connections, endocannabinoid neurotransmitters, and G-protein-coupled receptors cannabinoid-1 (CB1) and cannabinoid-2 (CB2). The binding of cannabinoids, whether endogenous or exogenous, to these receptors, produces a range of physiological effects [16]. The ECB receptors are dispersed centrally and peripherally, as shown in Figure 1 [17].

3. Molecular Effect of Cannabinoids on the Central Nervous System

The impact of cannabinoids on the molecular level in the brain is one of the crucial elements that need to be thoroughly explained.

3.1. Role of CB1 and CB2 Receptors

CB1 receptors, which are primarily present in nerve terminals, are responsible for the inhibition of the release of neurotransmitters. On the other hand, CB2 receptors are expressed mainly by immune cells, and among their roles is the regulation of cytokine production and immune cell movement both inside and outside of the central nervous system. Additionally, recent research has shown that some neurons and blood arteries in the brain have CB2 receptors [18,19]. The ECB system mediates neuroprotective actions via glutamatergic neurons, while inflammatory responses are regulated via GABAergic neurons and astrocytes. Many neuronal disorders, including age-related neurodegeneration, are linked to changes and dysfunction of the ECB system [19]. Age-related cognitive decline is associated with decreased CB1 receptor expression in the hippocampus, and CB1 receptor deletion from GABAergic hippocampal neurons results in neuronal death and elevated inflammation [20]. Moreover, changes in CB1 receptor signaling that are unique to glutamatergic neurons may impact age-related cognitive decline and decreased synaptic integrity and plasticity. Cannabinoids that signal through the CB1 receptor cause synaptic plasticity, cell migration, and neuronal growth, whereas cannabinoids that signal through the CB2 receptor are associated with mechanisms that stop, slow down, and repair damage caused by inflammation, as illustrated in Figure 2 [21].

3.2. Molecular Effect of Cannabinoids and ECB System in the Hippocampus

Higher-order brain activities rely on coordinating various delicately balanced systems like an orchestra, which executes the rhythms that shape our cognitive processes, affect our behavior, and create our memory. The hippocampus has drawn more attention than any other brain region. Consequently, it is an essential component of mnemonic systems in various species, including humans. Additionally, several studies have suggested that the hippocampus is a center for conscious and unconscious experience [22,23]. It is essential to explore the molecular mechanisms controlling hippocampus circuitries to understand the comprehend neural processes. Molecular effectors are seen as CNS modulators indicating that their actions impact cognition and behavior. The hippocampus plays a role in cognitive functions such as memory, learning, and sensory integration [24,25].
Additionally, it contains a lot of CB1 receptors, which are crucial for controlling pathophysiological processes [25,26]. The CB1 receptors are found mainly in the hippocampus’ GABAergic neurons and are also present in glutamatergic neurons, astrocytes, and subcellular compartments [27,28]. Cannabinoid signaling regulates the release of cholinergic and dopaminergic neurotransmitters in addition to the typical excitatory/inhibitory transmission regulation by CB1 receptors [29,30]. Acetylcholine (ACh) and GABA are released at mixed GABAergic synapses established explicitly by the cholinergic terminals in the hippocampus [31]. In this situation, regulating dopaminergic and cholinergic transmission in the hippocampus under CB1 receptor-dependent may control cognitive and affective processes.
Interestingly, cannabinoid antagonist use has been linked to improved cognitive abilities. This can be explained by increased acetylcholine levels brought on by cholinergic disinhibition in the hippocampus. In ACh neurotransmission, Δ9-tetrahydrocannabinol (Δ9-THC) exhibits a biphasic, dose-dependent impact. Because ACh levels are governed by dopamine receptor activation, only high dosages of THC can reduce their levels. As a result, while septal D1 receptor signaling increases ACh efflux, hippocampal D2 receptor activation induces ACh suppression triggered by large dosages of THC [32]. Dopaminergic terminal activity in the ventral tegmental region is increased when endogenous or exogenous cannabinoids activate the intrahippocampal CB1 receptor. These systems must be appropriately controlled for the hippocampus to function at its peak, particularly during stressful situations when alterations in neurotransmitter levels may cause maladaptive changes connected to neuropsychiatric disorders [33]. This molecular mechanism emphasizes the critical role of the endocannabinoid system in the progression or management of neurodegenerative disease, implying the possibility of developing treatments that can cure MS as well as many other neurodegenerative disorders.

3.3. Effect of Cannabinoids on Mitochondria and Metabolic Pathway

The brain accounts for only 2% of total body weight in mammals but utilizes up to 20% of the body’s energy production [34,35]. Mitochondria are crucial elements of eukaryotic cell functions [34,36,37]. The role of neuronal energetics in brain physiology and pathology is the subject of extensive research. The molecular mechanisms linking mitochondrial activity to brain functions remain unclear. CB1 receptors have recently been discovered in the mitochondria of hippocampus astroglia cells [27], although it is still unclear how they might affect astrocytic activity and possibly modify brain networks. The activation of the mitochondrial CB1 receptor (mtCB1) has been linked to alterations in bioenergetics and mitochondrial respiration that influence hippocampus synaptic transmission and, in turn, memory consolidation [38,39]. By turning on a subgroup of mitochondrial G proteins called Gi proteins, the mtCB1 receptor slows down mitochondrial respiration. Proteins involved in oxidative phosphorylation are not phosphorylated as much as before as a result of this activation, which also decreases protein kinase A (PKA) activity in the mitochondria. Since synaptic transmission requires a significant amount of energy, any disruptions in mitochondrial respiration directly impact synaptic activity. Long-term memory is also disturbed when the mtCB1 receptor is activated because it decreases ATP and prevents excitatory synaptic transmission in the CA1-CA3 circuit [39]. A recent study connected the disturbance of glucose metabolism and lactate generation to the activation of astroglia mtCB1 receptors [28]. THC administration results in decreased mitochondrial protein phosphorylation, which changes reactive oxygen species (ROS) levels, causing the transcription factor hypoxia-inducible factor 1 to be downregulated (HIF-1). Since the HIF-1 pathway promotes glycolysis, inhibiting it has a detrimental impact on glucose metabolism and lactate generation.

4. Pharmacokinetics and Pharmacodynamics Characteristics of Cannabinoids

Cannabinoids have been proven to have anti-inflammatory, antiviral, and anticancer characteristics, according to studies on the pharmacodynamics of cannabinoids [40]. Recognizing the pharmacokinetics and pharmacodynamics characteristics of cannabinoids is crucial to understanding the effects of drugs on the body. Most of the pharmacodynamics and pharmacokinetic data were obtained through studies on cannabinoid users or healthy volunteers. Several factors impact the pharmacokinetics of cannabinoids, such as prior cannabinoid usage, pharmacogenetics, body size, disease status, food, and microbiome. The dosage form of cannabinoids and the route of administration impact the pharmacokinetics parameters of cannabinoids, as shown in Table 1 [41]. The biodistribution of cannabinoids can be affected by their lipophilicity, where THC has a high distribution volume (5.7–10 L/kg) due to its high lipophilicity. The distribution of CBD is also affected by its lipophilicity and has a high volume of distribution, and it can rapidly penetrate the brain, adipose tissue, and other organs [42]. Another factor that may increase the volume of distribution is the chronic regular administration of cannabinoids which precipitates tissue accumulation over time [42,43]. The liver predominantly metabolizes cannabinoids. A tiny amount of extra-hepatic metabolism exists in tissues such as the brain, intestines, and lungs [44]. The cytochrome P450 (CYP 450) enzymes, specifically CYP2C9, CYP2C19, and CYP3A4, metabolize THC in the liver. Decarboxylation, epoxidation, and oxidation are all steps in THC metabolism that occur before conjugation. The two primary THC metabolites are Δ11-hydroxy-THC (Δ11-OH-THC) and Δ11-carboxy-THC (Δ11- COOH-THC), which are produced when THC is hydroxylated and oxidized, respectively. Tissues that express CYP 450 participate in the extra-hepatic metabolism of THC [45].
Additionally, CBD is metabolized in the liver via the CYP 450 isoenzymes CYP2C19 and CYP3A4, where it is then subjected to different metabolic processes before being excreted [46]. The route of elimination is also a pharmacokinetic characteristic in which THC and CBD metabolites are eliminated through the urine, feces, and, to a lesser extent, bile. The elimination rate is influenced by several variables, including the type of dosage form and the patient’s characteristics [40]. Until now, no clear guideline involves specific instruction about the required dose of cannabinoids. Consequently, there is a need to conduct studies on the kinetics of the drug with different patient populations and diseases, specifically the MS population.
Table 1. Effect of various dosage forms on pharmacokinetics parameters (PKM) of cannabinoids.
Table 1. Effect of various dosage forms on pharmacokinetics parameters (PKM) of cannabinoids.
PKMAbsorptionBioavailabilityPeak Plasma ConcentrationDuration of ActionAdvantagesDisadvantages
Dosage Form
InhalationHigh absorption = 10–60%THC = 2–65%, CBD = 6–31%Peak plasma concentrations of THC and CBD are reached quickly, within 3–10 min.1–4 hSuitable bioavailability and rapid onset of action.Variability between patients based on lung function.
OralLow absorption = 2–14%THC = 5–10%, CBD = 6–20% Due to the effect of 1st pass metabolism.Achieve peak concentration (60–120 min).6 hIt provides a sufficient duration of action.Delayed onset of action.
TopicalIrregular absorptionSkin barriers hinder bioavailability due to the lipophilic nature of the drug.Steady-state condition is achieved within = 17 h.THC = 14 h, CBD = 72 hReduction in the side effects associated with systemic administration of the drugs.Poor bioavailability due to skin barrier.
Systemic intravenousHigh absorption rateHigh bioavailability like inhaled dosage form.Within 10 min.4 hRapid action and high bioavailability.Require an aqueous vehicle due to poor water solubility.
Ref.[15,43,44][15,47,48][15,49][49,50][50,51][15,52]

5. Pharmacogenetics and Cannabinoids

Gene polymorphisms implicated in drug action, metabolism, and transport in the body may be the reason for response variation in MS patients responding to cannabinoid therapy. The most recent data on gene polymorphisms influencing cannabinoids transport, activity, and metabolism will be discussed here. The genetic alteration that can affect the response to cannabinoid therapy can be divided into variations of genes that code for cannabinoid receptors or those that code for metabolizing enzymes.

