IMR Press / FBE / Volume 14 / Issue 1 / DOI: 10.31083/j.fbe1401001
Open Access Review
Autism and associated disorders: cannabis as a potential therapy
Show Less
1 Department of Biomedical and Pharmaceutical Sciences, Touro College of Pharmacy, New York, NY 10027, USA
2 Department of Medicinal Chemistry, School of Pharmacy, College of Health Sciences, Mekelle University, 231 Tigray, Ethiopia
3 Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA
*Correspondence: mariana.babayeva@touro.edu (Mariana Babayeva)
Academic Editor: Igor Lavrov
Front. Biosci. (Elite Ed) 2022, 14(1), 1; https://doi.org/10.31083/j.fbe1401001
Submitted: 25 October 2021 | Revised: 23 November 2021 | Accepted: 10 December 2021 | Published: 13 January 2022
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Autism spectrum disorder (ASD) is a group of disabilities with impairments in physical, verbal, and behavior areas. Regardless the growing frequency of autism, no medicine has been formed for the management of the ASD primary symptoms. The most frequently prescribed drugs are off-label. Therefore, there is necessity for an advance tactic for the treatment of autism. The endocannabinoid system has a central role in ruling emotion and social behaviors. Dysfunctions of the system donate to the behavioral deficits in autism. Therefore, the endocannabinoid system represents a potential target for the development of a novel autism therapy. Cannabis and associated compounds have produced substantial research attention as a capable therapy in neurobehavioral and neurological syndromes. In this review we examine the potential benefits of medical cannabis and related compounds in the treatment of ASD and concurrent disorders.

Keywords
Autism
Endocannabinoid system
Medical cannabis
CBD
THC
CBDV
1. Introduction

Autism spectrum disorders is neurodevelopment disorders with wide range of impairments in social communication and restricted and repetitive behaviors [1]. The intellectual capability of individuals with ASDs is highly variable and ranges from severe impairment to superior performance. Both, Autism and ASD are used interchangeably. In the USA, the prevalence of ASD is approximately 4.5 times greater in boys than in girls [2]. ASD occurs in all racial, ethnic, and socioeconomic groups, although white children are more likely to be diagnosed with ASD than black or Hispanic [1]. According to the WHO, on a worldwide basis, it is estimated that 1 in 160 children exhibit ASD [3].

Autism is a behavioral diagnosis, the exact cause of which is unknown and currently biomarkers have not been identified. Several factors including environmental, biological and genetics play a role in the pathogenesis of ASD. Approximately 15–20% of ASD cases were found to be associated with genetic mutations [4]. Fragile X mental retardation 1 (FMR1) is the most common single gene mutation identified in autistic individuals [5, 6, 7, 8, 9]. Other single gene mutations that have been shown to be associated with ASD include tuberous sclerosis, neurofibromatosis, Angelman syndrome, and Rett syndrome [5, 10]. Immune dysfunction and inflammation as well as fetal exposure to antiepileptic drugs contribute to the pathogenesis of autism [11, 12, 13, 14, 15, 16, 17].

The endocannabinoid system (ECS) has been investigated for its association with ASD because of its role in regulating emotion and social behaviors. The endocannabinoid system involves the cannabinoid receptors (CB1 and CB2) and their endogenous ligands (the endocannabinoids) as well as the enzymes involved in the biosynthesis and inactivation of the endocannabinoids [18]. The primary endocannabinoids are N-arachidonoyl-ethanolamine (anandamide, AEA) and 2-arachidonoylglycerol (2-AG). Enzymes involved in the synthesis of AEA and 2-AG are N-acylphosphatidylethanolamine-specific phospholipase D (NAPE-PLD) and diacylglycerol lipase (DAGL), respectively. Endocannabinoids are inactivated by hydrolytic enzymes, fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL). Inhibitors of FAAH and MAGL may influence signaling of ECS. The endocannabinoids act as retrograde messengers on presynaptic cannabinoid receptors to lower the release of neurotransmitters (e.g., monoamine, opioids, GABA, glutamate, acetylcholine) and to impact a wide range of biological processes [18].

Several studies have suggested that dysfunctions in the components of the endocannabinoid system may contribute to the behavioral deficits and neuroinflammation observed in autism [19, 20, 21, 22, 23, 24]. In animal models of ASD, modulation of the endocannabinoid system has been shown to improve certain ASD-associated social and cognitive impairments [25, 26, 27, 28].

Although the prevalence of autism is increasing [29], a pharmaceutical has not been developed for the treatment of the core symptoms of ASD. Management of ASD calls for a multidisciplinary approach and mainly involves behavioral and educational interventions [1, 30, 31]. Pharmacological therapy attempts to address ASD-associated comorbidities including seizure, violent behavior, psychosis, anxiety, depression, bipolar disorder, and attention-deficit hyperactivity disorder [30, 31, 32]. The most frequently prescribed classes of drugs for ASD patients include antidepressants, stimulants, antipsychotics, anticonvulsants, hypotensive agents, anxiolytics/sedatives/hypnotics, and melatonin [33, 34, 35].

Marijuana is regulated as a schedule 1 substance by the US federal government. However, 36 states, the District of Columbia, Guam, Puerto Rico, and the U.S. Virgin Islands have comprehensive medical cannabis programs. In addition, the remaining 14 states allow use of cannabidiol (CBD) with minimal or no delta-9-tetrahydrocannabinol (THC) for medical reasons in limited situations [36]. Among these, 12 states approved cannabis for the treatment of ASD and self-injurious or aggressive autistic behavior [37]. Most of European countries have legalized medical cannabis in recent years. The European Union recently legalized CBD and hemp products. However, some countries legalized only derivatives of the cannabis plant and not flowers or other natural forms of the plant [38].

The medicinal use of cannabis in ancient China dates back to about 2700 BC [39]. According to the US drug enforcement administration (DEA), hemp and cannabis comprise separate parts of the cannabis plant [40]. Three species of cannabis including cannabis sativa, cannabis indica, and cannabis ruderalis have been identified [41, 42, 43]. Cannabis contains more than 100 cannabinoids; CBD and THC are the focus of most studies [44, 45]. THC is the main psychoactive constituent and is a partial agonist of CB1 and CB2 receptors. THC can produce neuroprotective, analgesic, appetite stimulant, antiemetic, and antiglaucoma effects [46, 47]. In contrast to THC, CBD has little affinity for CB1 and CB2 receptors but functions as an indirect antagonist of cannabinoid agonists. CBD is also an inverse agonist of CB2 receptors. CBD can counteract some of the functional consequences of CB1 activation in the brain and exhibits anti-inflammatory, antioxidant, anticonvulsant, and neuroprotective effects and reduces THC psychoactivity [45, 48, 49]. Other cannabinoids including non-psychoactive cannabidivarin (CBDV) also contribute to the cannabis medicinal effects. Studies in animal models and humans advocated for anti-inflammatory, neuroprotective, anxiolytic, and antipsychotic properties of cannabis compounds.

Here we present a comprehensive review on: (i) Changes in the endocannabinoid system in autism (ii) Effect of cannabis on autism (iii) Effect of cannabis on autism-associated disorders.

2. Changes in the endocannabinoid system in autism

Therapy for autism has been difficult to establish because it is a multifactorial disorder. ASD manifests due to a combination of genetic, immunological, and environmental factors that result in communication and behavioral problems [50, 51, 52]. Many studies have shown that the ECS plays a crucial role in regulating emotional and social behavior [53, 54, 55, 56, 57]. Dysfunction in the ECS was connected with the pathology of neurodevelopmental disorders, most specifically ASD [24, 58]. Pathophysiological mechanisms producing the neurobehavioral deficits in ASD include aberrant synaptic plasticity, immune dysfunction, and metabolic disturbances, all of which are regulated by ECS [14, 35].

The ECS represents a complex system of lipid signaling pathways [59, 60]. The ECS plays an important role in the development of the central nervous system (CNS) [61, 62, 63]. CB1 receptors are located in the central nervous system, peripheral nervous system, and peripheral organs. In the CNS, CB1 receptors are concentrated in the cerebellum, hippocampus, and the basal ganglia, which are areas of dysfunction in autism [64, 65, 66]. CB1 receptor activation results in glutamate release, inhibition of synaptic transmission and regulation of synaptogenesis, axonal outgrowth, differentiation, migration, and proliferation [61, 67, 68, 69].

Autism is also associated with dysregulation of the immune system [15, 70, 71]. CB2 receptors are located in immune cells and may control the movement of inflammatory cells [64, 72, 73, 74, 75, 76]. CB2 receptors are also expressed in microglia and astrocytes, which may be critical to ASD-related neuroinflammation [77]. Immune dysfunction, increased autoimmune activity and inflammatory responses are associated with microglial activation in patients with ASD [78]. Microglia activation, increased levels of inflammatory cytokines and chemokines, and augmented expression of the microglial activation-related genes are the results of the pro-inflammatory status of the immune system in autism [78, 79, 80, 81]. Moreover, the mRNA and protein for CB2 receptor and endocannabinoid enzymes were significantly changed in animal models of autism, demonstrating the involvement of the ECS in ASD-associated immunological disturbances [52, 70]. Studies focused on children with autism have shown alterations of the immune system such as variation in monocyte and macrophage reactions, abnormal T helper cytokine levels, reduced numbers of lymphocytes, and abnormal immunoglobulin levels [82, 83, 84, 85, 86, 87]. In addition, an increase in pro-inflammatory cytokines was associated with more regressive forms of autism and more pronounced stereotypical behaviors [78].

Several investigations revealed the involvement of the ECS in neuromodulation as well as in the regulation of emotional responses, behavioral reactivity, and social interaction in ASD [19, 24, 88]. Disruption of this system may impair social communication, social play, and reciprocity [19]. Some studies investigated the association of acetaminophen with social behaviors and ASD confirmed activation and involvement of the ECS in autism [89, 90, 91, 92, 93, 94, 95, 96].

Additional studies have established the connection between the endocannabinoid system and autism. Lowered CB1 receptor level was found in postmortem brains of autistic patients [78]. Stimulation of the CB1 receptors either directly by the synthetic cannabinoid receptor agonist WIN55212-2, or indirectly by a 2-AG hydrolysis inhibitor, increases the spatial memory performance of rats under stress conditions [97]. Polymorphisms in the gene encoding the CB1 receptor, CNR1, were found to modulate striatal responses and gaze duration to social reward cues [98, 99], indicating that changes in endocannabinoid affinity to the CB1 receptors may lead to deficits in social rewards observed in autism. Additionally, the CB1 receptor has neuroprotective capacities manifested by decreasing tumor necrosis factor (TNF)-α levels in neurodegenerative conditions [100].

Animal studies implied that social play behavior boosts AEA levels in several brain regions [55, 56, 101]. High AEA concentrations, following inhibition of FAAH, result in CB1 receptor activation and improved social play behavior [89, 102, 103]. Plasma concentrations of AEA were found to be lower in children with ASD compared to healthy controls [18, 104]. In addition, FAAH inhibition boosts levels of N-acylethanolamines such as oleoylethanolamide (OEA) and palmitoylethanolamide (PEA) in hippocampus. High OEA and PEA levels can compete with AEA for FAAH enzyme and lead to reduced AEA metabolism and enhanced activity of the CB1 receptors [101]. Moreover, as neither OEA nor PEA have affinity for CB1 receptors, it is possible that competition for FAAH forces AEA activity back to the CB1 receptors [19]. This suggests that a deficiency in social play behaviors may be caused by low AEA levels in critical brain areas. However, earlier studies have demonstrated that broad excitation of CB1 receptors interferes with the normal excitation of complex social acts, possibly by interfering with cognitive functions required for normal social interactions [101, 103]. Additionally, autism mouse models demonstrate a downregulation of G protein-coupled receptor 55, GPR55 and peroxisome proliferator-activated receptor, PPAR, which can be alternative receptors involved in social play behaviors [19]. Some of the behavioral changes may be mediated by AEA activation of other receptor targets or direct activation of PPARγs by OEA or PEA, since data indicate that activation of hippocampal PPARγ enhances cognitive performance [105]. PEA with its intestinal anti-inflammatory characteristics is a point of interest for autism since part of the autistic chronic inflammatory state is mediated via the gastrointestinal associated immune system [83, 106, 107]. The observed anti-inflammatory effects of PEA are exerted through activation of CB2, GPR55 and PPARγ receptors [108].

The endocannabinoids through CB1 receptors can reduce glutamate release, alter synaptic plasticity and, therefore, modulate neurotransmission in the basal ganglia. Cannabinoid signaling can release dopamine that counteracts the effects of induction of the CB1 receptor via dopamine D1-like receptors [109]. On the other hand, dopamine signaling via dopamine D2-like receptors may lead to upregulation of endocannabinoid signaling [109]. Endocannabinoid signaling also functions as a retrograde signaling system in GABAergic and glutamatergic synapses [110, 111].

Different genetic animal models are used to explain mechanisms of action of the endocannabinoids. The BTBR T+tf/J (BTBR) mouse strain have autism-like phenotype. CB1 density in the BTBR hippocampus is 15–20% higher than in other strains. GTPγS- stimulated binding of CB1 agonist cannabinoid CP55940 to Gi/o-coupled receptors in the BTBR animals is also elevated, indicating a potential for increased sensitivity [112, 113]. In BTBR model increased AEA action at CB1 receptors improved social impairment and reduced locomotor movement, implying an influence on irritability and repetitive behaviors [114]. Furthermore, the treatment of BTBR mice with the FAAH inhibitor, URB597, and the cannabinoid delta 9-tetrahydrocannabinol (Δ9-THC) ameliorated the social behavior deficits of BTBR mice [25, 114, 115]. There is also evidence for elevated CB2 receptor expression in the BTBR mouse brain [116]. Genomic studies revealed an upregulation of mRNA levels of the CB2A isoform, in the cerebellum of BTBR mice [115, 116]. A clinical study in young children demonstrated upregulation of CB2 gene expression in peripheral blood mononuclear cells of patients with autism [117]. The increase in CB2 expression may serve a compensatory role for the inflammation associated with autism [118, 119, 120, 121]. Therefore, keeping in mind that AEA suppresses the release of proinflammatory cytokines through a CB2-mediated mechanism [77], the enhancement of CB2 may be negative feedback to reduce the proinflammatory responses in ASD. Moreover, in the Fragile X syndrome (FXS) model of autism mRNA levels of the enzymes (DAGL and MAGL), were altered in the cerebellum and hippocampus, whereas levels of 2-AG in the same regions were not changed [55, 122]. In the FXC model, treatment with URB597 (selective inhibitor of the FAAH) resulted in an increased AEA activity, improved memory and anxiety-like behavior, and reversed the social impairment [81].