5.1. Variation of Genes That Code for Cannabinoid Receptors

The majority of studies on the effects of cannabinoids have focused only on a particular class of receptors that cannabinoids act on. There are more than one or two cannabinoid receptors with specific effects on the body’s function that require additional research, especially in the population of MS. The ECB system has various types of receptors, which may impact the activity of ligands in many physiological processes, and scientists have demonstrated in recent years that this mechanism is far more complex [53]. Variations in the expression of these receptors will affect cannabinoids and their effects on the body, as shown in Table 2.

5.1.1. Polymorphism of Cannabinoid Receptors CB1 and CB2

The discovery and cloning of the first and second cannabinoid receptors (CB1 and CB2) in 1990 and 1993 significantly raised our knowledge about cannabinoids. The CNR1 gene, which codes for the CB1 receptor, is found on human chromosome 6q14–15 and has four exons, the biggest of which is most frequently expressed in brain tissue [60]. The CB1 receptor is characterized by evolutionary conservatism. The CB receptor’s activation increases hunger and has sedative, analgesic, and antiemetic properties [61]. Contradictory findings were found in research linking the CNR1 gene’s single nucleotide polymorphism (SNP) with marijuana addiction, which also included the trinucleotide repeat locus (AAT) and the insertion-deletion (-3180T) polymorphism [54]. In 206 non-Hispanic Caucasians (92 subjects and 114 controls), Comings et al. found a link between (AAT) >5 repeats and drug dependency [62]. However, most trinucleotide repeat (AAT)n studies had contradictory findings [63,64]. Moreover, the Hartman Group found that in case-control samples, the CNR1 gene variant rs1049353 was associated with symptoms of cannabinoid dependency [65]. CNR2, which codes for CB2, is the second gene chosen. There are not many descriptions in the literature of studies on the CNR2 gene and addiction. Mutant CB2 receptors were then transfected into HEK293 cells, according to Carrasquer et al. This study showed that the CB2 polymorphic receptors might bind cannabinoid ligands at locations 63 and 316 and mediate signal transmission, which may help explain the etiology of some disorders [66].

5.1.2. Transient Receptor Potential Cation Channel Subfamily V Member 1 (TRPV1)

The transient receptor potential cation channel subfamily V member 1, also known as TRPV-1, is a ligand-gated, non-selective ion channel discovered as another receptor target for cannabinoids. TRPV-1 has been shown to express itself in a wide range of cells, including those of the immune system, the central and peripheral nervous systems, endothelium and epithelial cells, keratinocytes, and smooth muscle cells [11]. On primary afferent nerve cells, there is a more significant co-localization of cannabinoid receptors and TRPV-1. It has been demonstrated that the endocannabinoid AEA acts as an endo vanilloid by stimulating the TRPV-1 receptor, which mediates downstream signaling [67].
Until now, no association studies have shown a connection between the presence of TRPV1 gene polymorphism and marijuana addiction. Two well-known receptors, CB1 and CB2, are the focus of most studies on the functioning of the ECB system. CBD’s two enantiomeric variants have little or no affinity for the CB1 and CB2 receptors. Therefore, it is conceivable that the effectiveness of CBD is linked to its ability to reduce the activity of FAAH and increase anandamide levels (AEA). This would suggest that elevated MDR1 mRNA expression following CBD exposure depends on AEA’s simultaneous activation of CB2 and TRPV1 [53].

5.2. Variation in Genes That Code for Metabolizing Enzymes

The metabolism of cannabinoids is enzyme-dependent [68]. Oxidation, decarboxylation, and conjugation with glucuronic acid are the primary mechanisms used in metabolizing Δ9-THC. There is no doubt that environmental factors such as drug availability and social conditioning impact cannabinoid usage, but more research is required to understand the genetic effect on cannabinoids fully. The CYP gene family consists of 58 pseudogenes and 57 putatively functional genes in humans. Alterations in or inactivation of enzyme activity can result from CYP gene polymorphism and mutation. Cytochrome P450 (CYP, EC1.14.14.1) superfamily enzymes, specifically CYP3A4, are considered a significant metabolic pathway for THC, CBD, and CYP2C9 encoded by CYP2C9 and CYP3A4 genes are predicted to play an essential role in the primary metabolism of THC. The highly polymorphic CYP2D6 gene, a member of the CYP superfamily, has been implicated in discovering a phase I enzyme with over 91 distinct alleles [69]. The most prevalent CYP2D6 variants are *3, *4, and *5, with phenotypes indicative of poor metabolizers and decreased or absent enzyme activity. There is a wide range of CYP2D6 activity in people with genetic polymorphisms in the CYP2D6 gene. Moreover, xenobiotics and endobiotics are glucuronidated by a superfamily of enzymes known as UDP-Glucuronosyltransferases (UGTs).
The UDP-glucuronosyltransferases (UGTs) superfamily of enzymes is a key player in detoxifying and eliminating both endogenous and exogenous substances, such as cannabinoids and their metabolite, through the process of glucuronidation. Of all phase II drug metabolites, 35% or less are glucuronides [70]. The UGT families consist of UGT1, UGT2, UGT3, and UGT8 [71]. Additionally, recent research has shown new polymorphisms connected to the various enzyme alleles, as illustrated in Table 3. In addition to the disease-modifying therapies that treat critical MS, dose requirements for several commonly used drugs with a narrow therapeutic range may differ by more than 20-fold depending on genotype or enzyme expression status [72].

6. Adverse Effects of Cannabinoids

Cannabinoids are often dismissed by the general public as a safe substance, oblivious to any potential long-term health issues [83,84,85]. Comparing cannabinoid users with non-users in the general population reveals that cannabinoids negatively impact cognition [86]. There is growing evidence that acute cannabinoid use is linked to other neurocognitive decision-making deficits, including processing speed, sustained attention, verbal fluency, and executive functioning [87,88,89,90,91,92,93,94]. Chronic cannabinoid usage by teenagers and young adults over time affects cognitive abilities in several domains, including learning, memory, attention, decision-making, executive functioning, and psychomotor speed [85,95,96,97,98]. However, one systematic review investigated the adverse effects of cannabinoids in progressive MS patients who never used cannabinoids. The results indicated potential improvements in cognition with medicinal cannabinoids for MS patients. However, this effect is only for short-time use. It contradicts the findings that chronic use of whole-plant cannabinoids resulted in impairments in memory, attention, and executive and visuospatial function [99]. More research is required for the detailed investigation of the adverse effects of cannabinoid use both in the short and the long term.
A recent review on the adverse effects of cannabinoids has discussed not only the implications of cannabinoids on mental functions, psychiatric conditions, and cognitive and CNS alterations but also their effects on the respiratory system, the immune system, the reproductive system, and the cardiovascular system [100]. Therefore, the use of cannabinoids in the medical field must be conducted with great caution to benefit from their potential benefits while avoiding the possible risks.

7. Cannabinoids and the Management of Multiple Sclerosis

Earlier studies using an in vitro model of MS, the Experimental Autoimmune Encephalomyelitis (EAE) model, have shown that cannabinoids are highly effective in treating MS [101]. The cannabinoid receptor agonists Tetrahydrocannabinol, Δ8- and Δ9-THC appear to lessen the symptoms of EAE and significantly reduce the immune response in animals. Several studies have shown that clinical signs of EAE, such as tail flaccidity and generalized atonia, were delayed in onset and diminished in severity after these pharmacological treatments [102,103]. Additionally, one of those studies found that Δ9-THC reduced the histological evidence of EAE inflammation in the spinal cord of rats and guinea pigs [102]. Interestingly, dexanabinol, one of the synthetic cannabinoid ligands that lack the potential to act through CB1 or CB2 receptors, has also been shown to lessen EAE symptoms in rats [104].
Numerous clinical studies indicate that the pain and spasticity caused by MS and spinal cord injuries may be effectively reduced via cannabinoids, whose potential to reduce the signs and symptoms of MS and spinal cord damage is consistent with some of the drug’s conventional medical uses. Clinical investigations have shown that cannabinoid receptor agonists help lessen some of the MS-specific signs and symptoms, and these clinical trials are depicted in great detail in Table 4.
The promising effects of cannabinoids in managing various effects of MS are something that all of the research from 1996 to 2020 has in common. For example, neuropathic pain, one of the significant MS symptoms, has been demonstrated to be reduced by using cannabinoids. Several clinical studies involving various cannabinoids and dosage forms produced promising outcomes for the prevention and/or treatment of neuropathic pain. However, some clinical trials used personal anecdotes gleaned through distributing questionnaires to MS patients using cannabinoids for self-medication. The first survey used responses from 57 male and 55 female patients [105]. The results pointed out that more than 90% of these individuals who were experiencing MS symptoms, such as spasticity at the start of sleep, muscle discomfort, pain in the legs at night, tremor in the arms or head, and depression, have reported relief after using cannabinoids. Despite these findings, there is conflicting evidence about the effectiveness of cannabinoids in treating MS and spinal cord damage. It is currently unknown if cannabinoid receptor agonists prevent MS from progressing in patients with this condition or if they only lower the intensity of specific signs and symptoms. There is still some debate in the literature about the relative importance of CB1 and CB2 receptor activation and the causes of variations in response to cannabinoids. Many factors, such as dosage form, genetic makeup, or the molecular effects of cannabinoids at different doses, contribute to patient response variation.
Table 4. Various clinical trials on the use of cannabinoids with MS patients.
Table 4. Various clinical trials on the use of cannabinoids with MS patients.
Drug (Active Constituent)Dosage FormExperimental DesignOutcomesRef.
THC (5–10 mg)OralDouble-blind studyDecrease muscle spasms and enhance the walking ability of the patient.[106]
Nabilone
(1 mg), a synthetic THC mimic
OralOpen-Label studyDimension in the pain with MS patients.[107]
THC (10 mg oral or 15 mg rectal) Oral/rectalOpen-Label studyEnhancement of walking ability, passive mobility, and dimension in the pain with young MS patients.[108]
THC (7.5 mg)OralPlacebo-controlled, double-blind studyImproved muscle spasm perception.[109]
Sativex, a THC-CBD combination
(2.7 mg: 2.5 mg)
Oromucosal Controlled double-blind, randomized studyIt has an analgesic effect in addition to enhancing of quality of life of the patient (e.g., sleep improvement).[110]
Tobacco and cannabis resin-containing smokingInhalation Placebo-controlled trialDecrease in the Nystagmus amplitude and enhancement of visual ability.[111]
Dronabinol
(>25 mg), a synthetic THC mimic
OralRandomized, double-blind controlled studySignificant analgesic effect.[8]
THC (5–15 mg)Oral The single-blind study was a placebo-controlled trialEnhancement of the patient’s ability in handwriting and a significant decrease in spasticity and tremors.[112]
THC (5–10 mg)OralDouble-blind studyImprovement in tremors and ataxia with MS patient.[113]
Nabilone (1 mg), a synthetic THC mimicOralDouble-blind controlled trialsReduction in muscle spasms and tremors.[114]
THC (10–15 mg oral or 2.5–5 mg rectal)Oral/rectalOpen-Label studyAnalgesic effect with MS patient.[108]
Sativex, a THC-CBD combination
(2.7 mg: 2.5 mg)
OromucosalDouble-blind controlled trialsImprovement in muscle spasms in MS patients.[115]
THC (1 mg)OralOpen-Label studyHave an analgesic effect on patients.[107]
Sativex, a THC-CBD combination
(2.7 mg: 2.5 mg)
OromucosalDouble-blind controlled randomized trial Improving the resistant MS spasticity more effectively and clinically.[116]
Sativex, a THC-CBD combination
(2.7 mg: 2.5 mg)
OromucosalObservational, prospective controlled trialImprovement in the symptoms of MS in resistant patients. [117]
Sativex, a THC-CBD combination
(2.7 mg: 2.5 mg)
OromucosalDouble-blind controlled trialImprovement in the clinical states of MS patients.[118]
Sativex, a THC-CBD combination
(2.7 mg: 2.5 mg)
OromucosalRandomized controlled studyDecrees the neuropathic pain associated with MS patients.[119]
Sativex, a THC-CBD combination
(2.7 mg: 2.5 mg)
OromucosalOpen-Label studyThis study has proven the immunomodulatory effect of cannabinoids by detecting the gene expression of immune-related pathways.[120]
Sativex, a THC-CBD combination
(2.7 mg: 2.5 mg)
OromucosalRandomized controlled trialA significant reduction in the pain associated with MS.[121]
Sativex, a THC-CBD combination
(2.7 mg: 2.5 mg)
OromucosalControlled retrospective study It demonstrates an efficient and safe reduction in muscle spasms.[122]

8. Functions of Cannabinoids in MS

Animal models have been used to investigate the possible function of cannabinoids in preventing the progression of MS and providing neuroprotection. One of the most accepted mechanisms of action is immunosuppression. This effect was revealed in an in vivo study using an EAE model of mice and a daily dose of CBD, which showed dimensions in the T cell infiltration and neuroinflammation in the brain and spinal cord’s white matter pathways [123]. Another similar study used CBD alone or in combination with THC. In both techniques, there was a reduction in the proliferation and number of T cells, which impacted and reduced the degree of demyelination of neurons [124].
In vitro studies also supported the immunosuppression mechanism as the most dominant pathway of protection in MS. Two earlier studies used THC on either animal or human cell cultures, and the results showed inhibition or reduction of T cells’ proliferation [125,126]. A recent study has also demonstrated that THC decreases the number of natural killer cells (NK) [127]. Recent studies extended to the use of CBD in cell cultures. A study by Yang et al. investigated the use of CBD on T cells and showed that the proinflammatory phenotype of T cells was reversed [128]. It is also noteworthy that the effect of cannabinoids is not exclusive to T cells, as demonstrated by a study by Kozela et al., which showed that the use of CBD negatively impacted both T cells and B cells [129].

9. The Growing Field of Cannabinoid Therapeutics

Several clinical studies have been conducted on human individuals (Table 4). The results showed great promise for using cannabinoids in MS. Commercial cannabinoid formulations have advanced in recent years, but their use is still limited. The combined effects of THC and CBD can be observed in Sativex, one example of a commercial cannabinoid formulation. Many clinical trials have been conducted to evaluate the efficacy of Sativex as a supplemental therapy for patients with MS who have moderate to severe spasticity [116,130]. Nabiximols, a combination of CBD and Δ9-THC formulation, have also been used to manage spasticity associated with MS [131]. Neuropathic pain, a common symptom of MS that affects between 17% and 70% of patients, was also shown in a study to be reduced by Sativex. Additionally, Sativex is well tolerated by MS patients and has a low incidence of side effects [132].
Although the number of cannabinoids in formulations approved for the treatment of MS is still limited, several cannabinoids are used to treat a variety of other diseases and conditions, such as Nabilone for the management of Parkinson’s disease [133], Dronabinol, which is used to decrease anorexia, disturbed behavior [134], and nighttime agitation [135] in Alzheimer’s disease, and Cesamet for the treatment of chemotherapy-induced nausea in cancer patients [136]. All these indicate that the field of cannabinoid therapeutics is still in its infancy, but it will witness remarkable progress in the future.

10. Future Research on Cannabinoids

It has been discovered that cannabinoids may bind to a variety of locations, including the transient receptor potential vanilloid subtype 1 (TRPV1), the G-protein-coupled receptor 55 (GPR55), and the cannabinoid CB1 and CB2 receptors [100]. Despite the accepted scientific fact that cannabinoids act through interactions with the receptors of the ECB system, which are CB1 and CB2 [16], the idea that a drug may interact with several proteins to exert its biological function has slowly gained acceptance. Therefore, a recent review by Zhu et al. discussed the recent updates on natural products and the different approaches for target identification via binding affinity experiments for target recognition and validation and the biological function verification of the significance of this binding. This review also pointed out single-cell multi-omics and pathway enrichment analysis for identifying gene regulatory networks and the full impact of drug interactions on system biology [137]. Future research on cannabinoids using this approach will enable us to understand further the potential benefits and safety risks of cannabinoids for several diseases and conditions.

11. Concluding Remarks and Perspectives

Multiple sclerosis (MS) is a neurodegenerative condition in which inflammation and myelin degeneration lead to lesions, which have been found in the white matter of the brain stem, optic nerve, and spinal cord [2]. MS’s signs and symptoms depend on where the lesions are in the brain or spinal cord [5]. Symptomatic treatment aims to decrease the symptoms, but it is limited by its toxicity [8]. More than sixty physiologically active chemical substances, known as cannabinoids, can be created either naturally (phytocannabinoids), by animals (endocannabinoids), or artificially (synthetic cannabinoids) [11]. The therapeutic use of cannabinoids as a symptomatic treatment for MS has recently grown in popularity, where they exert their function through the endocannabinoid (ECB) system, which is a complex signaling system that includes the G-protein-coupled receptors cannabinoid-1 (CB1) and cannabinoid-2 (CB2) [16].
Cannabinoids have been proven to have anti-inflammatory, antiviral, and anticancer characteristics, according to studies on the pharmacodynamics of cannabinoids [40]. However, the effects and responses of cannabinoids can vary among individuals due to genetic variations in cannabinoid receptors or metabolizing enzymes, as shown by different studies in Table 2. Therefore, cannabinoid treatment should be tailored to an individual’s genomic state rather than used indiscriminately. The potential benefits of cannabinoids must also be balanced with the associated risks, including adverse effects on mental, cognitive, and physical functions and the respiratory, immune, reproductive, and cardiovascular systems [100]. Therefore, the medical use of cannabinoids must be approached with caution.
Since the 1990s, the therapeutic use of cannabinoids in MS has been studied through in vitro experiments, in vivo pre-clinical studies on animals, clinical trials on human subjects, and patient questionnaires assessing symptom relief after self-medication with cannabinoids. All these studies showed the potential therapeutic benefits of cannabinoids in MS. Some of them advanced to produce commercial therapeutic formulations of cannabinoids such as Sativex, which is used as a supplemental therapy for patients with MS who have moderate to severe spasticity [116,130], and Nabiximols, which has also been used for the management of spasticity associated with MS [131]. However, despite extensive previous research, further studies are needed on cannabinoids to enhance their safety and efficacy in treating MS and other diseases.