Studies on Shank3B-/- mouse with significant social interaction disorder, have shown that pharmacological augmentation of 2-AG levels by ZL184 (monoacylglycerol lipase inhibitor) normalized social interaction deficits [123]. Mutations in neuroligin-3 gene, 4 (NLGN 3, 4), which is involved in the formation and remodeling of CNS synapses, are associated with intellectual disability, seizures, and autism behaviors [124, 125]. Studies in mice with a neuroligin-3 amino acid substitution (R451C) and a neuroligin-3 deletion revealed that neuroligin-3 is specifically required for tonic endocannabinoid signaling, further confirming variations in endocannabinoid signaling can donate to the pathophysiologic mechanism of autism [23, 126]. Additionally, it was shown that a synthetic cannabinoid WIN55212-2 may reduce aggressive behavior of neuroligin-3 (NL3) R451C mouse model of ASD by modulation of CB1 receptor [127]. Another animal model with rats prenatally exposed to valproic acid (VPA), exhibits ASD-like abnormalities in sociability and nociception tests, and alterations of distinct elements of endocannabinoid system [19, 128]. VPA rats have lessened level of mRNA of PPARα and GPR55 in hippocampus and frontal cortex, decreased level of FAAH and abnormal AEA activity, favoring ASD like behavioral symptoms [19, 128]. It was reported that the FAAH inhibitor PF3845 enhanced AEA signaling and weakened the discrepancy in social behavior in VPA rats [19]. Another inhibitor of FAAH enzyme, URB597 improved social deficits, repetitive behaviors, and abnormal emotion-related behaviors in VPA-exposed offspring [78]. The results were produced by the deletion of postsynaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) subunits GluA1 and GluA2 and helped to characterize the mechanism of AEA signaling in the prefrontal cortex [78]. Moreover, increased 2-AG concentrations developed the improvement of behavior defects in VPA rat model after treatment with JZL184 [18].

Recent testimony clearly links alterations in the ECS with autism. The lowered endocannabinoid levels, degradation of enzymes and CB receptors up-regulation indicate reduced endocannabinoid signaling in ASD. Taken together, the findings lead to the hypothesis that influencing the ECS can normalize different behavioral patterns compromised in ASD and recommend the ECS as a novel target option for autism pharmacotherapy.

3. Cannabis in the treatment of autism
3.1 Preclinical studies

Cannabidiol is effective in the treatment of some neurodevelopmental conditions including ASD. CBD has low affinity for the CB1 and CB2 receptors, but m bind to these receptors. CBD has high affinity for the TRPV1 and the TRPV2 receptors involved in stimulation of the CB1 receptors [61, 78, 129]. In animals cannabidiol increases the serum levels of AEA by inhibiting the FAAH enzyme and normalizes the depletion of AEA tone and, as a result, improves the ASD symptoms [130, 131]. In humans CBD’s effect on AEA serum levels may be due to inhibition of the metabolism [132]. Since decreased AEA level produces social impairment in genetic models of ASD, intensifying AEA signaling may inverse the behavioral disorders. The administration of CBD to C57BL/6J mice (model with compulsive behavior) alleviated marble-burying behavior, which is analogous to repetitive and compulsive behaviors observed in ASD [78]. There is also preclinical evidence supporting that CBD improved autism-like social behavior in mice with Dravet syndrome and the social and cognitive dysfunctions in rat model of schizophrenia [133, 134]. In murine models CBD modulated immune function by reducing activation of microglia and decreasing expression of chemokine ligands and interleukins [129]. In another study, cannabidiol reduced neuronal excitability and transmission through inflammatory pathways by inhibition of adenosine reuptake and modulation of the release of pro-inflammatory cytokine tumor necrosis factor alpha (TNFα) [135]. In addition, chronic CBD administration rescued several autistic-like behaviors (anxiety- and depression-like behavior, poor social interaction, and increased rearing behavior, as well as reference memory and working memory) in Scn1a+/- mice (Dravet syndrome animal model) [133, 136]. CBD did not induce any adverse effects on motor function, giving further support for the benefits and safety of using this cannabinoid as ASD therapy.

THC also can produce a positive effect on the neurodevelopmental conditions such as autism. THC treatment increased locomotor behavior and reduced the depressogenic profile in BTBR mice with an autism-like phenotype [115].

Another widely studied nonpsychotropic phytocannabinoid is cannabidivarin (CBDV). Although the mechanism of actions of CBDV is still unclear, it was reported that CBDV might produce its effects through voltage dependent anion selective channel protein 1 (VDAC1), or through the activation and desensitization of TRPV1 channels [135]. CBDV reduces neuronal excitability and neuronal transmission and may also inhibit adenosine reuptake or modulate the release of pro-inflammatory cytokine tumor necrosis factor alpha (TNFα) [137]. CBDV as TRPV1-antagonist with anti-inflammatory activities, decreases inflammation involving cytokine production [138, 139, 140]. A therapeutic potential of CBDV was supported by the certain effects of the cannabinoid in Mecp2 seizure animal model (Rett syndrome model) and VPA model [128, 141, 142, 143]. CBDV increased the AEA and OEA levels, reduced DAGL-a expression and reduced CB1 and CB2 receptor levels in the Mecp2 mice [144, 145]. As a result, CBDV restored the compromised general health status, the behavioral deficit, the sociability, and the brain weight and produced a rescue of memory deficits in this animal model [144, 145]. Surprisingly, CBDV restored neurotrophic factor levels and ribosomal protein six phosphorylation in the mice, though both were expected to be impaired in ASD [145]. In a study with VPA rats CBDV repaired hippocampal endocannabinoid signaling and neuroinflammation [146]. This cannabinoid produced an increase in CB1 receptor, FAAH and MAGL levels, enhanced GFAP, CD11b, and TNFα levels and caused microglia activation in the hippocampus [146]. As a result, social impairments, short-term memory deficits, repetitive behaviors and hyperlocomotion were restored [146]. On the other hand, chronic administration of CBDV induced an increase in glial fibrillary acidic protein (GFAP, associated with CNS inflammation) in control and VPA animals [146]. Overall, the ASD-like behavioral changes were repaired by CBDV treatment suggesting that the beneficial effects of CBDV could be related to the restoration of the ECS abnormalities in the hippocampus. The correlation between improvement of the behavioral deficits and modulation of the ECS was consistent with other studies [78, 104, 127, 128].

These current data strongly link alterations of the ECS with ASD and provide evidence supporting the ability of cannabis or/and certain cannabinoids to improve anomalies similar to core and associated symptoms of ASD. However, the exact mechanisms of action of cannabinoids in ASD patients remains unclear [73, 76]. The non-endocannabinoid mechanisms may involve the regulation of glutamatergic and GABAergic neurotransmission and receptors including the GPR55, 5-HT1, α3 and α1 glycine receptors, and TPPA1 channel [56, 108, 110, 111, 133]. Moreover, CBD may also indirectly act through neuropeptides such as oxytocin and vasopressin that are involved in social bonding, compromised in ASD [54, 81].

3.2 Clinical studies

Cannabis has an extensive range of clinical applications including treatment of multiple sclerosis, Tourette syndrome, Parkinson’s disease, epilepsy, glaucoma, nausea, depression and pain [37, 59, 60, 147, 148, 149, 150, 151]. Epidiolex® (CBD) has been FDA- and EMA-approved for two epilepsy syndromes related to ASD: Dravet and Lennox-Gastaut Syndrome [149, 150]. Autism is associated with disruption of the endocannabinoid system [19, 88, 152]. Cannabis and some cannabinoids have ability to modulate the ECS and, therefore, improve behavioral deficits in autism.

Use of cannabinoids for autism has a growing interest in social media. A number of anecdotal self-reported cases show that ASD children who failed traditional pharmacologic therapy have responded to cannabis treatment. Parents of these children have reported remarkable improvements. A child with autism has spoken first words after receiving cannabis oil and finally, developed significant language skills [153]. In another case, FDA-approved medications produced life-threatening toxicities in 10-year-old boy with autism. He was subsequently started on cannabis; six years after the six-months-terminal diagnosis, the boy was sociable and successful [153]. Similarly, other boys with severe ASD, treated with various therapies did not result in alleviating autistic symptoms. The boys showed dramatic improvement in communication skills and interactions after they were given cannabis [154, 155, 156]. Smoking cannabis improved the sociability, vocabulary and reduced anxiety of a 20-year-old ASD man [157]. Another boy with a brain tumor, autism, severe seizures, and self-destructive behavior was treated with cannabis and showed remarkable progress [158].

Despite the growing interest, there are very limited clinical data on the impact of cannabis on autism. In a prospective single-case-study dronabinol (THC) produced improvement in hyperactivity, irritability, stereotyped behaviors, and speech in an ASD boy [159]. In another open label study dronabinol also produced significant improvements in self-injurious behavior of seven out of 10 mentally retarded adolescents [160].

Positive results in autistic patients treated with cannabis as well as increasing anecdotal reports of cannabis beneficial effects on autistic children have led to more scientific testing. In January 2017, Shaare Zedek Medical Center initiated a phase II clinical trial (NCT02956226) to evaluate the safety and efficacy of cannabis (CBD: THC, 20:1 ratio) in children with autism [161]. This three-month study involved 150 patients (aged 5–21 years) with mild to severe autism. The investigators informed cannabinoid therapy was correlated with reduction in disrupting behavior, the CGI-I (Clinical Global Impression-Improvement) scale improved in 49% patients against 21% control patients. Median SRS (Social Responsiveness Scale) score increased 14.9 points versus 3.6 points on placebo. The therapy produced a decrease in body weight in obese patients. This is particularly important as antipsychotics are linked to substantial weight gain. No significant adverse incidents were reported [162]. Common adverse events included somnolence and decreased appetite, reported for 28% and 25% on whole-plant extract, respectively (n = 95); 23% and 21% on pure-cannabinoids (n = 93), and 8% and 15% on placebo (n = 94). Even the study demonstrated that medical cannabis has the ability to improve ASD disrupting behaviors with adequate acceptability, evidence for efficacy of cannabinoids is insufficient. Further testing was recommended.

More clinical trials were conducted recently. A retrospective study assessed effect of cannabis (CBD:THC, 20:1 ratio) in 60 ASD children with severe behavioral problems [104]. This therapy resulted in improved behavioral outbreaks in 61% patients. Anxiety, communications as well as disruptive behaviors were also improved in 39%, 47%, and 29%, respectively. The Autism Parenting Stress Index reported 33% less stress [104]. Adverse events were sleep disturbances, irritability, and loss of appetite. One patient with a higher THC dose had a brief psychotic event [104]. In an open-label prospective study, 188 ASD patients received cannabis oil with 30% CBD and 1.5% THC (20:1). After six months of treatment, 60.0% of the patients were evaluated. Substantial progress was reported in 30.1%, moderate in 53.7%, minor or no improvement in 6.4% and 8.6% of the patients, respectively. The most common side effect was restlessness [163]. The same CBD:THC ratio (20:1) was used in a prospective study with 53 autistic patients [164]. It was reported that self-injury and rage attacks improved in 67.6% and worsened in 8.8%; hyperactivity better in 68.4% and deteriorated in 2.6%; sleep problems improved in 71.4% and aggravated in 4.7% patients. Adverse effects, sleepiness and alteration of appetite were weak [164]. In a double-blind study with 34 men (half with ASD) CBD amplified fractional amplitude of low-frequency fluctuations (fALFF) in the cerebellar vermis and the right fusiform gyrus and modified vermal functional connectivity in ASD group only [165].

Recently an observational trial (CT03699527) with 200 children with ASD was completed. Goal of this investigation was to assess disposition, pharmacokinetics and pharmacodynamics of medical cannabis products including CBD and to provide evidenced based dosing guidance for these products to the children [78]. Another open clinical trial (NCT03900923) for CBD in youth with ASD (n = 30) was recently announced [166]. Aims of the investigation are to detect the optimal CBD doses and to characterize outcomes for upcoming studies. Another placebo-controlled trial (NCT03537950) examines brain reaction to a single dose of CBD and CBDV in men (n = 38) with and with no autism [167]. A research program at Montefiore Medical Center, Albert Einstein College of Medicine is conducting a 12-week Phase 2 double-blind, randomized, placebo-controlled trial (NCT03202303) with CBDV in 100 children and adolescents with autism [168]. An open-label clinical trial (NCT03849456) investigating CBDV effect in ASD patients was terminated due to enrollment challenges during COVID-19 pandemic [81].

In addition, recent case series studied the impact of cannabis on Fragile X patients. It was reported reductions in social evasion and anxiety, improvements in sleep, motor and language skills after the CBD treatment. Two out of three patients exhibited anxiety reappearance following termination of the therapy. CBD was shown to be a medicinal candidate for FXS patients. However, rigorous clinical trials are required to support this finding [169].

Based on available data cannabis was found as safe and effective choice to alleviate ASD signs. But cannabis produces not only therapeutic effects but also creates dangerous health complications. One major concern is the impact of cannabis on brain. Cannabis impairs normal brain development and may result in potentially non-reversible neurocognitive changes [170]. Early cannabis use was correlated with a significant drop in IQ at age 38 years [171]. Cannabis use has been shown to impair cognitive functions from basic, such as motor coordination to more complex executive functions [172]. Chronic cannabis was associated with cognitive problems such as addiction, distorted perceptions, difficulty in thinking and problem solving, working memory deficits, and abnormal social behavior [152, 171, 172, 173, 174, 175]. These deficits vary in severity and depend on the quantity, recency, age of onset and duration of cannabis use. Moreover, individuals with cannabis-related impairments have been found to have trouble for successful recovery, putting them at increased risk for relapse to cannabis use. Further, termination of cannabis use did not fully restore neuropsychological functioning among adolescent-onset cannabis users [173]. Cannabis use is also specified as a potential source, aggravator, or masker of psychiatric symptoms particularly in young people [176, 177, 178, 179]. It is difficult to recognize patients who benefit and who build side effects [180]. For instance, while cannabis might be beneficial in persons with one phenotype, it may have no effect or severe adverse outcomes in persons with other phenotypes [181]. Therefore, all positive and negative effects of medical cannabis have to be carefully assessed in large studies.

4. Cannabis in the treatment of autism-associated disorders

An exciting hypothesis about the medicinal potential of cannabis is that cannabis/cannabinoids may improve disorders co-occurring with ASD, including ADHD, seizures, anxiety, mood disorders, and sleep disturbances.