Author Contributions

All persons who meet authorship criteria are listed as authors. All authors certify that they have participated sufficiently in the work to take public responsibility for the content, including participation in the concept, design, writing, or manuscript revision. Furthermore, each author certifies that this material or similar material has not been and will not be submitted to or published in any other publication before its appearance in this journal. The review’s conceptualization and design were carried out by R.A.N., A.K. and A.A.; R.A.N. drafted the manuscript; A.K. and A.A. reviewed the manuscript for important intellectual content. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All figures (Figure 1 and Figure 2) in this manuscript were constructed using Biorender (www.biorender.com). Additionally, a publication license was obtained from Biorender for the use of these figures in this manuscript, which allows for the reproduction and distribution of the figures.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kobelt, G.; Thompson, A.; Berg, J.; Gannedahl, M.; Eriksson, J. New Insights into the Burden and Costs of Multiple Sclerosis in Europe. Mult. Scler. J. 2017, 23, 1123–1136. [Google Scholar] [CrossRef] [Green Version]
  2. Absinta, M.; Sati, P.; Masuzzo, F.; Nair, G.; Sethi, V.; Kolb, H.; Ohayon, J.; Wu, T.; Cortese, I.C.M.; Reich, D.S. Association of Chronic Active Multiple Sclerosis Lesions with Disability In Vivo. JAMA Neurol. 2019, 76, 1474–1483. [Google Scholar] [CrossRef]
  3. Reich, D.S.; Lucchinetti, C.F.; Calabresi, P.A. Multiple Sclerosis. N. Engl. J. Med. 2018, 378, 169–180. [Google Scholar] [CrossRef] [PubMed]
  4. Gerhard, L.; Dorstyn, D.S.; Murphy, G.; Roberts, R.M. Neurological, Physical and Sociodemographic Correlates of Employment in Multiple Sclerosis: A Meta-Analysis. J. Health Psychol. 2020, 25, 92–104. [Google Scholar] [CrossRef] [PubMed]
  5. Sebastião, E.; Wood, T.; Motl, R.W.; Santinelli, F.B.; Barbieri, F.A.; Zanesco, A. The Importance of Promoting Physical Activity and Exercise Training as Adjuvant Therapy for People with Multiple Sclerosis. Motriz Rev. Educ. Fis. 2022, 28, e10220016021. [Google Scholar] [CrossRef]
  6. Chountoulesi, M.; Demetzos, C. Promising Nanotechnology Approaches in Treatment of Autoimmune Diseases of Central Nervous System. Brain Sci. 2020, 10, 338. [Google Scholar] [CrossRef] [PubMed]
  7. Broadley, S.A.; Barnett, M.H.; Boggild, M.; Brew, B.J.; Butzkueven, H.; Heard, R.; Hodgkinson, S.; Kermode, A.G.; Lechner-Scott, J.; Macdonell, R.A.L.; et al. Therapeutic Approaches to Disease Modifying Therapy for Multiple Sclerosis in Adults: An Australian and New Zealand Perspective Part 3 Treatment Practicalities and Recommendations. J. Clin. Neurosci. 2014, 21, 1857–1865. [Google Scholar] [CrossRef] [PubMed]
  8. Zajicek, J.; Fox, P.; Sanders, H.; Wright, D.; Vickery, J.; Nunn, A.; Thompson, A. Cannabinoids for Treatment of Spasticity and Other Symptoms Related to Multiple Sclerosis (CAMS Study): Multicentre Randomised Placebo-Controlled Trial. Lancet 2003, 362, 1517–1526. [Google Scholar] [CrossRef]
  9. Dąbrowski, G.; Skrajda, M. Cannabinoids from Cannabis sp.: Mechanism of their activity and potential health benefits in human body. J. Educ. Health Sport 2017, 7, 936–945. [Google Scholar] [CrossRef]
  10. Hollister, L.E. Health Aspects of Cannabis. Pharmacol. Rev. 1986, 38, 1–20. [Google Scholar] [CrossRef]
  11. Garbutcheon-Singh, K.B.; Smith, S.D. Cannabinoids Interaction with Transient Receptor Potential Family and Implications in the Treatment of Rosacea. Dermatol. Ther. 2021, 34, e15162. [Google Scholar] [CrossRef]
  12. Pennypacker, S.D.; Romero-Sandoval, E.A. CBD and THC: Do They Complement Each Other Like Yin and Yang? Pharmacother. J. Hum. Pharmacol. Drug Ther. 2020, 40, 1152–1165. [Google Scholar] [CrossRef]
  13. Grotenhermen, F. Cannabinoids. Curr. Drug Targets CNS Neurol. Disord. 2005, 4, 507–530. [Google Scholar] [CrossRef]
  14. Hanuš, L.O. Pharmacological and Therapeutic Secrets of Plant and Brain (Endo)Cannabinoids. Med. Res. Rev. 2009, 29, 213–271. [Google Scholar] [CrossRef] [PubMed]
  15. Murphy, F.; Sales, P.; Murphy, S.; Averill, S.; Lau, N.; Sato, S.-O. Baby Boomers and Cannabis Delivery Systems. J. Drug Issues 2015, 45, 293–313. [Google Scholar] [CrossRef]
  16. Meyer, H.C.; Lee, F.S.; Gee, D.G. The Role of the Endocannabinoid System and Genetic Variation in Adolescent Brain Development. Int. J. Neuropsychopharmacol. 2018, 43, 21–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Nesto, R.W.; Mackie, K. Endocannabinoid System and Its Implications for Obesity and Cardiometabolic Risk. Eur. Heart J. Suppl. 2008, 10, B34–B41. [Google Scholar] [CrossRef] [Green Version]
  18. Ashton, J.C.; Friberg, D.; Darlington, C.L.; Smith, P.F. Expression of the Cannabinoid CB2 Receptor in the Rat Cerebellum: An Immunohistochemical Study. Neurosci. Lett. 2006, 396, 113–116. [Google Scholar] [CrossRef] [PubMed]
  19. Gong, J.P.; Onaivi, E.S.; Ishiguro, H.; Liu, Q.R.; Tagliaferro, P.A.; Brusco, A.; Uhl, G.R. Cannabinoid CB2 Receptors: Immunohistochemical Localization in Rat Brain. Brain Res. 2006, 1071, 10–23. [Google Scholar] [CrossRef]
  20. Mitrirattanakul, S.; Ramakul, N.; Guerrero, A.V.; Matsuka, Y.; Ono, T.; Iwase, H.; Mackie, K.; Faull, K.F.; Spigelman, I. Site-Specific Increases in Peripheral Cannabinoid Receptors and Their Endogenous Ligands in a Model of Neuropathic Pain. Pain 2006, 126, 102–114. [Google Scholar] [CrossRef] [Green Version]
  21. Maresz, K.; Pryce, G.; Ponomarev, E.D.; Marsicano, G.; Croxford, J.L.; Shriver, L.P.; Ledent, C.; Cheng, X.; Carrier, E.J.; Mann, M.K.; et al. Direct Suppression of CNS Autoimmune Inflammation via the Cannabinoid Receptor CB1 on Neurons and CB2 on Autoreactive T Cells. Nat. Med. 2007, 13, 492–497. [Google Scholar] [CrossRef] [PubMed]
  22. Lisman, J.; Buzsáki, G.; Eichenbaum, H.; Nadel, L.; Rangananth, C.; Redish, A.D. Viewpoints: How the Hippocampus Contributes to Memory, Navigation and Cognition. Nat. Neurosci. 2017, 20, 1434–1447. [Google Scholar] [CrossRef] [PubMed]
  23. Behrendt, R.P. Hippocampus as a Wormhole: Gateway to Consciousness. Wiley Interdiscip. Rev. Cogn. Sci. 2017, 8, e1446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Scarante, F.F.; Vila-Verde, C.; Detoni, V.L.; Ferreira-Junior, N.C.; Guimarães, F.S.; Campos, A.C. Cannabinoid Modulation of the Stressed Hippocampus. Front. Mol. Neurosci. 2017, 10, 411. [Google Scholar] [CrossRef] [Green Version]
  25. Busquets-Garcia, A.; Desprez, T.; Metna-Laurent, M.; Bellocchio, L.; Marsicano, G.; Soria-Gomez, E. Dissecting the Cannabinergic Control of Behavior: The Where Matters. Bioessays 2015, 37, 1215–1225. [Google Scholar] [CrossRef]
  26. Hampson, R.E.; Deadwyler, S.A. Cannabinoids, Hippocampal Function and Memory. Life Sci. 1999, 65, 715–723. [Google Scholar] [CrossRef]
  27. Gutiérrez-Rodríguez, A.; Bonilla-Del Río, I.; Puente, N.; Gómez-Urquijo, S.M.; Fontaine, C.J.; Egaña-Huguet, J.; Elezgarai, I.; Ruehle, S.; Lutz, B.; Robin, L.M.; et al. Localization of the Cannabinoid Type-1 Receptor in Subcellular Astrocyte Compartments of Mutant Mouse Hippocampus. Glia 2018, 66, 1417–1431. [Google Scholar] [CrossRef] [Green Version]
  28. Jimenez-Blasco, D.; Busquets-Garcia, A.; Hebert-Chatelain, E.; Serrat, R.; Vicente-Gutierrez, C.; Ioannidou, C.; Gómez-Sotres, P.; Lopez-Fabuel, I.; Resch-Beusher, M.; Resel, E.; et al. Glucose Metabolism Links Astroglial Mitochondria to Cannabinoid Effects. Nature 2020, 583, 603–608. [Google Scholar] [CrossRef]
  29. Marsicano, G.; Goodenough, S.; Monory, K.; Hermann, H.; Eder, M.; Cannich, A.; Azad, S.C.; Cascio, M.G.; Ortega-Gutiérrez, S.; van der Stelt, M.; et al. CB1 Cannabinoid Receptors and On-Demand Defense against Excitotoxicity. Science 2003, 302, 84–88. [Google Scholar] [CrossRef] [Green Version]
  30. Soria-Gomez, E.; Metna, M.; Bellocchio, L.; Busquets-Garcia, A.; Marsicano, G. The Endocannabinoid System in the Control of Behavior. In Handbook of Neurobehavioral Genetics and Phenotyping; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2017; pp. 323–355. [Google Scholar] [CrossRef]
  31. Degroot, A.; Köfalvi, A.; Wade, M.R.; Davis, R.J.; Rodrigues, R.J.; Rebola, N.; Cunha, R.A.; Nomikos, G.G. CB1 Receptor Antagonism Increases Hippocampal Acetylcholine Release: Site and Mechanism of Action. Mol. Pharmacol. 2006, 70, 1236–1245. [Google Scholar] [CrossRef]
  32. Tzavara, E.T.; Wade, M.; Nomikos, G.G. Biphasic Effects of Cannabinoids on Acetylcholine Release in the Hippocampus: Site and Mechanism of Action. J. Neurosci. 2003, 23, 9374–9384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Wyrofsky, R.R.; Reyes, B.A.S.; Zhang, X.Y.; Bhatnagar, S.; Kirby, L.G.; van Bockstaele, E.J. Endocannabinoids, Stress Signaling, and the Locus Coeruleus-Norepinephrine System. Neurobiol. Stress 2019, 11, 100176. [Google Scholar] [CrossRef] [PubMed]
  34. MacAskill, A.F.; Kittler, J.T. Control of Mitochondrial Transport and Localization in Neurons. Trends Cell Biol. 2010, 20, 102–112. [Google Scholar] [CrossRef] [PubMed]
  35. Attwell, D.; Laughlin, S.B. An Energy Budget for Signaling in the Grey Matter of the Brain. J. Cereb. 2001, 21, 1133–1145. [Google Scholar] [CrossRef]
  36. Mattson, M.P.; Gleichmann, M.; Cheng, A. Mitochondria in Neuroplasticity and Neurological Disorders. Neuron 2008, 60, 748–766. [Google Scholar] [CrossRef] [Green Version]
  37. Laughlin, S.B.; de Ruyter Van Steveninck, R.R.; Anderson, J.C. The Metabolic Cost of Neural Information. Nat. Neurosci. 1998, 1, 36–41. [Google Scholar] [CrossRef]
  38. Loureiro, M.; Renard, J.; Zunder, J.; Laviolette, S.R. Hippocampal Cannabinoid Transmission Modulates Dopamine Neuron Activity: Impact on Rewarding Memory Formation and Social Interaction. Int. J. Neuropsychopharmacol. 2014, 40, 1436–1447. [Google Scholar] [CrossRef] [Green Version]
  39. Hebert-Chatelain, E.; Desprez, T.; Serrat, R.; Bellocchio, L.; Soria-Gomez, E.; Busquets-Garcia, A.; Pagano Zottola, A.C.; Delamarre, A.; Cannich, A.; Vincent, P.; et al. A Cannabinoid Link between Mitochondria and Memory. Nature 2016, 539, 555–559. [Google Scholar] [CrossRef]
  40. Whyte, D.A.; Al-Hammadi, S.; Balhaj, G.; Brown, O.M.; Penefsky, H.S.; Souid, A.K. Cannabinoids Inhibit Cellular Respiration of Human Oral Cancer Cells. Pharmacology 2010, 85, 328–335. [Google Scholar] [CrossRef]
  41. Rubens, M. Political and Medical Views on Medical Marijuana and Its Future. Soc. Work Public Health 2014, 29, 121–131. [Google Scholar] [CrossRef] [PubMed]
  42. Devinsky, O.; Cilio, M.R.; Cross, H.; Fernandez-Ruiz, J.; French, J.; Hill, C.; Katz, R.; di Marzo, V.; Jutras-Aswad, D.; Notcutt, W.G.; et al. Cannabidiol: Pharmacology and Potential Therapeutic Role in Epilepsy and Other Neuropsychiatric Disorders. Epilepsia 2014, 55, 791–802. [Google Scholar] [CrossRef] [Green Version]
  43. Kalant, H. Medicinal Use of Cannabis: History and Current Status. Pain Res. Manag. 2001, 6, 80–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Huestis, M.A. Pharmacokinetics and Metabolism of the Plant Cannabinoids, Δ9-Tetrahydrocannibinol, Cannabidiol and Cannabinol. Handb. Exp. Pharmacol. 2005, 168, 657–690. [Google Scholar] [CrossRef]
  45. Lucas, C.J.; Galettis, P.; Schneider, J. The Pharmacokinetics and the Pharmacodynamics of Cannabinoids. Br. J. Clin. Pharmacol. 2018, 84, 2477–2482. [Google Scholar] [CrossRef] [Green Version]
  46. Gonçalves, J.; Rosado, T.; Soares, S.; Simão, A.Y.; Caramelo, D.; Luís, Â.; Fernández, N.; Barroso, M.; Gallardo, E.; Duarte, A.P. Cannabis and Its Secondary Metabolites: Their Use as Therapeutic Drugs, Toxicological Aspects, and Analytical Determination. Medicines 2019, 6, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Grotenhermen, F. Cannabinoids for therapeutic use. Am. J. Drug Deliv. 2004, 2, 229–240. [Google Scholar] [CrossRef]
  48. Zhornitsky, S.; Potvin, S. Cannabidiol in Humans—The Quest for Therapeutic Targets. Pharmaceuticals 2012, 5, 529–552. [Google Scholar] [CrossRef]
  49. Bridgeman, M.B.; Abazia, D.T. Medicinal Cannabis: History, Pharmacology, And Implications for the Acute Care Setting. Pharm. Ther. 2017, 42, 180. [Google Scholar]
  50. Badowski, M.E. A Review of Oral Cannabinoids and Medical Marijuana for the Treatment of Chemotherapy-Induced Nausea and Vomiting: A Focus on Pharmacokinetic Variability and Pharmacodynamics. Cancer Chemother. Pharmacol. 2017, 80, 441–449. [Google Scholar] [CrossRef] [Green Version]
  51. De Luis, D.A.; Sagrado, M.G.; Aller, R.; Izaola, O.; Conde, R.; Romero, E. C358A Missense Polymorphism of the Endocannabinoid Degrading Enzyme Fatty Acid Amide Hydrolase (FAAH) and Insulin Resistance in Patients with Diabetes Mellitus Type 2. Diabetes Res. Clin. Pract. 2010, 88, 76–80. [Google Scholar] [CrossRef]
  52. Pellesi, L.; Licata, M.; Verri, P.; Vandelli, D.; Palazzoli, F.; Marchesi, F.; Cainazzo, M.M.; Pini, L.A.; Guerzoni, S. Pharmacokinetics and Tolerability of Oral Cannabis Preparations in Patients with Medication Overuse Headache (MOH)—A Pilot Study. Eur. J. Clin. Pharmacol. 2018, 74, 1427–1436. [Google Scholar] [CrossRef]
  53. Arnold, J.C.; Hone, P.; Holland, M.L.; Allen, J.D. CB2 and TRPV1 Receptors Mediate Cannabinoid Actions on MDR1 Expression in Multidrug Resistant Cells. Pharmacol. Rep. 2012, 64, 751–757. [Google Scholar] [CrossRef]
  54. Agrawal, A.; Wetherill, L.; Dick, D.M.; Xuei, X.; Hinrichs, A.; Hesselbrock, V.; Kramer, J.; Nurnberger, J.I., Jr.; Schuckit, M.; Bierut, L.J.; et al. Evidence for association between polymorphisms in the cannabinoid receptor 1 (CNR1) gene and cannabis dependence. Am. J. Med. Genet. 2009, 150B, 736–740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Harismendy, O.; Bansal, V.; Bhatia, G.; Nakano, M.; Scott, M.; Wang, X.; Dib, C.; Turlotte, E.; Sipe, J.C.; Murray, S.S.; et al. Population Sequencing of Two Endocannabinoid Metabolic Genes Identifies Rare and Common Regulatory Variants Associated with Extreme Obesity and Metabolite Level. Genome Biol. 2010, 11, R118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Verdejo-García, A.; Beatriz Fagundo, A.; Cuenca, A.; Rodriguez, J.; Cuyás, E.; Langohr, K.; de Sola Llopis, S.; Civit, E.; Farré, M.; Peña-Casanova, J.; et al. COMT Val158met and 5-HTTLPR Genetic Polymorphisms Moderate Executive Control in Cannabis Users. Int. J. Neuropsychopharmacol. 2013, 38, 1598–1606. [Google Scholar] [CrossRef] [Green Version]
  57. Agrawal, A.; Edenberg, H.J.; Foroud, T.; Bierut, L.J.; Dunne, G.; Hinrichs, A.L.; Nurnberger, J.I.; Crowe, R.; Kuperman, S.; Schuckit, M.A.; et al. Association of GABRA2 with Drug Dependence in the Collaborative Study of the Genetics of Alcoholism Sample. Behav. Genet. 2006, 36, 640–650. [Google Scholar] [CrossRef]
  58. Gelernter, J.; Kranzler, H.; Cubells, J. Genetics of Two μ Opioid Receptor Gene (OPRM1) Exon I Polymorphisms: Population Studies, and Allele Frequencies in Alcohol- and Drug-Dependent Subjects. Mol. Psychiatry 1999, 4, 476–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Han, S.; Yang, B.Z.; Kranzler, H.R.; Oslin, D.; Anton, R.; Farrer, L.A.; Gelernter, J. Linkage Analysis Followed by Association Show NRG1 Associated with Cannabis Dependence in African Americans. Biol. Psychiatry 2012, 72, 637–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Zhang, P.W.; Ishiguro, H.; Ohtsuki, T.; Hess, J.; Carillo, F.; Walther, D.; Onaivi, E.S.; Arinami, T.; Uhl, G.R. Human Cannabinoid Receptor 1: 5′ Exons, Candidate Regulatory Regions, Polymorphisms, Haplotypes and Association with Polysubstance Abuse. Mol. Psychiatry 2004, 9, 916–931. [Google Scholar] [CrossRef] [Green Version]