4.1 Attention deficit hyperactivity disorder

Attention-deficit hyperactivity disorder (ADHD) and ASD are frequently comorbid, have similar genetics and cognitive deficits [182, 183]. ADHD is neurodevelopmental illness characterized by inattention, hyperactivity, and impulsivity due to lack of neurotransmitter dopamine in the brain [184]. Treatments for ADHD involve stimulant medications, but they may have unpleasant side outcomes. Some people utilize cannabis as ADHD therapy as cannabis restores brain dopamine levels without any adverse effects [185].

Moreover, clinical and anecdotal data recommend cannabis as a therapy for ADHD. In a study of 268 separate online discussion threads, 25% of people thought that cannabis was helpful [186]. However, efficiency of cannabis was not entirely established [187]. Another controlled clinical trial investigated effect of Sativex (THC:CBD, 1:1 ratio) on 30 ADHD patients. The study showed improvements in hyperactivity/impulsivity, inattention, and emotional lability [188]. Smoking cannabis improved the driving skills of a cannabis-user with ADHD during a time of abstinence [189]. Dronabinol produced improvement in self-injurious behaviors in adolescents [190]. High cannabis doses were related to ADHD medication reduction and a lower ADHD self-report scale score [191]. However, some research studies demonstrated that cannabis does not have impact on cognitive skills, or perhaps make the skills worse [192]. Moreover, the ability of cannabinoids to cross the placenta and affect fetal neurodevelopment may produce hyperactivity, impulsivity, and inattention symptoms in childhood [193]. Further research is needed to estimate usefulness of cannabis in the management of ADHD.

4.2 Seizure disorders

Epilepsy is one of the associated disorders in autism with an occurrence rate of 5–40% and with the highest incidence reported in adolescents and young adults [194, 195, 196, 197, 198, 199]. Most studies showed increased risk of epilepsy in ASD patients, and there is also evidence indicating higher risk of ASD in individuals with epilepsy [199, 200]. Careful selection of antiepileptic drugs and close monitoring of adverse effects is essential due to overlapping co-morbidities [201, 202].

Cannabis was used for the treatment of epilepsy for centuries [203, 204, 205]. Cannabis, cannabinoids, and endocannabinoids have been extensively studied and found to have antiseizure activities [206, 207, 208]. In animal seizure models, AEA, THC, and WIN55212-2 exhibited potent anticonvulsant effects through CB1 activation [209, 210]. But the CBD antiseizure effect was mediated by several mechanisms such as ENT transporter, GPR55, TRPV1, 5-HT1 and glycine receptors [207, 211, 212].

In open label study in patients with Lennox-Gastaut syndrome and drug-resistant seizures, CBD given as add-on therapy reduced seizure frequency in these highly treatment-resistant patients [213, 214]. CBD also significantly reduced convulsive-seizure frequency in 120 young patients with the Dravet syndrome and drug-resistant seizures [213].

Epidiolex was the first FDA approved compound in cannabis for therapy of three treatment-resistant cases of seizures: Lennox-Gaustat Syndrome, Dravet Syndrome and Tuberous Sclerosis Complex, in patients 1 year of age and older [214]. This approval ensures the quality and effectiveness of CBD in seizure disorders.

4.3 Anxiety

Up to 40% of children with autism have at least one diagnosed anxiety syndrome [215, 216, 217]. The current anxiety treatment is cognitive-behavioral therapy that may not work for ASD children and anti-anxiety medications. Patients build a quick tolerance to these medications that causes thousands of overdose-associated deaths every year [218].

The effect of cannabis on anxiety is complex. THC and CBD produce opposing impacts on brain activity. THC is a psychoactive stimulant whereas CBD calms psychoactivity down [217]. THC relaxing effect is short-term and might develop memory deficiency and cognitive damage [219]. Moreover, THC has been associated with development of psychosis and increased anxiety [220, 221]. In contrast, CBD inhibits the anxiogenic and psychotogenic effects provoked by THC and produces anxiolytic effect [222, 223]. CBD has a pharmacological profile like atypical antipsychotic drugs and inhibits the FAAH enzyme increasing the levels of AEA [99, 224, 225, 226]. High level of AEA was linked to reduced stress, anxiety, and depression. Other mechanisms of canabidiol anxiolytic effect may be activation of metabotropic receptors for serotonin or adenosine and interaction with TRPV1, GABAA and PPAR receptors [226, 227, 228, 229, 230]. CBD has demonstrated efficacy in animal models of anxiety and stress [231, 232, 233, 234]. Preclinical data strongly suggest that CBD has a potential as therapy for generalized anxiety disorder, panic disorder, social anxiety disorder, obsessive–compulsive disorder, and post-traumatic stress disorder as well as prevention of the long-term anxiogenic effects of stress [235, 236, 237].

Clinical experimental data support preclinical findings. Cannabidiol pretreatment drastically diminished anxiety, cognitive and speech defects and improved forgetting traumatic memories in patients with social anxiety disorder [238, 239, 240, 241, 242]. Brain scans displayed blood flow changes in limbic and paralimbic brain zones as an evidence of anti-anxiety effect of the cannabinoid [242]. A large retrospective case series were conducted in 72 adults with CBD as add-on therapy. Anxiety scores decreased in 79.2%, 15.3% patients experienced worsening anxiety symptoms [243]. Other studies also reported CBD was efficient in reduction anxiety signs in different patient populations [244, 245, 246]. However, additional studies are required to verify long-term effectiveness and safety and establish appropriate dosing.

4.4 Sleep disorders

Sleep difficulties are the very frequent ASD-related syndromes influencing up to 80% of autistic children [247, 248, 249, 250]. Nonpharmacological treatments for sleep problems in ASD children include general behavioral treatments, sleep hygiene and parent training [251, 252]. Pharmacological therapy of autistic sleep disorders includes hypnotic medications, which cause significant adverse effects such as addiction, serious withdrawal, and complex sleep-related behaviors [253]. Melatonin may also cause sleep conditions. Individuals with ASD have abnormal melatonin metabolism, low melatonin levels and abnormalities in genes associated with melatonin production [35, 254, 255]. Use of melatonin in ASD has resulted in improved sleep parameters with minimal adverse effects [34, 35, 256, 257].

Cannabis was studied for treatment of sleep disorders and was shown to have low potency sedative effect in mice [258]. In a large retrospective study with 166 patients, 79% reported improved sleep quality and considerable reduction of sleep time following cannabis use [259]. Cannabis components, THC and CBD cause different effects on sleep. Earlier studies have shown that THC produced a somnolent effect in humans [260, 261, 262, 263]. However, in recent studies THC produced no effect on nocturnal sleep, reduced sleep expectancy as well as increased daytime sleep [264]. THC also significantly decreased duration of nighttime sleep, suggesting development of tolerance to the sedative effect [265]. CBD appeared to counteract the activity of THC by activating neurons in awaken-inducing brain zones including lateral hypothalamus and/or dorsal nuclei and increasing dopamine extracellular levels [264, 266, 267, 268]. The CBD awakening properties were not inhibited by the sleep-inducing AEA [267, 269]. CBD increased wakefulness during light-on period, increased sleep lights-off period and prevented sleep rebound after total sleep deficiency [265, 266, 267, 268, 270]. In a trial with 72 adults CBD upgraded sleep scores in 66.7% patients [243]. Oral administration of cannabidiol to ASD children has also shown improvement in ASD-associated sleep disorders [164]. CBD appears to optimize sleep and improve ASD-associated sleep problems and may have a beneficial value in autistic sleep conditions [271, 272, 273, 274].

4.5 Mood disorders

A recent study discovered mood illnesses are prevalent in ASD patients. Children with ASD and ADHD were 2.7 times more expected to have mood ailments, compared with autistic patients without ADHD [275]. Autistic children can be mistakenly diagnosed as autism and bipolar disorder have some common symptoms. In many cases “mania” symptoms are also signs of autism [276]. ASD children are four-fold more likely to suffer depression [277]. Therapy of co-occurring autism and mood conditions is complex and standard pharmacotherapy may be ineffective.

The ECS has essential involvement in mood disorders. The system activity of the system may be modified by cannabinoids [278]. Both THC and CBD have potential to lessen symptoms of mood disorders. CBD produces antidepressant effects and demonstrates antipsychotic properties in depression, anxiety, and bipolar disorders [279, 280, 281, 282, 283, 284]. THC in combination with CBD confirmed the antipsychotic effect [285]. However, results on effect of cannabis whole plant on mood conditions are confusing. In line with some studies that reported anxiety and increase symptoms of depression, many cannabis users describe an improvement in mood [286]. While the relationship between cannabis use and psychiatric illnesses was recorded [287, 288], longitudinal research assessing the connection of cannabis with psychiatric conditions produced varied outcomes [289]. A number of studies correlated cannabis with increased risk of depression, anxiety, bipolar disorder, substance use disorders and psychosis [290, 291, 292, 293, 294, 295, 296], whereas others did not agree with this conclusion [295, 296, 297]. Understanding long-term consequences of cannabis use is especially important in managing pediatric conditions. Studies showed children using cannabis frequently have greater chance of depression, anxiety, schizophrenia, or bipolar disorder in later life [298, 299, 300, 301]. However, not every cannabis user builds psychotic disease [188, 193, 302]. People with family history of a psychotic disease, or with schizotypal character, or with specific genes, might have the risk of psychotic disorder as a result of consistent cannabis use [303].

Not many studies advocate for the use of cannabis for psychiatric disorders [304]. The benefits have to be balanced against the significantly better recognized risks and the adverse consequences of cannabis for young people [305, 306, 307]. Further investigation should be conducted in patients with autism and mood disorders [283, 308].

5. Cannabis genomic and biotechnology advancements

Application of genomic technologies including high-throughput sequencing has enabled significant advancements in the characterization of different plant species. Fundamental knowledge of plant gene content and genomic variation have been elucidated using biotechnology approaches. For most crops, the biotechnology tools are well developed. However, until recently the tools were under-developed for cannabis because of the prolonged ban of recreational cannabis as well as the strict regulation of hemp. Progress in cannabis has been made recently in the heterologous expression of cannabis genes, as well as in the study of the function of representative genes in model biological systems.

The heterologous expression of cannabinoids in yeast has been recently reported [309]. Key advantages of the yeast expression system for cannabinoid synthesis include ease of extraction and purification, less potential for heavy metal contamination and customization of cannabiboids. The full biogenesis of the main cannabinoids was generated in Saccharomyces cerevisiae. Study demonstrated the potential for a platform to produce cannabinoids, allowing for rigorous study of the compounds. Ultimately, the platform might be utilized in the development of therapies for a range of human health complications.

To characterize molecular differences between cannabis and hemp, the sequences of the respective plant genomes were analyzed [310]. Plant genomes frequently contain duplicated genes; gene amplification is a proven mechanism for increasing expression levels [311]. Overall, the sequence comparison demonstrated that there were few differences in gene median read depth (MRD) between a cannabis strain Purple Kush (PK) and the hemp cultivar Finola [310]. One exception is AAE3, a gene encoding an unknown function in PK. It is postulated that AAE3 may have a role in cannabinoid biosynthesis [311]. It is believed that the large expansion of AAE3 occurred through the insertion of pseudogenes into the PK genome. The differences in expression of the cannabinoid enzymes between PK and Finola are attributed to genetic variations that result in changes in gene expression. Recently, whole genome and transcriptome mapping of cannabis has been obtained using next generation sequencing (NGS) methods [312]. The information may be combined with proteomics and metabolomics to detect secondary cannabis metabolites. More significantly, NGS data provides the foundation for introducing genetic engineering in cannabis [313].

Because of the wide use of Cannabis as drugs, the effects of Cannabis sativa gene products are under investigation. The challenge is the number of different chemical components present in crude extracts. CBD has been studied in rodent models, but its effects remain incomplete. Evaluation of CBD in zebrafish showed that adult wild-type zebrafish altered behavior when exposed to 40 mg/mL of cannabidiol [314]. The locomotory ability of the zebrafish was influenced by the CBD, in contrast to the control of zebrafish exposed to water alone [314]. The CBD treated zebrafish exhibited a significant decrease of swimming performance, both in velocity as well as distance. Gene expression studies demonstrated differential gene expression, upregulation of some genes (il 1b and il17a/f2) and correspondingly down-regulation of other genes (tgfba, s100a10b, ighm and cd4-1).

While biotechnology of cannabis is still early, the availability of affordable next generation sequencing platforms together with heterologous expression systems and model biological systems, will accelerate the progress in the characterization of the cannabis genome and potential medicinal candidates.

6. Summary

Autism spectrum disorders are developmental disabilities producing substantial social, communication and behavioral disorders. No medication was created for the therapy of the main ASD symptoms. Pharmacological therapy attempts to address ASD-associated comorbidities. Many of the drugs used are off-label.

Due to its vital role in regulating emotion and social behaviors, the endocannabinoid system has been investigated for its association with ASD. Studies indicate contribution of endocannabinoid system dysfunction to ASD pathogenesis and suggest the ECS disfunction contribute to the behavioral deficits and neuroinflammation observed in autism. Cannabis and cannabinoids interact with the ECS and may improve ASD-associated social and cognitive impairments. Therefore, the ECS represents a possible goal for the development of ASD treatment and cannabis/cannabinoids may be effective as pharmacological therapy.

Although, clinical studies have shown promising results of cannabis treatment in ASD and associated disorders, there are limited data supporting clear effect of cannabis/cannabinoids in different phenotypes of ASD. More clinical investigations are needed to discover the efficacy, safety, and dosing of the therapy. This would be a significant advance in the treatment of autism and could lead to improved functioning and quality of life for the patients and their families.

Abbreviations

ASD, autism spectrum disorders; CBD, cannabidiol; THC, delta-9-tetrahydrocannabinol; CBDV, Cannabidivarin; ECS, endocannabinoid system; EC, endocannabinoid; CB1, cannabinoid receptor 1; CB2, cannabinoid receptor 2; AEA, anandamide; 2-AG, 2-arachidonoylglycerol; FAAH, fatty acid amide hydrolase; MAGL, monoacylglycerol lipase.

Author contributions

MB wrote chapters—Abstract, Cannabis in the treatment of autism, and Cannabis in the treatment of autism-associated disorders. HA wrote chapters—Introduction and updated Attention deficit hyperactivity disorder and Sleep disorder. PB wrote chapter—Changes in the endocannabinoid system in autism. ZL wrote—Cannabis genomic and biotechnology advancements and updated Changes in the endocannabinoid system in autism.

Ethics approval and consent to participate

Not applicable.

Acknowledgment

Thanks to all the peer reviewers for their opinions and suggestions.

Funding

This research received no external funding.

Conflict of interest

The authors declare no conflict of interest.