  61. Di Marzo, V.; Matias, I. Endocannabinoid Control of Food Intake and Energy Balance. Nat. Neurosci. 2005, 8, 585–589. [Google Scholar] [CrossRef] [PubMed]
  62. Li, T.; Liu, X.; Zhu, Z.H.; Zhao, J.; Hu, X.; Ball, D.M.; Sham, P.C.; Collier, D.A. No Association between (AAT)n Repeats in the Cannabinoid Receptor Gene (CNR1) and Heroin Abuse in a Chinese Population. Mol. Psychiatry 2000, 5, 128–130. [Google Scholar] [CrossRef] [PubMed]
  63. Covault, J.; Gelernter, J.; Kranzler, H. Association Study of Cannabinoid Receptor Gene (CNR1) Alleles and Drug Dependence. Mol. Psychiatry 2001, 6, 501–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bühler, K.M.; Huertas, E.; Echeverry-Alzate, V.; Giné, E.; Moltó, E.; Montoliu, L.; López-Moreno, J.A. Risky Alcohol Consumption in Young People Is Associated with the Fatty Acid Amide Hydrolase Gene Polymorphism C385A and Affective Rating of Drug Pictures. Mol. Genet. Genom. 2014, 289, 279–289. [Google Scholar] [CrossRef]
  65. Hartman, C.A.; Hopfer, C.J.; Haberstick, B.; Rhee, S.H.; Crowley, T.J.; Corley, R.P.; Hewitt, J.K.; Ehringer, M.A. The Association between Cannabinoid Receptor 1 Gene (CNR1) and Cannabis Dependence Symptoms in Adolescents and Young Adults. Drug Alcohol Depend. 2009, 104, 11–16. [Google Scholar] [CrossRef] [Green Version]
  66. Carrasquer, A.; Nebane, N.M.; Williams, W.M.; Song, Z.H. Functional Consequences of Nonsynonymous Single Nucleotide Polymorphisms in the CB2 Cannabinoid Receptor. Pharmacogenet. Genom. 2010, 20, 157–166. [Google Scholar] [CrossRef]
  67. Bíró, T.; Tóth, B.I.; Marincsák, R.; Dobrosi, N.; Géczy, T.; Paus, R. TRP Channels as Novel Players in the Pathogenesis and Therapy of Itch. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2007, 1772, 1004–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Diao, X.; Scheidweiler, K.B.; Wohlfarth, A.; Zhu, M.; Pang, S.; Huestis, M.A. Strategies to Distinguish New Synthetic Cannabinoid FUBIMINA (BIM-2201) Intake from Its Isomer THJ-2201: Metabolism of FUBIMINA in Human Hepatocytes. Forensic Toxicol. 2016, 34, 256–267. [Google Scholar] [CrossRef] [Green Version]
  69. Stout, S.M.; Cimino, N.M. Exogenous Cannabinoids as Substrates, Inhibitors, and Inducers of Human Drug Metabolizing Enzymes: A Systematic Review. Drug Metab. Rev. 2014, 46, 86–95. [Google Scholar] [CrossRef] [Green Version]
  70. Evans, W.E.; Relling, M.V. Pharmacogenomics: Translating Functional Genomics into Rational Therapeutics. Science 1999, 286, 487–491. [Google Scholar] [CrossRef] [Green Version]
  71. Zhang, X.; Meng, X.; Wang, Y.; Yan, W.; Yang, J. Comprehensive Analysis of UGT1A1 Genetic Polymorphisms in Chinese Tibetan and Han Populations. Biochem. Genet. 2012, 50, 967–977. [Google Scholar] [CrossRef]
  72. Ingelman-Sundberg, M. Genetic Susceptibility to Adverse Effects of Drugs and Environmental Toxicants: The Role of the CYP Family of Enzymes. Mutat. Res. Fund. Mol. M 2001, 482, 11–19. [Google Scholar] [CrossRef] [PubMed]
  73. Saghafi, F.; Salehifar, E.; Janbabai, G.; Zaboli, E.; Hedayatizadeh-Omran, A.; Amjadi, O.; Moradi, S. CYP2D6*3 (A2549del), *4 (G1846A), *10 (C100T) and *17 (C1023T) Genetic Polymorphisms in Iranian Breast Cancer Patients Treated with Adjuvant Tamoxifen. Biomed. Rep. 2018, 9, 446. [Google Scholar] [CrossRef] [Green Version]
  74. Murayama, N.; Nakamura, T.; Saeki, M.; Soyama, A.; Saito, Y.; Sai, K.; Ishida, S.; Itoda, M.; Ozawa, S.; Sawada, J.I.; et al. CYP3A4 Gene Polymorphisms Influence Testosterone 6β-Hydroxylation. Drug Metab. Pharmacokinet. 2002, 17, 150–156. [Google Scholar] [CrossRef] [PubMed]
  75. Mailloux, J.; Medwid, S.; Facey, A.; Sung, I.; Russell, L.E.; Tirona, R.G.; Kim, R.B.; Schwarz, U.I. In-Vitro Characterization of Coding Variants with Predicted Functional Implications in the Efflux Transporter Multidrug Resistance Protein 4 (MRP4, ABCC4). Pharm. Genet. Genom. 2022, 32, 111–116. [Google Scholar] [CrossRef] [PubMed]
  76. Dai, D.P.; Wang, Y.H.; Wang, S.H.; Geng, P.W.; Hu, L.M.; Hu, G.X.; Cai, J.P. In Vitro Functional Characterization of 37 CYP2C9 Allelic Isoforms Found in Chinese Han Population. Acta Pharmacol. 2013, 34, 1449–1456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Villeneuve, L.; Girard, H.; Fortier, L.C.; Gagné, J.F.; Guillemette, C. Novel Functional Polymorphisms in the UGT1A7 and UGT1A9 Glucuronidating Enzymes in Caucasian and African-American Subjects and Their Impact on the Metabolism of 7-Ethyl-10-Hydroxycamptothecin and Flavopiridol Anticancer Drugs. J. Pharmacol. Exp. Ther. 2003, 307, 117–128. [Google Scholar] [CrossRef] [Green Version]
  78. Saeki, M.; Saito, Y.; Jinno, H.; Sai, K.; Ozawa, S.; Sawada, J.I.; Komamura, K.; Kamakura, S.; Kitakaze, M.; Ueno, K.; et al. Three Novel Single Nucleotide Polymorphisms in UGT1A9. Drug Metab. Pharmacokinet. 2003, 18, 146–149. [Google Scholar] [CrossRef]
  79. Jinno, H.; Saeki, M.; Saito, Y.; Tanaka-Kagawa, T.; Hanioka, N.; Sai, K.; Kaniwa, N.; Ando, M.; Shirao, K.; Minami, H.; et al. Functional Characterization of Human UDP-Glucuronosyltransferase 1A9 Variant, D256N, Found in Japanese Cancer Patients. J. Pharmacol. Exp. Ther. 2003, 306, 688–693. [Google Scholar] [CrossRef] [Green Version]
  80. Bosma, P.J. Inherited Disorders of Bilirubin Metabolism. J. Hepatol. 2003, 38, 107–117. [Google Scholar] [CrossRef]
  81. Moghrabi, N.; Sutherland, L.; Wooster, R.; Povey, S.; Boxer, M.; Burchell, B. Chromosomal Assignment of Human Phenol and Bilirubin UDP-Glucuronosyltransferase Genes (UGT1A-Subfamily). Ann. Hum. Genet. 1992, 56, 81–91. [Google Scholar] [CrossRef]
  82. Miles, K.; Stern, S.; Smith, P.; Kessler, F.; Ali, S.; Ritter, J. An investigation of human and rat liver microsomal mycophenolic acid glucuronidation: Evidence for a principal role of UGT1A enzymes and species differences in UGT1A specificity. Drug Metab. Dispos. 2005, 33, 1513–1520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Volkow, N.D.; Baler, R.D.; Compton, W.M.; Weiss, S.R.B. Adverse Health Effects of Marijuana Use. N. Engl. J. Med. 2014, 370, 2219–2227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Carliner, H.; Brown, Q.L.; Sarvet, A.L.; Hasin, D.S. Cannabis Use, Attitudes, and Legal Status in the US: A Review. Prev. Med. 2017, 104, 13–23. [Google Scholar] [CrossRef] [PubMed]
  85. Broyd, S.J.; van Hell, H.H.; Beale, C.; Yuecel, M.; Solowij, N. Acute and Chronic Effects of Cannabinoids on Human Cognition—A Systematic Review. Biol. Psychiatry 2016, 79, 557–567. [Google Scholar] [CrossRef] [Green Version]
  86. Duperrouzel, J.C.; Granja, K.; Pacheco-Colón, I.; Gonzalez, R. Adverse Effects of Cannabis Use on Neurocognitive Functioning: A Systematic Review of Meta-Analytic Studies. J. Dual Diagn. 2020, 16, 43–57. [Google Scholar] [CrossRef]
  87. Lorenzetti, V.; Chye, Y.; Silva, P.; Solowij, N.; Roberts, C.A. Does Regular Cannabis Use Affect Neuroanatomy? An Updated Systematic Review and Meta-Analysis of Structural Neuroimaging Studies. Eur. Arch. Psychiatry Clin. Neurosci. 2019, 269, 59–71. [Google Scholar] [CrossRef] [PubMed]
  88. Nusbaum, A.T.; Whitney, P.; Cuttler, C.; Spradlin, A.; Hinson, J.M.; McLaughlin, R.J. Altered Attentional Control Strategies but Spared Executive Functioning in Chronic Cannabis Users. Drug Alcohol Depend. 2017, 181, 116–123. [Google Scholar] [CrossRef]
  89. Becker, M.P.; Collins, P.F.; Luciana, M. Neurocognition in College-Aged Daily Marijuana Users. J. Clin. Exp. Neuropsychol. 2014, 36, 379–398. [Google Scholar] [CrossRef] [Green Version]
  90. Gonzalez, R.; Schuster, R.M.; Mermelstein, R.J.; Vassileva, J.; Martin, E.M.; Diviak, K.R. Performance of Young Adult Cannabis Users on Neurocognitive Measures of Impulsive Behavior and Their Relationship to Symptoms of Cannabis Use Disorders. J. Clin. Exp. Neuropsychol. 2012, 34, 962–976. [Google Scholar] [CrossRef] [Green Version]
  91. Grant, J.E.; Chamberlain, S.R.; Schreiber, L.; Odlaug, B.L. Neuropsychological Deficits Associated with Cannabis Use in Young Adults. Drug Alcohol Depend. 2012, 121, 159–162. [Google Scholar] [CrossRef] [Green Version]
  92. Griffith-Lendering, M.F.H.; Huijbregts, S.C.J.; Vollebergh, W.A.M.; Swaab, H. Motivational and Cognitive Inhibitory Control in Recreational Cannabis Users. J. Clin. Exp. Neuropsychol. 2012, 34, 688–697. [Google Scholar] [CrossRef] [PubMed]
  93. Bartholomew, J.; Holroyd, S.; Heffernan, T.M. Does Cannabis Use Affect Prospective Memory in Young Adults? J. Psychopharmacol. 2010, 24, 241–246. [Google Scholar] [CrossRef] [PubMed]
  94. Huestegge, L.; Kunert, H.-J.; Radach, R. Long-Term Effects of Cannabis on Eye Movement Control in Reading. Psychopharmacology 2010, 209, 77–84. [Google Scholar] [CrossRef] [PubMed]
  95. Curran, H.V.; Freeman, T.P.; Mokrysz, C.; Lewis, D.A.; Morgan, C.J.A.; Parsons, L.H. Keep off the Grass? Cannabis, Cognition and Addiction. Nat. Rev. Neurosci. 2016, 17, 293–306. [Google Scholar] [CrossRef] [PubMed]
  96. Ganzer, F.; Bröning, S.; Kraft, S.; Sack, P.-M.; Thomasius, R. Weighing the Evidence: A Systematic Review on Long-Term Neurocognitive Effects of Cannabis Use in Abstinent Adolescents and Adults. Neuropsychol. Rev. 2016, 26, 186–222. [Google Scholar] [CrossRef]
  97. Lisdahl, K.M.; Wright, N.E.; Medina-Kirchner, C.; Maple, K.E.; Shollenbarger, S. Considering Cannabis: The Effects of Regular Cannabis Use on Neurocognition in Adolescents and Young Adults. Curr. Addict. Rep. 2014, 1, 144–156. [Google Scholar] [CrossRef] [Green Version]
  98. Lubman, D.I.; Cheetham, A.; Yücel, M. Cannabis and Adolescent Brain Development. Pharmacol. Ther. 2015, 148, 1–16. [Google Scholar] [CrossRef]
  99. Landrigan, J.; Bessenyei, K.; Leitner, D.; Yakovenko, I.; Fisk, J.D.; Prentice, J.L. A Systematic Review of the Effects of Cannabis on Cognition in People with Multiple Sclerosis. Mult. Scler. Relat. Disord. 2022, 57, 103338. [Google Scholar] [CrossRef]
  100. Cohen, K.; Weizman, A.; Weinstein, A. Positive and Negative Effects of Cannabis and Cannabinoids on Health. Clin. Pharmacol. Ther. 2019, 105, 1139–1147. [Google Scholar] [CrossRef]
  101. Baxter, A.G. The Origin and Application of Experimental Autoimmune Encephalomyelitis. Nat. Rev. Immunol. 2007, 7, 904–912. [Google Scholar] [CrossRef]
  102. Lyman, W.; Sonett, J.; Brosnan, C.; Bornstein, M. Δ9-Tetrahydrocannabinol: A novel treatment for experimental autoimmune encephalomyelitis. J. Neuroimmunol. 1989, 23, 73–81. [Google Scholar] [CrossRef] [PubMed]
  103. Wirguin, I.; Mechoulam, R.; Breuer, A.; Schezen, E.; Weidenfeld, J.; Brenner, T. Suppression of Experimetal Autoimmune Encephalomyelitis by Cannabinoids. Immunopharmacology 1994, 28, 209–214. [Google Scholar] [CrossRef] [PubMed]
  104. Achiron, A.; Miron, S.; Lavie, V.; Margalit, R.; Biegon, A. Dexanabinol (HU-211) Effect on Experimental Autoimmune Encephalomyelitis: Implications for the Treatment of Acute Relapses of Multiple Sclerosis. J. Neuroimmunol. 2000, 102, 26–31. [Google Scholar] [CrossRef] [PubMed]
  105. 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, 44–48. [Google Scholar] [CrossRef] [PubMed]
  106. Petro, D.J.; Ellenberger, C. Treatment of Human Spasticity with Δ9-Tetrahydrocannabinol. J. Clin. Pharmacol. 1981, 21, 413S–416S. [Google Scholar] [CrossRef] [PubMed]
  107. Hamann, W.; di Vadi, P.P. Analgesic Effect of the Cannabinoid Analogue Nabilone Is Not Mediated by Opioid Receptors. Lancet 1999, 353, 560. [Google Scholar] [CrossRef]
  108. Brenneisen, R.; Egli, A.; Elsohly, M.A.; Henn, V.; Spiess, Y. The Effect of Orally and Rectally Administered Delta 9-Tetrahydrocannabinol on Spasticity: A Pilot Study with 2 Patients. Int. J. Clin. Pharmacol. Ther. 1996, 34, 446–452. [Google Scholar]
  109. Ungerleider, J.T.; Andyrsiak, T.; Fairbanks, L.; Ellison, G.W.; Myers, L.W. Delta-9-THC in the Treatment of Spasticity Associated with Multiple Sclerosis. Adv. Alcohol Subst. Abuse 1988, 7, 39–50. [Google Scholar] [CrossRef]
  110. Rog, D.J.; Nurmikko, T.J.; Friede, T.; Young, C.A. Randomized, Controlled Trial of Cannabis-Based Medicine in Central Pain in Multiple Sclerosis. Neurology 2005, 65, 812–819. [Google Scholar] [CrossRef]
  111. Schon, F.; Hart, P.E.; Hodgson, T.L.; Pambakian, A.L.M.; Ruprah, M.; Williamson, E.M.; Kennard, C. Suppression of Pendular Nystagmus by Smoking Cannabis in a Patient with Multiple Sclerosis. Neurology 1999, 53, 2209. [Google Scholar] [CrossRef]
  112. Clifford, D.B. Tetrahydrocannabinol for Tremor in Multiple Sclerosis. Ann. Neurol. 1983, 13, 669–671. [Google Scholar] [CrossRef] [PubMed]
  113. Meinck, H.M.; Schönle, P.W.; Conrad, B. Effect of Cannabinoids on Spasticity and Ataxia in Multiple Sclerosis. J. Neurol. 1989, 236, 120–122. [Google Scholar] [CrossRef] [PubMed]
  114. Martyn, C.N.; Illis, L.S.; Thom, J. Nabilone in the Treatment of Multiple Sclerosis. Lancet 1995, 345, 579. [Google Scholar] [CrossRef] [PubMed]
  115. Wade, D.T.; Robson, P.; House, H.; Makela, P.; Aram, J. A Preliminary Controlled Study to Determine Whether Whole-Plant Cannabis Extracts Can Improve Intractable Neurogenic Symptoms. Clin. Rehabil. 2003, 17, 21–29. [Google Scholar] [CrossRef]
  116. Markovà, J.; Essner, U.; Akmaz, B.; Marinelli, M.; Trompke, C.; Lentschat, A.; Vila, C. Sativex® as Add-on Therapy vs. Further Optimized First-Line ANTispastics (SAVANT) in Resistant Multiple Sclerosis Spasticity: A Double-Blind, Placebo-Controlled Randomised Clinical Trial. Int. J. Neurosci. 2019, 129, 119–128. [Google Scholar] [CrossRef]
  117. Flachenecker, P.; Henze, T.; Zettl, U.K. Nabiximols (THC/CBD Oromucosal Spray, Sativex®) in Clinical Practice—Results of a Multicenter, Non-Interventional Study (MOVE 2) in Patients with Multiple Sclerosis Spasticity. Eur. Neurol. 2014, 71, 271–279. [Google Scholar] [CrossRef]
  118. Squintani, G.; Donato, F.; Turri, M.; Deotto, L.; Teatini, F.; Moretto, G.; Erro, R. Cortical and Spinal Excitability in Patients with Multiple Sclerosis and Spasticity after Oromucosal Cannabinoid Spray. J. Neurol. Sci. 2016, 370, 263–268. [Google Scholar] [CrossRef]
  119. Russo, M.; Naro, A.; Leo, A.; Sessa, E.; D’Aleo, G.; Bramanti, P.; Calabrò, R.S. Evaluating Sativex® in Neuropathic Pain Management: A Clinical and Neurophysiological Assessment in Multiple Sclerosis. Pain Med. 2016, 17, 1145–1154. [Google Scholar] [CrossRef] [Green Version]
  120. Sorosina, M.; Clarelli, F.; Ferr, L.; Osiceanu, A.M.; Unal, N.T.; Mascia, E.; Martinelli, V.; Comi, G.; Benigni, F.; Esposito, F.; et al. Clinical Response to Nabiximols Correlates with the Downregulation of Immune Pathways in Multiple Sclerosis. Eur. J. Neurol. 2018, 25, 70. [Google Scholar] [CrossRef]
  121. Turri, M.; Teatini, F.; Donato, F.; Zanette, G.; Tugnoli, V.; Deotto, L.; Bonetti, B.; Squintani, G. Pain Modulation after Oromucosal Cannabinoid Spray (SATIVEX®) in Patients with Multiple Sclerosis: A Study with Quantitative Sensory Testing and Laser-Evoked Potentials. Medicines 2018, 5, 59. [Google Scholar] [CrossRef] [Green Version]
  122. Patti, F.; Messina, S.; Solaro, C.; Amato, M.P.; Bergamaschi, R.; Bonavita, S.; Bossio, R.B.; Morra, V.B.; Costantino, G.F.; Cavalla, P.; et al. Efficacy and Safety of Cannabinoid Oromucosal Spray for Multiple Sclerosis Spasticity. J. Neurol. Neurosurg. Psychiatry 2016, 87, 944–951. [Google Scholar] [CrossRef] [PubMed]