References
[1]
Center for Disease Control and Prevention (CDC). 2021 Community Report. What is Autism Spectrum Disorder? — CDC. Spotlight On: Racial and Ethnic Differences in Children Identified with Autism Spectrum Disorder (ASD). 2021. https://www.cdc.gov/ncbddd/autism/addm-community-report/spotlight-on-closing-racial-gaps.html (Accessed: 17 November 2021).
[2]
Christensen DL, Baio J, Van Naarden Braun K, Bilder D, Charles J, Constantino JN, et al. Centers for Disease Control and Prevention (CDC). Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years–Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2012. The Morbidity and Mortality Weekly Report (MMWR) Surveillance Summaries. 2016; 65: 1–23.
[3]
WHO. Media centre Autism spectrum disorders. 2021. Avaliable at: http://www.who.int/mediacentre/factsheets/autism-spectrum-disorders/en/ (Accessed: 23 March 2021).
[4]
Ghosh A, Michalon A, Lindemann L, Fontoura P, Santarelli L. Drug discovery for autism spectrum disorder: challenges and opportunities. Nature Reviews Drug Discovery. 2013; 12: 777–790.
[5]
Betancur C. Etiological heterogeneity in autism spectrum disorders: more than 100 genetic and genomic disorders and still counting. Brain Research. 2011; 1380: 42–77.
[6]
Rosti RO, Sadek AA, Vaux KK, Gleeson JG. The genetic landscape of autism spectrum disorders. Developmental Medicine and Child Neurology. 2014; 56: 12–18.
[7]
Harris SW, Hessl D, Goodlin-Jones B, Ferranti J, Bacalman S, Barbato I, et al. Autism profiles of males with fragile X syndrome. American Journal of Mental Retardation. 2008; 113: 427–438.
[8]
Bailey DB Jr, Mesibov GB, Hatton DD, Clark RD, Roberts JE, Mayhew L. Autistic behavior in young boys with fragile X syndrome. Journal of Autism and Developmental Disorders. 1998; 28: 499–508.
[9]
Belmonte MK, Bourgeron T. Fragile X syndrome and autism at the intersection of genetic and neural networks. Nature Neuroscience. 2006; 9: 1221–1225.
[10]
Zoghbi HY, Bear MF. Synaptic Dysfunction in Neurodevelopmental Intellectual Disabilities. Cold Spring Harbor Perspectives in Biology. 2012; 4: 1–22.
[11]
Bentham Science Publisher BSP. Immunoexcitotoxicity as a Central Mechanism of Autism Spectrum Disorders. In Strunecka A (Ed.) Cellular and Molecular Biology of Autism Spectrum Disorders (pp. 47–72). Bentham Science Publishers Ltd: Sharjah, UAE. 2010.
[12]
Blaylock RL. Immune Dysfunction in Autism Spectrum Disorder. In Strunecka A (Ed.) Cellular and Molecular Biology of Autism Spectrum Disorders (pp. 73–81). Bentham Science Publishers Ltd: Sharjah, UAE. 2010.
[13]
Meltzer A, Van de Water J. The Role of the Immune System in Autism Spectrum Disorder. Neuropsychopharmacology. 2017; 42: 284–298.
[14]
Careaga M, Water J, Ashwood P. Immune dysfunction in autism: a pathway to treatment. Neurotherapeutics. 2010; 7: 283–292.
[15]
Onore C, Careaga M, Ashwood P. The role of immune dysfunction in the pathophysiology of autism. Brain, Behavior, and Immunity. 2012; 26: 383–392.
[16]
Samsam M, Ahangari R, Naser SA. Pathophysiology of autism spectrum disorders: revisiting gastrointestinal involvement and immune imbalance. World Journal of Gastroenterology. 2014; 20: 9942–9951.
[17]
Wood AG, Nadebaum C, Anderson V, Reutens D, Barton S, O’Brien TJ, et al. Prospective assessment of autism traits in children exposed to antiepileptic drugs during pregnancy. Epilepsia. 2015; 56: 1047–1055.
[18]
Zou M, Liu Y, Xie S, Wang L, Li D, Li L, et al. Alterations of the endocannabinoid system and its therapeutic potential in autism spectrum disorder. Open Biology. 2021; 11: rsob.200306.
[19]
Kerr DM, Downey L, Conboy M, Finn DP, Roche M. Alterations in the endocannabinoid system in the rat valproic acid model of autism. Behavioural Brain Research. 2013; 249: 124–132.
[20]
Su T, Yan Y, Li Q, Ye J, Pei L. Endocannabinoid System Unlocks the Puzzle of Autism Treatment via Microglia. Front. Psychiatry. 2021; 12: 734837
[21]
Chakrabarti B, Persico A, Battista N, Maccarrone M. Endocannabinoid Signaling in Autism. Neurotherapeutics. 2015; 12: 837–847.
[22]
Krueger DD, Brose N. Evidence for a common endocannabinoid-related pathomechanism in autism spectrum disorders. Neuron. 2013; 78: 408–410.
[23]
Speed HE, Masiulis I, Gibson JR, Powell CM. Increased Cortical Inhibition in Autism-Linked Neuroligin-3R451C Mice is Due in Part to Loss of Endocannabinoid Signaling. PLoS ONE. 2015; 10: e0140638.
[24]
Zamberletti E, Gabaglio M, Parolaro D. The Endocannabinoid System and Autism Spectrum Disorders: Insights from Animal Models. International Journal of Molecular Sciences. 2017; 18: 1916.
[25]
Schultz S, Siniscalco D. Endocannabinoid system involvment in autism spectrum disorder: An overview with potential therapeutic application. AIMS Molecular Science. 2019; 6: 27-37
[26]
Busquets-Garcia A, Gomis-González M, Guegan T, Agustín-Pavón C, Pastor A, Mato S, et al. Targeting the endocannabinoid system in the treatment of fragile X syndrome. Nature Medicine. 2013; 19: 603–607.
[27]
Gomis-González M, Busquets-Garcia A, Matute C, Maldonado R, Mato S, Ozaita A. Possible Therapeutic Doses of Cannabinoid Type 1 Receptor Antagonist Reverses Key Alterations in Fragile X Syndrome Mouse Model. Genes. 2016; 7: 1–12.
[28]
Qin M, Zeidler Z, Moulton K, Krych L, Xia Z, Smith CB. Endocannabinoid-mediated improvement on a test of aversive memory in a mouse model of fragile X syndrome. Behavioural Brain Research. 2015; 291: 164–171.
[29]
Centre of Disease Control and Prevention. Data & Statistics Prevalence Risk Factors and Characteristics. 2020. Available at: http://www.cdc.gov/ncbddd/autism/data.html (Accessed: 28 March 2020).
[30]
Riesgo R, Gottfried C, and Becker M. Clinical Approach in Autism: Management and Treatment. Recent Advances in Autism Spectrum Disorders. 2013.
[31]
Anagnostou E, Zwaigenbaum L, Szatmari P, Fombonne E, Fernandez BA, Woodbury-Smith M, et al. Autism spectrum disorder: advances in evidence-based practice. Canadian Medical Association Journal. 2014; 186: 509–519.
[32]
Benvenuto A, Battan B, Porfirio MC, Curatolo P. Pharmacotherapy of autism spectrum disorders. Brain and Development. 2013; 35: 119–127.
[33]
Oswald DP, Sonenklar NA. Medication Use among Children with Autism Spectrum Disorders. Journal of Child and Adolescent Psychopharmacology. 2007; 17: 348–355.
[34]
Malow B, Adkins KW, McGrew SG, Wang L, Goldman SE, Fawkes D, et al. Melatonin for Sleep in Children with Autism: a Controlled Trial Examining Dose, Tolerability, and Outcomes. Journal of Autism and Developmental Disorders. 2012; 42: 1729–1737.
[35]
ROSSIGNOL DA, FRYE RE. Melatonin in autism spectrum disorders: a systematic review and meta-analysis. Developmental Medicine & Child Neurology. 2011; 53: 783–792.
[36]
National Conference on State Legislatures. State Medical Marijuana Laws. 2021. Avaliable at: http://www.ncsl.org/research/health/state-medical-marijuana-laws.aspx (Accessed: 19 September 2021).
[37]
ProCon.org. Legal medical marijuana states and DC. 2021. https://medicalmarijuana.procon.org/legal-medical-marijuana-states-and-dc/ (Accessed: 19 September 2021).
[38]
The Cannigma. Where Is Weed Legal in Europe? 2021. Updated European Marijuana Laws. 2021. Available at: cannigma.com (Accessed: 17 November 2021).
[39]
Pain S. A potted history. Nature. 2015; 525: S10–S11.
[40]
DEA. Implementation of the Agriculture Improvement Act of 2018. 2020. Available at: https://www.federalregister.gov/documents/2020/08/21/2020-17356/implementation-of-the-agriculture-improvement-act-of-2018 (Accessed: 29 April 2021).
[41]
Hillig KW, Mahlberg PG. A chemotaxonomic analysis of cannabinoid variation in Cannabis (Cannabaceae). American Journal of Botany. 2004; 91: 966–975.
[42]
Pollio A. The Name of Cannabis: a Short Guide for Nonbotanists. Cannabis and Cannabinoid Research. 2016; 1: 234–238.
[43]
Small E. Evolution and Classification of Cannabis sativa (Marijuana, Hemp) in Relation to Human Utilization. The Botanical Review. 2015; 81: 189–294.
[44]
Russo E, Guy GW. A tale of two cannabinoids: the therapeutic rationale for combining tetrahydrocannabinol and cannabidiol. Medical Hypotheses. 2005; 66: 234–246.
[45]
Russo EB. Taming THC: potential cannabis synergy and phytocannabinoid-terpenoid entourage effects. British Journal of Pharmacology. 2011; 163: 1344–1364.
[46]
Fishbein M, Gov S, Assaf F, Gafni M, Keren O, Sarne Y. Long-term behavioral and biochemical effects of an ultra-low dose of Δ9-tetrahydrocannabinol (THC): neuroprotection and ERK signaling. Experimental Brain Research. 2012; 221: 437–448.
[47]
Nahas G, Harvey DJ, Sutin K, Turndorf H, Cancro R. A molecular basis of the therapeutic and psychoactive properties of cannabis (Δ9-tetrahydrocannabinol). Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2002; 26: 721–730.
[48]
Burstein S. Cannabidiol (CBD) and its analogs: A review of their effects on inflammation. Bioorganic & Medicinal Chemistry. 2015; 23: 1377–1385.
[49]
Pisanti S, Malfitano AM, Ciaglia E, Lamberti A, Ranieri R, Cuomo G, et al. Cannabidiol: State of the art and new challenges for therapeutic applications. Pharmacology & Therapeutics. 2017; 175: 133–150.
[50]
Ruggeri B, Sarkans U, Schumann G, Persico AM. Biomarkers in autism spectrum disorder: the old and the new. Psychopharmacology. 2014; 231: 1201–1216.
[51]
Xu N, Li X, Zhong Y. Inflammatory Cytokines: Potential Biomarkers of Immunologic Dysfunction in Autism Spectrum Disorders. Mediators of Inflammation. 2015; 2015: 1–10.
[52]
Brigida AL, Schultz S, Cascone M, Antonucci N, Siniscalco D. Endocannabinod Signal Dysregulation in Autism Spectrum Disorders: A Correlation Link between Inflammatory State and Neuro-Immune Alterations. International Journal of Molecular Sciences. 2017; 18: 1425.
[53]
Lutz B. Endocannabinoid signals in the control of emotion. Current Opinion in Pharmacology. 2009; 9: 46–52.
[54]
Viveros M, Marco E, Llorente R, López-Gallardo M. Endocannabinoid System and Synaptic Plasticity: Implications for Emotional Responses. Neural Plasticity. 2007; 2007: 52908.
[55]
Marco EM, Rapino C, Caprioli A, Borsini F, Maccarrone M, Laviola G. Social encounter with a novel partner in adolescent rats: Activation of the central endocannabinoid system. Behavioural Brain Research. 2011; 220: 140–145.
[56]
Marco EM, Scattoni ML, Rapino C, Ceci C, Chaves N, Macrì S, et al. Emotional, endocrine and brain anandamide response to social challenge in infant male rats. Psychoneuroendocrinology. 2013; 38: 2152–2162.
[57]
Sciolino NR, Bortolato M, Eisenstein SA, Fu J, Oveisi F, Hohmann AG, et al. Social isolation and chronic handling alter endocannabinoid signaling and behavioral reactivity to context in adult rats. Neuroscience. 2010; 168: 371–386.
[58]
Pietropaolo S, Bellocchio L, Bouzón-Arnáiz I, Yee BK. The role of the endocannabinoid system in autism spectrum disorders: Evidence from mouse studies. Progress in Molecular Biology and Translational Science. 2020; 2: 183–208.
[59]
Babayeva M, Assefa H, Basu P, Chumki S, Loewy Z. Marijuana Compounds: a Nonconventional Approach to Parkinson’s Disease Therapy. Parkinson’s Disease. 2016; 2016: 1279042.
[60]
Babayeva M, Fuzailov M, Rozenfeld P and Basu P. Marijuana Compounds: A Non-Conventional Therapeutic Approach to Epilepsy in Children. Journal Addiction & Neuropharmacology. 2014; 1: 1
[61]
Cheung K, Mitchell M, Heussler H. Cannabidiol and Neurodevelopmental Disorders in Children. Frontiers in Psychiatry. 2021; 12: 643442
[62]
Campolongo P, Trezza V, Palmery M, Trabace L, Cuomo V. Developmental exposure to cannabinoids causes subtle and enduring neurofunctional alterations. International Review of Neurobiology. 2009; 85: 117–133.
[63]
Anavi-Goffer S, Mulder J. The polarized life of the endocannabinoid system in CNS development. ChemBioChem. 2009; 10: 1591–1598.
[64]
Drysdale AJ, Platt B. Cannabinoids: mechanisms and therapeutic applications in the CNS. Current Medicinal Chemistry. 2003; 10: 2719–2732.
[65]
Bauman ML, Kemper TL. Neuroanatomic observations of the brain in autism: a review and future directions. International Journal of Developmental Neuroscience. 2005; 23: 183–187.
[66]
Courchesne E, Pierce K, Schumann CM, Redcay E, Buckwalter JA, Kennedy DP, et al. Mapping Early Brain Development in Autism. Neuron. 2007; 56: 399–413.
[67]
Silva EAD Junior, Medeiros WMB, Torro N, Sousa JMM, Almeida IBCM, Costa FBD, et al. Cannabis and cannabinoid use in autism spectrum disorder: a systematic review. Trends Psychiatry Psychother. 2021. (in press)
[68]
Harkany T, Mackie K, Doherty P. Wiring and firing neuronal networks: endocannabinoids take center stage. Current Opinion in Neurobiology. 2008; 18: 338–345.
[69]
Keimpema E, Barabas K, Morozov YM, Tortoriello G, Torii M, Cameron G, et al. Differential Subcellular Recruitment of Monoacylglycerol Lipase Generates Spatial Specificity of 2-Arachidonoyl Glycerol Signaling during Axonal Pathfinding. Journal of Neuroscience. 2010; 30: 13992–14007.
[70]
Goines PE, Ashwood P. Cytokine dysregulation in autism spectrum disorders (ASD): Possible role of the environment. Neurotoxicology and Teratology. 2013; 36: 67–81.
[71]
Tonhajzerova I, Ondrejka I, Mestanik M, Mikolka P, Hrtanek I, Mestanikova A, et al. Inflammatory Activity in Autism Spectrum Disorder. Advances in Experimental Medicine and Biology. 2015; 173: 93–98.
[72]
Lunn CA, Reich E, Bober L. Targeting the CB2 receptor for immune modulation. Expert Opinion on Therapeutic Targets. 2006; 10: 653–663.
[73]
Pertwee RG, Howlett AC, Abood ME, Alexander SPH, Di Marzo V, Elphick MR, et al. International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid Receptors and their Ligands: beyond CB1and CB2. Pharmacological Reviews. 2010; 62: 588–631.
[74]
Carrier E, Patel S, Hillard C. Endocannabinoids in Neuroimmunology and Stress. Current Drug Target -CNS & Neurological Disorders. 2005; 4: 657–665.
[75]
Rajesh M, Mukhopadhyay P, Bátkai S, Haskó G, Liaudet L, Huffman JW, et al. CB2-receptor stimulation attenuates TNF-alpha-induced human endothelial cell activation, transendothelial migration of monocytes, and monocyteendothelial adhesion. The American Journal of Physiology-Heart and Circulatory Physiology. 2007; 293: H2210–H2218.
[76]
Tolón RM, Núñez E, Pazos MR, Benito C, Castillo AI, Martínez-Orgado JA, et al. The activation of cannabinoid CB2 receptors stimulates in situ and in vitro beta-amyloid removal by human macrophages. Brain Research. 2009; 1283: 148–154.
[77]
Cencioni MT, Chiurchiù V, Catanzaro G, Borsellino G, Bernardi G, Battistini L, et al. Anandamide suppresses proliferation and cytokine release from primary human T-lymphocytes mainly via CB2 receptors. PLoS ONE. 2010; 5: e8688.
[78]
Nezgovorova V, Ferretti CJ, Taylor BP, Shanahan E, Uzunova G, Hong K, et al. Potential of cannabinoids as treatments for autism spectrum disorders. Journal of Psychiatric Research. 2021; 137: 194–201.
[79]
Gandal MJ, Zhang P, Hadjimichael E, Walker RL, Chen C, Liu S, et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science. 2018; 362: eaat8127.
[80]
Griesi-Oliveira K, Fogo MS, Pinto BGG, Alves AY, Suzuki AM, Morales AG, et al. Transcriptome of iPSC-derived neuronal cells reveals a module of co-expressed genes consistently associated with autism spectrum disorder. Molecular Psychiatry. 2021; 26: 1589–1605.
[81]
Carbone E, Manduca A, Cacchione C, Vicari S, Trezza V. Healing autism spectrum disorder with cannabinoids: a neuroinflammatory story. Neuroscience & Biobehavioral Reviews. 2021; 121: 128–143
[82]
Warren RP, Singh VK, Averett RE, Odell JD, Maciulis A, Burger RA, et al. Immunogenetic studies in autism and related disorders. Molecular and Chemical Neuropathology. 1996; 28: 77–81.
[83]
Jyonouchi H, Geng L, Ruby A, Zimmerman-Bier B. Dysregulated Innate Immune Responses in Young Children with Autism Spectrum Disorders: their Relationship to Gastrointestinal Symptoms and Dietary Intervention. Neuropsychobiology. 2005; 51: 77–85.
[84]
Ashwood P, Wills S, van de Water J. The immune response in autism: A new frontier for autism research. Journal of Leukocyte Biology. 2006; 80: 1–15.
[85]
Molloy C, Morrow A, Meinzenderr J, Schleifer K, Dienger K, Manningcourtney P, et al. Elevated cytokine levels in children with autism spectrum disorder. Journal of Neuroimmunology. 2006; 172: 198–205.
[86]
Enstrom AM, Lit L, Onore CE, Gregg JP, Hansen RL, Pessah IN, et al. Altered gene expression and function of peripheral blood natural killer cells in children with autism. Brain, Behavior, and Immunity. 2009; 23: 124–133.
[87]
Li X, Chauhan A, Sheikh AM, Patil S, Chauhan V, Li X, et al. Elevated immune response in the brain of autistic patients. Journal of Neuroimmunology. 2009; 207: 111–116.
[88]
Batista TH, Giusti-Paiva A, Vilela FC. Maternal protein malnutrition induces autism-like symptoms in rat offspring. Nutritional Neuroscience. 2019; 22: 655–663.
[89]
Umathe SN, Manna SSS, Utturwar KS, Jain NS. Endocannabinoids mediate anxiolytic-like effect of acetaminophen via CB1 receptors. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2009; 33: 1191–1199.
[90]
Schultz ST. Can autism be triggered by acetaminophen activation of the endocannabinoid system? Acta Neurobiologiae Experimentalis. 2010; 70: 227–231.
[91]
Högestätt ED, Jönsson BAG, Ermund A, Andersson DA, Björk H, Alexander JP, et al. Conversion of Acetaminophen to the Bioactive N-Acylphenolamine am404 via Fatty Acid Amide Hydrolase-dependent Arachidonic Acid Conjugation in the Nervous System. Journal of Biological Chemistry. 2005; 280: 31405–31412.
[92]
Bertolini A, Ferrari A, Ottani A, Guerzoni S, Tacchi R, Leone S. Paracetamol: New Vistas of an Old Drug. CNS Drug Reviews. 2006; 12: 250–275.
[93]
De Petrocellis L, Bisogno T, Davis JB, Pertwee RG, Di Marzo V. Overlap between the ligand recognition properties of the anandamide transporter and the VR1 vanilloid receptor: inhibitors of anandamide uptake with negligible capsaicin-like activity. FEBS Letters. 2000; 483: 52–56.
[94]
Zygmunt PM, Chuang H, Movahed P, Julius D, Högestätt ED. The anandamide transport inhibitor am404 activates vanilloid receptors. European Journal of Pharmacology. 2000; 396: 39–42.
[95]
Fegley D, Kathuria S, Mercier R, Li C, Goutopoulos A, Makriyannis A, et al. Anandamide transport is independent of fatty-acid amide hydrolase activity and is blocked by the hydrolysis-resistant inhibitor am1172. Proceedings of the National Academy of Sciences. 2004; 101: 8756–8761.
[96]
Szallasi A, Di Marzo V. New perspectives on enigmatic vanilloid receptors. Trends in Neurosciences. 2000; 23: 491–497.
[97]
Morena M, De Castro V, Gray JM, Palmery M, Trezza V, Roozendaal B, et al. Training-Associated Emotional Arousal Shapes Endocannabinoid Modulation of Spatial Memory Retrieval in Rats. Journal of Neuroscience. 2015; 35: 13962–13974.
[98]
Chakrabarti B, Kent L, Suckling J, Bullmore E, Baron-Cohen S. Variations in the human cannabinoid receptor (CNR1) gene modulate striatal responses to happy faces. European Journal of Neuroscience. 2006; 23: 1944–1948.
[99]
Chakrabarti B, Baron-Cohen S. Variation in the human cannabinoid receptor CNR1 gene modulates gaze duration for happy faces. Molecular Autism. 2011; 2: 10.
[100]
Zhao P, Leonoudakis D, Abood ME, Beattie EC. Cannabinoid receptor activation reduces TNFα-Induced surface localization of AMPAR-type glutamate receptors and excitotoxicity. Neuropharmacology. 2010; 58: 551–558.
[101]
Trezza V, Damsteegt R, Manduca A, Petrosino S, Van Kerkhof LWM, Pasterkamp RJ, et al. Endocannabinoids in Amygdala and Nucleus Accumbens Mediate Social Play Reward in Adolescent Rats. Journal of Neuroscience. 2012; 32: 14899–14908.
[102]
Marzo Vd, Bisogno T, Sugiura T, Melck D, Petrocellis LD. The novel endogenous cannabinoid 2-arachidonoylglycerol is inactivated by neuronal- and basophil-like cells: connections with anandamide. Biochemical Journal. 1998; 331: 15–19.
[103]
Trezza V, Vanderschuren LJMJ. Bidirectional cannabinoid modulation of social behavior in adolescent rats. Psychopharmacology. 2008; 197: 217–227.
[104]
Aran A, Eylon M, Harel M, Polianski L, Nemirovski A, Tepper S, et al. Lower circulating endocannabinoid levels in children with autism spectrum disorder. Molecular Autism. 2019; 10: 2.
[105]
Denner LA, Rodriguez-Rivera J, Haidacher SJ, Jahrling JB, Carmical JR, Hernandez CM, et al. Cognitive enhancement with rosiglitazone links the hippocampal PPARgamma and ERK MAPK signaling pathways. Journal of Neuroscience. 2012; 32: 16725–16735.
[106]
de Magistris L, Familiari V, Pascotto A, Sapone A, Frolli A, Iardino P, et al. Alterations of the Intestinal Barrier in Patients with Autism Spectrum Disorders and in their first-degree Relatives. Journal of Pediatric Gastroenterology & Nutrition. 2010; 51: 418–424.
[107]
de Magistris L, Picardi A, Siniscalco D, Riccio MP, Sapone A, Cariello R, et al. Antibodies against Food Antigens in Patients with Autistic Spectrum Disorders. BioMed Research International. 2013; 2013: 729349.
[108]
Gao F, Zhang L, Su T, Li L, Zhou R, Peng M, et al. Signaling Mechanism of Cannabinoid Receptor-2 Activation-Induced β-Endorphin Release. Molecular Neurobiology. 2016; 53: 3616–3625.
[109]
van der Stelt M, Di Marzo V. The endocannabinoid system in the basal ganglia and in the mesolimbic reward system: implications for neurological and psychiatric disorders. European Journal of Pharmacology. 2003; 480: 133–150.
[110]
Fernández-Ruiz J, Hernández M, Ramos JA. Cannabinoid-dopamine interaction in the pathophysiology and treatment of CNS disorders. CNS Neuroscience & Therapeutics. 2010; 16: e72–e91.
[111]
Croxford JL. Therapeutic Potential of Cannabinoids in CNS Disease. CNS Drugs. 2003; 17: 179–202.
[112]
Howlett AC, Abood ME. CB 1 and CB 2 Receptor Pharmacology. Cannabinoid Pharmacology. 2017; 53: 169–206.
[113]
Gould GG, Burke TF, Osorio MD, Smolik CM, Zhang WQ, Onaivi ES, et al. Enhanced novelty-induced corticosterone spike and upregulated serotonin 5-HT1a and cannabinoid CB1 receptors in adolescent BTBR mice. Psychoneuroendocrinology. 2014; 39: 158–169.
[114]
Wei D, Dinh D, Lee D, Li D, Anguren A, Moreno-Sanz G, et al. Enhancement of Anandamide-Mediated Endocannabinoid Signaling Corrects Autism-Related Social Impairment. Cannabis and Cannabinoid Research. 2016; 1: 81–89.
[115]
S. Onaivi E, Benno R, Halpern T, Mehanovic M, Schanz N, Sanders C, et al. Consequences of Cannabinoid and Monoaminergic System Disruption in a Mouse Model of Autism Spectrum Disorders. Current Neuropharmacology. 2011; 9: 209–214.
[116]
Liu QR, Pan CH, Hishimoto A, Li CY, Xi ZX, Llorente-Berzal A, et al. Species differences in cannabinoid receptor 2 (CNR2gene): identification of novel human and rodent CB2 isoforms, differential tissue expression and regulation by cannabinoid receptor ligands. Genes, Brain and Behavior. 2009; 8: 519–530.
[117]
Siniscalco D, Sapone A, Giordano C, Cirillo A, de Magistris L, Rossi F, et al. Cannabinoid Receptor Type 2, but not Type 1, is up-Regulated in Peripheral Blood Mononuclear Cells of Children Affected by Autistic Disorders. Journal of Autism and Developmental Disorders. 2013; 43: 2686–2695.
[118]
Depino AM. Peripheral and central inflammation in autism spectrum disorders. Molecular and Cellular Neuroscience. 2013; 53: 69–76.
[119]
Leleu-Chavain N, Desreumaux P, Chavatte P, Millet R. Therapeutical potential of CB2receptors in immune-related diseases. Current Molecular Pharmacology. 2013; 6: 183–203.
[120]
Rom S, Persidsky Y. Cannabinoid Receptor 2: Potential Role in Immunomodulation and Neuroinflammation. Journal of Neuroimmune Pharmacology. 2013; 8: 608–620.
[121]
Chiurchiù V, Battistini L, Maccarrone M. Endocannabinoid signaling in innate and adaptive immunity. The Journal of Immunology. 2015; 144: 352–364.
[122]
Maccarrone M, Rossi S, Bari M, De Chiara V, Rapino C, Musella A, et al. Abnormal mGlu 5 Receptor/Endocannabinoid Coupling in Mice Lacking FMRP and BC1 RNA. Neuropsychopharmacology. 2010; 35: 1500–1509.
[123]
Folkes OM, Báldi R, Kondev V, Marcus DJ, Hartley ND, Turner BD, et al. An endocannabinoid-regulated basolateral amygdala–nucleus accumbens circuit modulates sociability. Journal of Clinical Investigation. 2020; 130: 1728–1742.
[124]
Jamain S, Quach H, Betancur C, Råstam M, Colineaux C, Gillberg IC, et al. Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism. Nature Genetics. 2003; 34: 27–29.
[125]
Comoletti D. The Arg451Cys-Neuroligin-3 Mutation Associated with Autism Reveals a Defect in Protein Processing. Journal of Neuroscience. 2004; 24: 4889–4893.
[126]
Földy C, Malenka RC, Südhof TC. Autism-associated neuroligin-3 mutations commonly disrupt tonic endocannabinoid signaling. Neuron. 2013; 78: 498–509.
[127]
Hosie S, Malone DT, Liu S, Glass M, Adlard PA, Hannan AJ, et al. Altered Amygdala Excitation and CB1 Receptor Modulation of Aggressive Behavior in the Neuroligin-3R451C Mouse Model of Autism. Frontiers in Cellular Neuroscience. 2018; 12: 234.
[128]
Servadio M, Melancia F, Manduca A, di Masi A, Schiavi S, Cartocci V, et al. Targeting anandamide metabolism rescues core and associated autistic-like symptoms in rats prenatally exposed to valproic acid. Translational Psychiatry. 2016; 6: e902–e902.
[129]
Premoli M, Aria F, Bonini SA, Maccarinelli G, Gianoncelli A, Pina SD, et al. Cannabidiol: Recent advances and new insights for neuropsychiatric disorders treatment. Life Sciences. 2019; 224: 120–127.
[130]
Schultz S, Gould GG, Antonucci N, Brigida AL, Siniscalco D. Endocannabinoid System Dysregulation from Acetaminophen Use May Lead to Autism Spectrum Disorder: Could Cannabinoid Treatment Be Efficacious? Molecules. 2021; 26: 7.
[131]
Loss CM, Teodoro L, Rodrigues GD, Moreira LR, Peres FF, Zuardi AW, et al. Is Cannabidiol During Neurodevelopment a Promising Therapy for Schizophrenia and Autism Spectrum Disorders? Frontiers in Pharmacology. 2021; 11: 635763.
[132]
Elmes MW, Kaczocha M, Berger WT, Leung K, Ralph BP, Wang L, et al. Fatty Acid-binding Proteins (FABPs) are Intracellular Carriers for Δ9-Tetrahydrocannabinol (THC) and Cannabidiol (CBD). Journal of Biological Chemistry. 2015; 290: 8711–8721.
[133]
Kaplan JS, Stella N, Catterall WA, Westenbroek RE. Cannabidiol attenuates seizures and social deficits in a mouse model of Dravet syndrome. Proceedings of the National Academy of Sciences. 2017; 114: 11229–11234.
[134]
Osborne AL, Solowij N, Weston-Green K. A systematic review of the effect of cannabidiol on cognitive function: Relevance to schizophrenia. Neuroscience & Biobehavioral Reviews. 2017; 72: 310–324.
[135]
Iannotti FA, Pagano E, Moriello AS, Alvino FG, Sorrentino NC, D’Orsi L, et al. Effects of non-euphoric plant cannabinoids on muscle quality and performance of dystrophic mdx mice. British Journal of Pharmacology. 2019; 176: 1568–1584.
[136]
Patra PH, Serafeimidou-Pouliou E, Bazelot M, Whalley BJ, Williams CM, McNeish AJ. Cannabidiol improves survival and behavioural co-morbidities of Dravet syndrome in mice. British Journal of Pharmacology. 2020; 177: 2779–2792.
[137]
Martín-Moreno AM, Reigada D, Ramírez BG, Mechoulam R, Innamorato N, Cuadrado A, et al. Cannabidiol and other Cannabinoids Reduce Microglial Activation in Vitro and in Vivo: Relevance to Alzheimer’s Disease. Molecular Pharmacology. 2011; 79: 964–973.
[138]
De Petrocellis L, Ligresti A, Moriello AS, Allarà M, Bisogno T, Petrosino S, et al. Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. British Journal of Pharmacology. 2011; 163: 1479–1494.
[139]
Amada N, Yamasaki Y, Williams CM, Whalley BJ. Cannabidivarin (CBDV) suppresses pentylenetetrazole (PTZ)-induced increases in epilepsy-related gene expression. PeerJ. 2013; 1: e214.
[140]
Pagano E, Romano B, Iannotti FA, Parisi OA, D’Armiento M, Pignatiello S, et al. The non-euphoric phytocannabinoid cannabidivarin counteracts intestinal inflammation in mice and cytokine expression in biopsies from UC pediatric patients. Pharmacological Research. 2019; 149: 104464.
[141]
Hill TDM, Cascio M, Romano B, Duncan M, Pertwee RG, Williams CM, et al. Cannabidivarin-rich cannabis extracts are anticonvulsant in mouse and rat via a CB1 receptor-independent mechanism. British Journal of Pharmacology. 2013; 170: 679–692.
[142]
Poleg S, Golubchik P, Offen D, Weizman A. Cannabidiol as a suggested candidate for treatment of autism spectrum disorder. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2019; 89: 90–96.
[143]
Bonini SA, Premoli M, Tambaro S, Kumar A, Maccarinelli G, Memo M, et al. Cannabis sativa: a comprehensive ethnopharmacological review of a medicinal plant with a long history. Journal of Ethnopharmacology. 2018; 227: 300–315.
[144]
Vigli D, Cosentino L, Raggi C, Laviola G, Woolley-Roberts M, De Filippis B. Chronic treatment with the phytocannabinoid Cannabidivarin (CBDV) rescues behavioural alterations and brain atrophy in a mouse model of Rett syndrome. Neuropharmacology. 2018; 140: 121–129.
[145]
Zamberletti E, Gabaglio M, Piscitelli F, Brodie JS, Woolley-Roberts M, Barbiero I, et al. Cannabidivarin completely rescues cognitive deficits and delays neurological and motor defects in male Mecp2 mutant mice. Journal of Psychopharmacology. 2019; 33: 894–907.
[146]
Zamberletti E, Gabaglio M, Woolley-Roberts M, Bingham S, Rubino T, Parolaro D. Cannabidivarin treatment ameliorates autism-like behaviors and restores hippocampal endocannabinoid system and glia alterations induced by prenatal valproic acid exposure in rats. Frontiers in Cellular Neuroscience. 2019; 13: 367.
[147]
Ahn Y, Han SH, Kim MG, Hong K, Kim WJ, Suh HJ, et al. Anti-depressant effects of ethanol extract from Cannabis sativa (hemp) seed in chlorpromazine-induced Drosophila melanogaster depression model. Pharmaceutical Biology. 2021; 59: 998–1007.
[148]
Babson KA, Sottile J, Morabito D. Cannabis, Cannabinoids, and Sleep: a Review of the Literature. Current Psychiatry Reports. 2017; 19: 23.
[149]
Devinsky O, Verducci C, Thiele EA, Laux LC, Patel AD, Filloux F, et al. Open-label use of highly purified CBD (Epidiolex®) in patients with CDKL5 deficiency disorder and Aicardi, Dup15q, and Doose syndromes. Epilepsy & Behavior. 2018; 86: 131–137.
[150]
Devinsky O, Cross JH, Laux L, Marsh E, Miller I, Nabbout R, et al. Trial of Cannabidiol for Drug-Resistant Seizures in the Dravet Syndrome. New England Journal of Medicine. 2017; 376: 2011–2020.
[151]
Ben Amar M. Cannabinoids in medicine: a review of their therapeutic potential. Journal of Ethnopharmacology. 2006; 105: 1–25.
[152]
Bossong MG, Niesink RJM. Adolescent brain maturation, the endogenous cannabinoid system and the neurobiology of cannabis-induced schizophrenia. Progress in Neurobiology. 2010; 92: 370–385.
[153]
Leafly. Jeremy Kossen. How Does Cannabis Consumption Affect Autism? 2016. Available at: https://www.leafly.com/news/health/how-does-cannabis-consumption-affect-autism (Accessed: 18 February 2021).
[154]
Washington Post. Myung-Ok Lee M. I made my son cannabis cookies. 2017. Available at: https://www.washingtonpost.com/opinions/i-made-my-son-cannabis-cookies-they-changed-his-life/2017/01/06/699b1d20-d1ef-11e6-a783-cd3fa950f2fd_story.html?utm_term=.f2dfbd34a7ac (Accessed: 27 September 2021).
[155]
AAMC (The American Alliance for Medical Cannabis). Using Medical Cannabis to Treat Autism Spectrum Disorder. 2014. Available at: http://www.letfreedomgrow.com/cmu/SamsStory.htm (Accessed: 8 August 2021).
[156]
CBS Philly. Abrams M. Mom Spurs Marijuana-Autism Study In Philadelphia. 2017. Available at: http://philadelphia.cbslocal.com/2017/12/16/mom-spurs-marijuana-autism-study-in-philadelphia/ (Accessed: 29 June 2021).
[157]
Huffpost. Miles K. Marijuana-Like Chemical May Help Autism And Fragile X Syndrome Symptoms. 2012. Available at: https://www.huffingtonpost.com/2012/09/27/marijuana-chemical-autism-fragile-x_n_1920320.html (Accessed: 14 July 2021).
[158]
MassRoots. Rabinski G. Can Cannabis Treat Autism? 2015. Available at: https://www.massroots.com/learn/cannabis-treatment-autism/ (Accessed: 15 July 2021).
[159]
Kurz R, Blaas K. Use of dronabinol (delta-9-THC) in autism: A prospective single-case-study with an early infantile autistic child. Cannabinoids. 2010; 5: 4–6.
[160]
Kruger T, Christophersen E. An Open Label Study of the Use of Dronabinol (Marinol) in the Management of Treatment-Resistant Self-Injurious Behavior in 10 Retarded Adolescent Patients. Journal of Developmental & Behavioral Pediatrics. 2006; 27: 433.
[161]
Aran A. Clinical Trial on Autistic Disorder: Cannabinoids - 99% pure cannabinoids mix, Placebo, Cannabinoids - whole plant extract. 2018. https://ichgcp.net/clinical-trials-registry/NCT02956226 (Accessed: 30 August 2021).
[162]
Aran A, Harel M, Cassuto H, Polyansky L, Schnapp A, Wattad N, et al. Cannabinoid treatment for autism: a proof-of-concept randomized trial. Molecular Autism. 2021; 12: 6.
[163]
Bar-Lev Schleider L, Mechoulam R, Saban N, Meiri G, Novack V. Real life Experience of Medical Cannabis Treatment in Autism: Analysis of Safety and Efficacy. Scientific Reports. 2019; 9: 200.
[164]
Barchel D, Stolar O, De-Haan T, Ziv-Baran T, Saban N, Fuchs DO, et al. Oral cannabidiol use in children with autism spectrum disorder to treat related symptoms and co-morbidities. Frontiers in Pharmacology. 2019; 9: 1521.
[165]
Pretzsch CM, Voinescu B, Mendez MA, Wichers R, Ajram L, Ivin G, et al. The effect of cannabidiol (CBD) on low-frequency activity and functional connectivity in the brain of adults with and without autism spectrum disorder (ASD). Journal of Psychopharmacology. 2019; 33: 1141–1148.
[166]
Castellanos F. Cannabidiol for ASD. Clinical Trial NCT03900923. 2019. https://clinicaltrials.gov/ct2/show/NCT03900923 (Accessed: 7 October 2021).
[167]
McAlonan G. Shifting Brain Excitation-Inhibition Balance in Autism Spectrum Disorder. 2016; https://clinicaltrials.gov/ct2/show/NCT03537950 (Accessed: 7 October 2021).
[168]
Hollander E. Cannabidivarin (CBDV) vs. Placebo in Children With Autism Spectrum Disorder (ASD). 2017. https://clinicaltrials.gov/ct2/show/NCT03202303 (Accessed: 7 October 2021).
[169]
Tartaglia N, Bonn-Miller M, Hagerman R. Treatment of Fragile X Syndrome with Cannabidiol: a Case Series Study and Brief Review of the Literature. Cannabis and Cannabinoid Research. 2019; 4: 3–9.
[170]
Lisdahl KM, Gilbart ER, Wright NE, Shollenbarger S. Dare to delay? The impacts of adolescent alcohol and marijuana use onset on cognition, brain structure, and function. Frontiers in Psychiatry. 2013; 4: 53.
[171]
Hadland SE, Knight JR, Harris SK. Medical Marijuana: review of the science and implications for developmental-behavioral pediatric practice. Journal of Developmental & Behavioral Pediatrics. 2015; 36: 115–123.
[172]
Crean RD, Crane NA, Mason BJ. An Evidence-Based Review of Acute and Long-Term Effects of Cannabis Use on Executive Cognitive Functions. Journal of Addiction Medicine. 2011; 5: 1–8.
[173]
Meier MH, Caspi A, Ambler A, Harrington H, Houts R, Keefe RSE, et al. Persistent cannabis users show neuropsychological decline from childhood to midlife. Proceedings of the National Academy of Sciences. 2012; 109: E2657–E2664.
[174]
Winstock AR, Ford C, Witton J. Assessment and management of cannabis use disorders in primary care. British Medical Journal. 2010; 340: c1571–c1571.
[175]
Schneider M. Puberty as a highly vulnerable developmental period for the consequences of cannabis exposure. Addiction Biology. 2008; 13: 253–263.
[176]
Moore TH, Zammit S, Lingford-Hughes A, Barnes TR, Jones PB, Burke M, et al. Cannabis use and risk of psychotic or affective mental health outcomes: a systematic review. The Lancet. 2007; 370: 319–328.
[177]
Malone DT, Hill MN, Rubino T. Adolescent cannabis use and psychosis: epidemiology and neurodevelopmental models. British Journal of Pharmacology. 2010; 160: 511–522.
[178]
Gerra G, Zaimovic A, Gerra M, Ciccocioppo R, Cippitelli A, Serpelloni G, et al. Pharmacology and Toxicology of Cannabis Derivatives and Endocannabinoid Agonists. Recent Patents on CNS Drug Discovery. 2010; 5: 46–52.
[179]
Grotenhermen F. The toxicology of cannabis and cannabis prohibition. Chemistry & Biodiversity. 2007; 4: 1744–1769.
[180]
Araujo DJ, Tjoa K, Saijo K. The endocannabinoid system as a window into microglial biology and its relationship to autism. Frontiers in Cellular Neuroscience. 2019; 13: 424.
[181]
Hu VW, Bi C. Phenotypic Subtyping and Re-analyses of Existing Transcriptomic Data from Autistic Probands in Simplex Families Reveal Differentially Expressed and ASD Trait-Associated Genes. Frontiers in Neurology. 2020; 11: 578972.
[182]
Karalunas SL, Hawkey E, Gustafsson H, Miller M, Langhorst M, Cordova M, et al. Overlapping and Distinct Cognitive Impairments in Attention-Deficit/Hyperactivity and Autism Spectrum Disorder without Intellectual Disability. Journal of Abnormal Child Psychology. 2018; 46: 1705–1716.
[183]
Attention-deficit / hyperactivity disorder (ADHD): Symptoms and diagnosis. 2021. Available at: https://www.cdc.gov/ncbddd/adhd/diagnosis.html (Accessed: 3 October 2021).
[184]
Mitchell JT, Sweitzer MM, Tunno AM, Kollins SH, McClernon FJ. “I use weed for my ADHD”: A qualitative analysis of online forum discussions on cannabis use and ADHD. PLoS ONE. 2016; 11: e0156614.
[185]
Tamm L, Epstein JN, Lisdahl KM, Molina B, Tapert S, Hinshaw SP, et al. Impact of ADHD and cannabis use on executive functioning in young adults. Drug and Alcohol Dependence. 2013; 133: 607–614.
[186]
Cooper RE, Williams E, Seegobin S, Tye C, Kuntsi J, Asherson P. Cannabinoids in attention-deficit/hyperactivity disorder: a randomised-controlled trial. European Neuropsychopharmacology. 2017; 27: 795–808.
[187]
Strohbeck-Kuehner P, Skopp G, Mattern R. Cannabis improves symptoms of ADHD. Cannabinoids. 2008; 3: 1–3.
[188]
Budney AJ. Review of the Validity and Significance of Cannabis Withdrawal Syndrome. American Journal of Psychiatry. 2004; 161: 1967–1977.
[189]
Hergenrather JY, Aviram J, Vysotski Y, Campisi-Pinto S, Lewitus GM, Meiri D. Cannabinoid and Terpenoid Doses are Associated with Adult ADHD Status of Medical Cannabis Patients. Rambam Maimonides Medical Journal. 2020; 11: e0001.
[190]
Zulauf CA, Sprich SE, Safren SA, Wilens TE. The Complicated Relationship between Attention Deficit/Hyperactivity Disorder and Substance Use Disorders. Current Psychiatry Reports. 2014; 16: 436.
[191]
Goldschmidt L, Day NL, Richardson GA. Effects of prenatal marijuana exposure on child behavior problems at age 10. Neurotoxicology and Teratology. 2000; 22: 325–336.
[192]
Wu C, Jew CP, Lu H. Lasting impacts of prenatal cannabis exposure and the role of endogenous cannabinoids in the developing brain. Future Neurology. 2011; 6: 459–480.
[193]
Wilens TE, Adamson J, Sgambati S, Whitley J, Santry A, Monuteaux MC, et al. Do Individuals with ADHD Self-Medicate with Cigarettes and Substances of Abuse? Results from a Controlled Family Study of ADHD. American Journal on Addictions. 2007; 16: 14–23.
[194]
Tuchman R, Rapin I. Epilepsy in autism. The Lancet Neurology. 2002; 1: 352–358.
[195]
Bolton PF, Carcani-Rathwell I, Hutton J, Goode S, Howlin P, Rutter M. Epilepsy in autism: features and correlates. British Journal of Psychiatry. 2011; 198: 289–294.
[196]
Amiet C, Gourfinkel-An I, Bouzamondo A, Tordjman S, Baulac M, Lechat P, et al. Epilepsy in Autism is Associated with Intellectual Disability and Gender: Evidence from a Meta-Analysis. Biological Psychiatry. 2008; 64: 577–582.
[197]
El Achkar CM, Spence SJ. Clinical characteristics of children and young adults with co-occurring autism spectrum disorder and epilepsy. Epilepsy & Behavior. 2015; 47: 183–190.
[198]
Clarke DF, Roberts W, Daraksan M, Dupuis A, McCabe J, Wood H, et al. The Prevalence of Autistic Spectrum Disorder in Children Surveyed in a Tertiary Care Epilepsy Clinic. Epilepsia. 2005; 46: 1970–1977.
[199]
Frye R. A review of traditional and novel treatments for seizures in autism spectrum disorder: findings from a systematic review and expert panel. Frontiers in Public Health. 2013; 1: 1–26.
[200]
Depositario-Cabacar DFT, Zelleke T. Treatment of epilepsy in children with developmental disabilities. Developmental Disabilities Research Reviews. 2010; 16: 239–247.
[201]
Lozano I. The Therapeutic Use of Cannabis sativa in Arabic Medicine. Journal of Cannabis Therapeutics. 2001; 1: 63–70.
[202]
Russo EB. Cannabis and epilepsy: an ancient treatment returns to the fore. Epilepsy & Behavior. 2017; 70: 292–297.
[203]
Ley W. On the efficacy of Indian hemp in some convulsive disorders. Provincial Medical Journal and Retrospect of the Medical Sciences. 1842; 4: 407–409.
[204]
Mechoulam R. Cannabis and epilepsy. Epilepsy & Behavior. 2017; 70: 278–279.
[205]
Devinsky O, Cilio MR, Cross H, Fernandez-Ruiz J, French J, Hill C, et al. Cannabidiol: Pharmacology and potential therapeutic role in epilepsy and other neuropsychiatric disorders. Epilepsia. 2014; 55: 791–802.
[206]
Friedman D, Sirven JI. Historical perspective on the medical use of cannabis for epilepsy: Ancient times to the 1980s. Epilepsy & Behavior. 2017; 70: 298–301.
[207]
Wallace MJ, Martin BR, DeLorenzo RJ. Evidence for a physiological role of endocannabinoids in the modulation of seizure threshold and severity. European Journal of Pharmacology. 2002; 452: 295–301.
[208]
Wallace MJ, Wiley JL, Martin BR, DeLorenzo RJ. Assessment of the role of CB1 receptors in cannabinoid anticonvulsant effects. European Journal of Pharmacology. 2001; 428: 51–57.
[209]
Ibeas Bih C, Chen T, Nunn AVW, Bazelot M, Dallas M, Whalley BJ. Molecular Targets of Cannabidiol in Neurological Disorders. Neurotherapeutics. 2015; 12: 699–730.
[210]
Rosenberg EC, Tsien RW, Whalley BJ, Devinsky O. Cannabinoids and Epilepsy. Neurotherapeutics. 2015; 12: 747–768.
[211]
Devinsky O, Marsh E, Friedman D, Thiele E, Laux L, Sullivan J, et al. Cannabidiol in patients with treatment-resistant epilepsy: an open-label interventional trial. The Lancet Neurology. 2016; 15: 270–278.
[212]
Thiele EA, Marsh ED, French JA, Mazurkiewicz-Beldzinska M, Benbadis SR, Joshi C, et al. Cannabidiol in patients with seizures associated with Lennox-Gastaut syndrome (GWPCARE4): a randomised, double-blind, placebo-controlled phase 3 trial. The Lancet. 2018; 391: 1085–1096.
[213]
Madan Cohen J, Checketts D, Dunayevich E, Gunning B, Hyslop A, Madhavan D, Villanueva V, Zolnowska M, Zuberi SM. Time to onset of cannabidiol treatment effects in Dravet syndrome: Analysis from two randomized controlled trials. Epilepsia. 2021; 62: 2218–2227.
[214]
EPIDIOLEX® (cannabidiol). A treatment Innovation. 2021. https://www.epidiolex.com/about-epidiolex#:~:text=EPIDIOLEX%3A%20A,year%20of%20age%20and%20older (Accessed: 4 June 2021).
[215]
van Steensel FJA, Bögels SM, Perrin S. Anxiety Disorders in Children and Adolescents with Autistic Spectrum Disorders: a Meta-Analysis. Clinical Child and Family Psychology Review. 2011; 14: 302–317.
[216]
Indiana University. Indiana Resource Center for Autism. 2016. Available at: https://www.iidc.indiana.edu/pages/anxiety-and-autism-spectrum-disorders (Accessed: 27 September 2021).
[217]
Fakhoury M. Could cannabidiol be used as an alternative to antipsychotics? Journal of Psychiatric Research. 2016; 80: 14–21.
[218]
NIH. Overdose Death Rate. 2021. Available at: https://www.drugabuse.gov/related-topics/trends-statistics/overdose-death-rates (Accessed: 25 June 2021).
[219]
Harvard Health Publishing. Medical Marijuana. 2018. Available at: https://www.health.harvard.edu/mind-and-mood/medical-marijuana-and-the-mind (Accessed: 7 April 2021).
[220]
Bhattacharyya S. Induction of Psychosis byΔ9-Tetrahydrocannabinol Reflects Modulation of Prefrontal and Striatal Function during Attentional Salience Processing. Archives of General Psychiatry. 2012; 69: 27.
[221]
Szuster RR, Pontius EB, Campos PE. Marijuana sensitivity and panic anxiety. The Journal of Clinical Psychiatry. 1988; 49: 427–429.
[222]
Moreira FA, Aguiar DC, Guimarães FS. Anxiolytic-like effect of cannabidiol in the rat Vogel conflict test. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2006; 30: 1466–1471.
[223]
Bhattacharyya S, Morrison PD, Fusar-Poli P, Martin-Santos R, Borgwardt S, Winton-Brown T, et al. Opposite effects of delta-9-tetrahydrocannabinol and cannabidiol on human brain function and psychopathology. Neuropsychopharmacology. 2010; 35: 764–774.
[224]
Zuardi AW, Crippa JAS, Hallak JEC, Moreira FA, Guimarães FS. Cannabidiol, a Cannabis sativa constituent, as an antipsychotic drug. Brazilian Journal of Medical and Biological Research. 2006; 39: 421–429.
[225]
McPartland JM, Duncan M, Di Marzo V, Pertwee RG. Are cannabidiol and Δ9-tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. British Journal of Pharmacology. 2015; 172: 737–753.
[226]
Fernández-Ruiz J, Sagredo O, Pazos MR, García C, Pertwee R, Mechoulam R, et al. Cannabidiol for neurodegenerative disorders: important new clinical applications for this phytocannabinoid? British Journal of Clinical Pharmacology. 2013; 75: 323–333.
[227]
Campos AC, de Paula Soares V, Carvalho MC, Ferreira FR, Vicente MA, Brandão ML, et al. Involvement of serotonin-mediated neurotransmission in the dorsal periaqueductal gray matter on cannabidiol chronic effects in panic-like responses in rats. Psychopharmacology. 2013; 226: 13–24.
[228]
Bakas T, van Nieuwenhuijzen PS, Devenish SO, McGregor IS, Arnold JC, Chebib M. The direct actions of cannabidiol and 2-arachidonoyl glycerol at GABAA receptors. Pharmacological Research. 2017; 119: 358–370.
[229]
Fogaça MV, Reis FMCV, Campos AC, Guimarães FS. Effects of intra-prelimbic prefrontal cortex injection of cannabidiol on anxiety-like behavior: Involvement of 5HT1a receptors and previous stressful experience. European Neuropsychopharmacology. 2014; 24: 410–419.
[230]
Soares VDP, Campos AC, Bortoli VCD, Zangrossi H, Guimarães FS, Zuardi AW. Intra-dorsal periaqueductal gray administration of cannabidiol blocks panic-like response by activating 5-HT1a receptors. Behavioural Brain Research. 2010; 213: 225–229.
[231]
Guimarães FS, Chiaretti TM, Graeff FG, Zuardi AW. Antianxiety effect of cannabidiol in the elevated plus-maze. Psychopharmacology. 1990; 100: 558–559.
[232]
Lemos JI, Resstel LB, Guimarães FS. Involvement of the prelimbic prefrontal cortex on cannabidiol-induced attenuation of contextual conditioned fear in rats. Behavioural Brain Research. 2010; 207: 105–111.
[233]
Schier A, Ribeiro N, Coutinho D, Machado S, Arias-Carrion O, Crippa J, et al. Antidepressant-Like and Anxiolytic-Like Effects of Cannabidiol: a Chemical Compound of Cannabis sativa. CNS & Neurological Disorders - Drug Targets. 2014; 13: 953–960.
[234]
Bitencourt RM, Pamplona FA, Takahashi RN. Facilitation of contextual fear memory extinction and anti-anxiogenic effects of am404 and cannabidiol in conditioned rats. European Neuropsychopharmacology. 2008; 18: 849–859.
[235]
Blessing EM, Steenkamp MM, Manzanares J, Marmar CR. Cannabidiol as a Potential Treatment for Anxiety Disorders. Neurotherapeutics. 2015; 12: 825–836.
[236]
Papagianni EP, Stevenson CW. Cannabinoid Regulation of Fear and Anxiety: an Update. Current Psychiatry Reports. 2019; 21: 38.
[237]
Campos AC, Guimarães FS. Evidence for a potential role for TRPV1 receptors in the dorsolateral periaqueductal gray in the attenuation of the anxiolytic effects of cannabinoids. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2009; 33: 1517–1521.
[238]
Zuardi AW, Cosme RA, Graeff FG, Guimarães FS. Effects of ipsapirone and cannabidiol on human experimental anxiety. Journal of Psychopharmacology. 1993; 7: 82–88.
[239]
Das RK, Kamboj SK, Ramadas M, Yogan K, Gupta V, Redman E, et al. Cannabidiol enhances consolidation of explicit fear extinction in humans. Psychopharmacology. 2013; 226: 781–792.
[240]
Schier ARDM, Ribeiro NPDO, e Silva ACDO, Hallak JEC, Crippa JAS, Nardi AE, et al. Cannabidiol, a Cannabis sativa constituent, as an anxiolytic drug. Revista Brasileira De Psiquiatria. 2012; 34: S104–S117.
[241]
Bergamaschi MM, Queiroz RHC, Chagas MHN, de Oliveira DCG, De Martinis BS, Kapczinski F, et al. Cannabidiol Reduces the Anxiety Induced by Simulated Public Speaking in Treatment-Naïve Social Phobia Patients. Neuropsychopharmacology. 2011; 36: 1219–1226.
[242]
Crippa JAS, Derenusson GN, Ferrari TB, Wichert-Ana L, Duran FL, Martin-Santos R, et al. Neural basis of anxiolytic effects of cannabidiol (CBD) in generalized social anxiety disorder: a preliminary report. Journal of Psychopharmacology. 2011; 25: 121–130.
[243]
Shannon S, Lewis N, Lee H, Hughes S. Cannabidiol in anxiety and sleep: a large case series. The Permanente Journal. 2019; 23: 18–04.
[244]
Elms L, Shannon S, Hughes S, Lewis N. Cannabidiol in the Treatment of Post-Traumatic Stress Disorder: a Case Series. The Journal of Alternative and Complementary Medicine. 2019; 25: 392–397.
[245]
Shannon S, Opila-Lehman J. Effectiveness of cannabidiol oil for pediatric anxiety and insomnia as part of posttraumatic stress disorder: a case report. The Permanente Journal. 2016; 20: 16.
[246]
Wright M, Di Ciano P, Brands B. Use of Cannabidiol for the Treatment of Anxiety: a Short Synthesis of Pre-Clinical and Clinical Evidence. Cannabis and Cannabinoid Research. 2020; 5: 191–196.
[247]
Reynolds AM, Malow BA. Sleep and Autism Spectrum Disorders. Pediatric Clinics of North America. 2011; 58: 685–698.
[248]
Herrmann S. Counting Sheep: Sleep Disorders in Children with Autism Spectrum Disorders. Journal of Pediatric Health Care. 2016; 30: 143–154.
[249]
Kozlowski AM, Matson JL, Belva B, Rieske R. Feeding and sleep difficulties in toddlers with autism spectrum disorders. Research in Autism Spectrum Disorders. 2012; 6: 385–390.
[250]
Tsai F, Chiang H, Lee C, Gau SS, Lee W, Fan P, et al. Sleep problems in children with autism, attention-deficit hyperactivity disorder, and epilepsy. Research in Autism Spectrum Disorders. 2012; 6: 413–421.
[251]
Abel E, Kim SY, Kellerman AM, Brodhead MT. Recommendations for Identifying Sleep Problems and Treatment Resources for Children with Autism Spectrum Disorder. Behavior Analysis in Practice. 2016; 10: 261–269.
[252]
Vriend JL, Corkum PV, Moon EC, Smith IM. Behavioral Interventions for Sleep Problems in Children with Autism Spectrum Disorders: Current Findings and Future Directions. Journal of Pediatric Psychology. 2011; 36: 1017–1029.
[253]
Russo E, Guy G, Robson P. Cannabis, pain and sleep: lessons from therapeutic clinical trials of Sativex a cannabis-based medicine. Chemistry & Biodiversity. 2007; 4: 1729–1743.
[254]
Gagnon K, Godbout R. Melatonin and Comorbidities in Children with Autism Spectrum Disorder. Current Developmental Disorders Reports. 2018; 5: 197–206.
[255]
Carmassi C, Palagini L, Caruso D, Masci I, Nobili L, Vita A, et al. Systematic review of sleep disturbances and circadian sleep desynchronization in autism spectrum disorder: Toward an integrative model of a self-reinforcing loop. Frontiers in Psychiatry. 2019; 10: 366.
[256]
Malow BA, Findling RL, Schroder CM, Maras A, Breddy J, Nir T, et al. Sleep, Growth, and Puberty after 2 Years of Prolonged-Release Melatonin in Children with Autism Spectrum Disorder. Journal of the American Academy of Child & Adolescent Psychiatry. 2021; 60: 252–261.e3.
[257]
Braam W, Smits MG, Didden R, Korzilius H. Exogenous melatonin for sleep problems in individuals with intellectual disability: A meta-analysis. Developmental Medicine and Child Neurology. 2009; 51: 340–349.
[258]
Pickens J. Sedative Activity of Cannabis in Relation to its delta trans THC and cannabidiol content. British Journal of Pharmacology. 1981; 72: 649–656.
[259]
Tringale R, Jensen C. Cannabis and Insomnia. O’Shaughnessy’s. Autumn. 2011; 5E9EC245-448E-17B2-C7CA-21C6BDC6852D.pdf (webydo.com) (Accessed: December 14, 2021)
[260]
Pivik RT, Zarcone V, Dement WC, Hollister LE. Delta-9-tetrahydrocannabinol and synhexl: Effects on human sleep patterns. Clinical Pharmacology & Therapeutics. 1972; 13: 426–435.
[261]
Hosko MJ, Kochar MS, Wang RIH. Effects of orally administered delta-9-tetrahydrocannabinol in man. Clinical Pharmacology & Therapeutics. 1973; 14: 344–352.
[262]
Feinberg I, Jones R, Walker JM, Cavness C, March J. Effects of high dosage delta-9-tetrahydrocannabinol on sleep patterns in man. Clinical Pharmacology & Therapeutics. 1975; 17: 458–466.
[263]
Feinberg I, Jones R, Walker J, Cavness C, Floyd T. Effects of marijuana extract and tetrahydrocannabinol on electroencephalographic sleep patterns. Clinical Pharmacology & Therapeutics. 1976; 19: 782–794.
[264]
Nicholson AN, Turner C, Stone BM, Robson PJ. Effect of delta 9 THC and Cannabidiol on Nocturnal Sleep and Early-morning Behavior in Young Adults. Journal of Clinical Psychopharmacology. 2004; 24: 305–313
[265]
Gorelick DA, Goodwin RS, Schwilke E, Schroeder JR, Schwope DM, Kelly DL, et al. Around-the-clock oral THC effects on sleep in male chronic daily cannabis smokers. The American Journal on Addictions. 2013; 22: 510–514.
[266]
Murillo-Rodríguez E, Millán-Aldaco D, Palomero-Rivero M, Mechoulam R, Drucker-Colín R. The nonpsychoactive Cannabis constituent cannabidiol is a wake-inducing agent. Behavioral Neuroscience. 2008; 122: 1378–1382
[267]
Murillo-Rodríguez E, Millán-Aldaco D, Palomero-Rivero M, Mechoulam R, Drucker-Colín R. Cannabidiol, a constituent ofCannabis sativa, modulates sleep in rats. FEBS Letters. 2006; 580: 4337–4345.
[268]
Murillo-Rodríguez E, Palomero-Rivero M, Millán-Aldaco D, Mechoulam R, Drucker-Colín R. Effects on sleep and dopamine levels of microdialysis perfusion of cannabidiol into the lateral hypothalamus of rats. Life Sciences. 2011; 88: 504–511.
[269]
Murillo-Rodríguez E, Vázquez E, Millán-Aldaco D, Palomero-Rivero M, Drucker-Colin R. Effects of the fatty acid amide hydrolase inhibitor URB597 on the sleep-wake cycle, c-Fos expression and dopamine levels of the rat. European Journal of Pharmacology. 2007; 562: 82–91.
[270]
Chagas MHN, Eckeli AL, Zuardi AW, Pena-Pereira MA, Sobreira-Neto MA, Sobreira ET, et al. Cannabidiol can improve complex sleep-related behaviours associated with rapid eye movement sleep behaviour disorder in Parkinson’s disease patients: a case series. Journal of Clinical Pharmacy and Therapeutics. 2014; 39: 564–566.
[271]
Murillo-Rodriguez E, Sarro-Ramirez A, Sanchez D, Mijangos-Moreno S, Tejeda-Padron A, Poot-Ake A, et al. Potential Effects of Cannabidiol as a Wake-Promoting Agent. Current Neuropharmacology. 2014; 12: 269–272.
[272]
Fusar-Poli L, Cavone V, Tinacci S, Concas I, Petralia A, Signorelli MS, et al. Cannabinoids for people with asd: A systematic review of published and ongoing studies. Brain Sciences. 2020; 10: 1–18.
[273]
Fleury-Teixeira P, Caixeta FV, Ramires da Silva LC, Brasil-Neto JP, Malcher-Lopes R. Effects of CBD-Enriched Cannabis sativa Extract on Autism Spectrum Disorder Symptoms: An Observational Study of 18 Participants Undergoing Compassionate Use. Frontiers in Neurology. 2019; 10: 1145.
[274]
Aran A, Cayam-Rand D. Medical cannabis in children. Rambam Maimonides Medical Journal. 2020; 11: 1–10.
[275]
Gordon-Lipkin E, Marvin AR, Law JK, Lipkin PH. Anxiety and Mood Disorder in Children with Autism Spectrum Disorder and ADHD. Pediatrics. 2018; 141: e20171377.
[276]
Dawson G, Rosanoff M. Autism and Bipolar Disorder. Autism and Bipolar Disorder. Springer: New York, NY. 2021.
[277]
Hudson CC, Hall L, Harkness KL. Prevalence of Depressive Disorders in Individuals with Autism Spectrum Disorder: a Meta-Analysis. Journal of Abnormal Child Psychology. 2019; 47: 165–175.
[278]
Ferber SG, Namdar D, Hen-Shoval D, Eger G, Koltai H, Shoval G, et al. The “Entourage Effect”: Terpenes Coupled with Cannabinoids for the Treatment of Mood Disorders and Anxiety Disorders. Current Neuropharmacology. 2020; 18: 87–96.
[279]
Shoval G, Shbiro L, Hershkovitz L, Hazut N, Zalsman G, Mechoulam R, et al. Prohedonic Effect of Cannabidiol in a Rat Model of Depression. Neuropsychobiology. 2016; 73: 123–129.
[280]
Zanelati T, Biojone C, Moreira F, Guimarães F, Joca S. Antidepressant-like effects of cannabidiol in mice: possible involvement of 5-HT1A receptors. British Journal of Pharmacology. 2010; 159: 122–128.
[281]
Iseger TA, Bossong MG. A systematic review of the antipsychotic properties of cannabidiol in humans. Schizophrenia Research. 2015; 162: 153–161.
[282]
Abrams DI. The therapeutic effects of Cannabis and cannabinoids: an update from the National Academies of Sciences, Engineering and Medicine report. European Journal of Internal Medicine. 2018; 49: 7–11.
[283]
Sarris J, Sinclair J, Karamacoska D, Davidson M, Firth J. Medicinal cannabis for psychiatric disorders: a clinically-focused systematic review. BMC Psychiatry. 2020; 20: 24.
[284]
Soares VP, Campos AC. Evidences for the Anti-panic Actions of Cannabidiol. Current Neuropharmacology. 2017; 15: 291–299.
[285]
Cuttler C, Spradlin A, McLaughlin RJ. A naturalistic examination of the perceived effects of cannabis on negative affect. Journal of Affective Disorders. 2018; 235: 198–205.
[286]
Womack SR, Shaw DS, Weaver CM, Forbes EE. Bidirectional Associations between Cannabis Use and Depressive Symptoms from Adolescence through Early Adulthood among at-Risk Young Men. Journal of Studies on Alcohol and Drugs. 2016; 77: 287–297.
[287]
Szoke A, Galliot A, Richard J, Ferchiou A, Baudin G, Leboyer M, et al. Association between cannabis use and schizotypal dimensions – a meta-analysis of cross-sectional studies. Psychiatry Research. 2014; 219: 58–66.
[288]
Degenhardt L, Ferrari AJ, Calabria B, Hall WD, Norman RE, McGrath J, et al. The global epidemiology and contribution of cannabis use and dependence to the global burden of disease: results from the GBD 2010 study. PLoS ONE. 2013; 8: e76635.
[289]
van Laar M, van Dorsselaer S, Monshouwer K, de Graaf R. Does cannabis use predict the first incidence of mood and anxiety disorders in the adult population? Addiction. 2007; 102: 1251–1260.
[290]
Degenhardt L, Coffey C, Romaniuk H, Swift W, Carlin JB, Hall WD, et al. The persistence of the association between adolescent cannabis use and common mental disorders into young adulthood. Addiction. 2013; 108: 124–133.
[291]
Patton GC, Coffey C, Carlin JB, Sawyer SM, Lynskey M. Reverse gateways? Frequent cannabis use as a predictor of tobacco initiation and nicotine dependence. Addiction. 2005; 100: 1518–1525.
[292]
Fergusson DM, Horwood LJ, Swain-Campbell NR. Cannabis dependence and psychotic symptoms in young people. Psychological Medicine. 2003; 33: 15–21.
[293]
van Os J. Cannabis Use and Psychosis: a Longitudinal Population-based Study. American Journal of Epidemiology. 2002; 156: 319–327.
[294]
Blanco C, Hasin DS, Wall MM, Flórez-Salamanca L, Hoertel N, Wang S, et al. Cannabis Use and Risk of Psychiatric Disorders: Prospective Evidence From a US National Longitudinal Study. JAMA Psychiatry. 2016; 73: 388.
[295]
Harder VS, Stuart EA, Anthony JC. Adolescent cannabis problems and young adult depression: male-female stratified propensity score analyses. The American Journal of Epidemiology. 2008; 168: 592–601.
[296]
Feingold D, Weiser M, Rehm J, Lev-Ran S. The association between cannabis use and mood disorders: a longitudinal study. Journal of Affective Disorders. 2015; 172: 211–218.
[297]
Rasic D, Weerasinghe S, Asbridge M, Langille DB. Longitudinal associations of cannabis and illicit drug use with depression, suicidal ideation and suicidal attempts among Nova Scotia high school students. Drug and Alcohol Dependence. 2013; 129: 49–53.
[298]
Patton GC. Cannabis use and mental health in young people: cohort study. British Medical Journal. 2002; 325: 1195–1198.
[299]
Coffey C, Carlin JB, Degenhardt L, Lynskey M, Sanci L, Patton GC. Cannabis dependence in young adults: an Australian population study. Addiction. 2002; 97: 187–194.
[300]
Bolla KI, Brown K, Eldreth D, Tate K, Cadet JL. Dose-related neurocognitive effects of marijuana use. Neurology. 2002; 59: 1337–1343.
[301]
Degenhardt L, Hall W. Is Cannabis Use a Contributory Cause of Psychosis? The Canadian Journal of Psychiatry. 2006; 51: 556–565.
[302]
Lucatch AM, Coles AS, Hill KP, George TP. Cannabis and Mood Disorders. Current Addiction Reports. 2018; 5: 336–345.
[303]
Hall W, Degenhardt L. The adverse health effects of chronic cannabis use. Drug Testing and Analysis. 2014; 6: 39–45.
[304]
Noel C. Evidence for the use of “medical marijuana” in psychiatric and neurologic disorders. Mental Health Clinician. 2017; 7: 29–38.
[305]
Kleber HD, Dupont RL. Physicians and Medical Marijuana. American Journal of Psychiatry. 2012; 169: 564–568.
[306]
Volkow ND, Baler RD, Compton WM, Weiss SRB. Adverse Health Effects of Marijuana Use. New England Journal of Medicine. 2014; 370: 2219–2227.
[307]
Hill KP. Medical Marijuana for Treatment of Chronic Pain and other Medical and Psychiatric Problems. The Journal of the American Medical Association. 2015; 313: 2474.
[308]
Botsford SL, Yang S, George TP. Cannabis and Cannabinoids in Mood and Anxiety Disorders: Impact on Illness Onset and Course, and Assessment of Therapeutic Potential. the American Journal on Addictions. 2020; 29: 9–26.
[309]
Luo X, Reiter MA, d’Espaux L, Wong J, Denby CM, Lechner A, et al. Complete biosynthesis of cannabinoids and their unnatural analogues in yeast. Nature. 2019; 567: 123–126.
[310]
van Bakel H, Stout JM, Cote AG, Tallon CM, Sharpe AG, Hughes TR, et al. The draft genome and transcriptome of Cannabis sativa. Genome Biology. 2011; 12: R102.
[311]
Pollack JR, Perou CM, Alizadeh AA, Eisen MB, Pergamenschikov A, Williams CF, et al. Genome-wide analysis of DNA copy-number changes using cDNA microarrays. Nature Genetics. 1999; 23: 41–46.
[312]
Kovalchuk I, Pellino M, Rigault P, van Velzen R, Ebersbach J, Ashnest JR, et al. The