  123. Nichols, J.M.; Kummari, E.; Sherman, J.; Yang, E.J.; Dhital, S.; Gilfeather, C.; Yray, G.; Morgan, T.; Kaplan, B.L.F. CBD Suppression of EAE Is Correlated with Early Inhibition of Splenic IFN-γ + CD8+ T Cells and Modest Inhibition of Neuroinflammation. J. Neuroimmune Pharmacol. 2021, 16, 346–362. [Google Scholar] [CrossRef] [PubMed]
  124. Al-Ghezi, Z.Z.; Miranda, K.; Nagarkatti, M.; Nagarkatti, P.S. Combination of Cannabinoids, Δ9-Tetrahydrocannabinol and Cannabidiol, Ameliorates Experimental Multiple Sclerosis by Suppressing Neuroinflammation through Regulation of MiRNA-Mediated Signaling Pathways. Front. Immunol. 2019, 10, 1921. [Google Scholar] [CrossRef]
  125. Klein, T.W.; Newton, C.; Friedman, H. Cannabinoid Receptors and Immunity. Immunol. Today 1998, 19, 373–381. [Google Scholar] [CrossRef]
  126. Nahas, G.G.; Morishima, A.; Desoize, B. Effects of Cannabinoids on Macromolecular Synthesis and Replication of Cultured Lymphocytes. Fed. Proc. 1977, 36, 1748–1752. [Google Scholar]
  127. Klein, T.W.; Kawakami, Y.; Newton, C.; Friedman, H. Marijuana Components Suppress Induction and Cytolytic Function of Murine Cytotoxic T Cells In Vitro and In Vivo. J. Toxicol. Environ. Health 1991, 32, 465–477. [Google Scholar] [CrossRef] [PubMed]
  128. Yang, X.; Bam, M.; Nagarkatti, P.S.; Nagarkatti, M. Cannabidiol Regulates Gene Expression in Encephalitogenic T Cells Using Histone Methylation and Noncoding RNA during Experimental Autoimmune Encephalomyelitis. Sci. Rep. 2019, 9, 15780. [Google Scholar] [CrossRef] [Green Version]
  129. Kozela, E.; Juknat, A.; Kaushansky, N.; Rimmerman, N.; Ben-Nun, A.; Vogel, Z. Cannabinoids Decrease the Th17 Inflammatory Autoimmune Phenotype. J. Neuroimmune Pharmacol. 2013, 8, 1265–1276. [Google Scholar] [CrossRef]
  130. Manera, C.; Bertini, S. Cannabinoid-Based Medicines and Multiple Sclerosis. Adv. Exp. Med. Biol. 2021, 1264, 111–129. [Google Scholar] [CrossRef]
  131. Allsop, D.J.; Copeland, J.; Lintzeris, N.; Dunlop, A.J.; Montebello, M.; Sadler, C.; Rivas, G.R.; Holland, R.M.; Muhleisen, P.; Norberg, M.M.; et al. Nabiximols as an Agonist Replacement Therapy during Cannabis Withdrawal: A Randomized Clinical Trial. JAMA Psychiatry 2014, 71, 281–291. [Google Scholar] [CrossRef] [Green Version]
  132. Nahas, G.G.; Frick, H.C.; Lattimer, J.K.; Latour, C.; Harvey, D. Pharmacokinetics of THC in Brain and Testis, Male Gametotoxicity and Premature Apoptosis of Spermatozoa. Hum. Psychopharmacol. Clin. Exp. 2002, 17, 103–113. [Google Scholar] [CrossRef] [PubMed]
  133. Sieradzan, K.A.; Fox, S.H.; Hill, M.; Dick, J.P.R.; Crossman, A.R.; Brotchie, J.M. Cannabinoids Reduce Levodopa-Induced Dyskinesia in Parkinson’s Disease: A Pilot Study. Neurology 2001, 57, 2108–2111. [Google Scholar] [CrossRef]
  134. Volicer, L.; Stelly, M.; Morris, J.; McLaughlin, J.; Volicer, B.J. Effects of Dronabinol on Anorexia and Disturbed Behavior in Patients with Alzheimer’s Disease. Int. J. Geriatr. Psychiatry 1997, 12, 913–919. [Google Scholar] [CrossRef]
  135. Walther, S.; Mahlberg, R.; Eichmann, U.; Kunz, D. Delta-9-Tetrahydrocannabinol for Nighttime Agitation in Severe Dementia. Psychopharmacology 2006, 185, 524–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Davis, M.P. Oral Nabilone Capsules in the Treatment of Chemotherapy-Induced Nausea and Vomiting and Pain. Expert Opin. Investig. Drugs 2008, 17, 85–95. [Google Scholar] [CrossRef] [PubMed]
  137. Zhu, Y.; Ouyang, Z.; Du, H.; Wang, M.; Wang, J.; Sun, H.; Kong, L.; Xu, Q.; Ma, H.; Sun, Y. New Opportunities and Challenges of Natural Products Research: When Target Identification Meets Single-Cell Multiomics. Acta Pharm. Sin. B 2022, 12, 4011–4039. [Google Scholar] [CrossRef]
Figure 1. Illustration of the endocannabinoid system and distribution of CB1 receptors. CB1: cannabinoid-1 receptor, CB2: cannabinoid-2 receptor, THC: tetrahydrocannabinol, CBD: cannabinoid.
Figure 1. Illustration of the endocannabinoid system and distribution of CB1 receptors. CB1: cannabinoid-1 receptor, CB2: cannabinoid-2 receptor, THC: tetrahydrocannabinol, CBD: cannabinoid.
Pharmaceutics 15 01151 g001
Figure 2. The physiological function of the CB2 receptor during leukocyte transmigration and inflammation, the transport of immune cells across the BBB is restricted by CB2 receptors. CB2: cannabinoid-2 receptor, NK: natural killer cells, B cells: B lymphocytes cells.
Figure 2. The physiological function of the CB2 receptor during leukocyte transmigration and inflammation, the transport of immune cells across the BBB is restricted by CB2 receptors. CB2: cannabinoid-2 receptor, NK: natural killer cells, B cells: B lymphocytes cells.
Pharmaceutics 15 01151 g002
Table 2. Genetic variation and their relation to cannabinoids response.
Table 2. Genetic variation and their relation to cannabinoids response.
ProteinGeneFunctionVariationEffectRef.
CB1CNR1Receptor63-9597T > CCannabinoids addiction[54]
CB2CNR2Receptor946C > TThe effect and the main function of the CB2 receptor are altered[54]
FAAH proteinFatty Acid Amide Hydrolase
FAAH
Biotransformation385C > AIt is associated with drug abuse[55]
Catechol-O-methyltransferase enzymeCOMTThe regulation and inactivation of catecholamine neurotransmitters in the brain472A > GModest controllable effect of cannabinoids consumption on executive functions[56]
GABAGABRA2Receptor231A > GNo significant effect on the drug dependence[57]
Mu opioid receptorOPRM1Receptor118A > GNo significant effect[58]
ErbB3, ErbB4NRG1Promotes the growth, differentiation, and survival of a wide range of cell types122-16329C > TAssociated with cannabinoids dependence[59]
Table 3. Various alleles and their relation to the metabolizing enzyme activity.
Table 3. Various alleles and their relation to the metabolizing enzyme activity.
GeneAlleleNucleotide ChangeEffectRef.
CYP2D6CYP2D6*3A2549delIt will produce a protein with little or no function, which means increased activity of cannabinoids in the body.[73]
CYP2D6CYP2D6*4G1846AThe activity of the enzyme is reduced, causing drug accumulation in the body.[73]
CYP2C9CYP2C9*2c.430C > TThe activity of the enzyme is reduced, causing drug accumulation in the body.[74]
CYP2C9CYP2C9*3c.1075AThe activity of the enzyme is reduced, causing drug accumulation in the body.[74]
CYP3A4CYP3A4*2664 T-CThe activity of the enzyme is reduced, causing an increase in the drug’s half-life time.[74]
CYP3A4CYP3A4*111088 C-TThe activity of the enzyme is reduced, causing an increase in the drug’s half-life time.[75]
CYP3A4CYP3A4*121117 C-TThe activity of the enzyme is reduced, causing an increase in the drug’s half-life time.[76]
CYP3A4CYP3A4*17566 T-CDecrease in the enzyme activity, which increases the half-life of the drug.[69]
UGT1AUGT1A9*3a98T-CReduction or inactivation of the enzyme. [77]
UGT1AUGT1A9*4726 T-GReduction or inactivation of the enzyme. [78]
UGT1AUGT1A9* 5766 G-AReduction or inactivation of the enzyme. [79]
UGT1AUGT1A1*31124 C-TIt will lead to the inactivation of the enzyme.[80]
UGT1AUGT1A1*101021C-TDimension in the enzyme activity. [81]
UGT1AUGT1A1*13508-510delInactivation of the enzyme. [82]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nouh, R.A.; Kamal, A.; Abdelnaser, A. Cannabinoids and Multiple Sclerosis: A Critical Analysis of Therapeutic Potentials and Safety Concerns. Pharmaceutics 2023, 15, 1151. https://doi.org/10.3390/pharmaceutics15041151

AMA Style

Nouh RA, Kamal A, Abdelnaser A. Cannabinoids and Multiple Sclerosis: A Critical Analysis of Therapeutic Potentials and Safety Concerns. Pharmaceutics. 2023; 15(4):1151. https://doi.org/10.3390/pharmaceutics15041151

Chicago/Turabian Style

Nouh, Roua A., Ahmed Kamal, and Anwar Abdelnaser. 2023. "Cannabinoids and Multiple Sclerosis: A Critical Analysis of Therapeutic Potentials and Safety Concerns" Pharmaceutics 15, no. 4: 1151. https://doi.org/10.3390/pharmaceutics15041151

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop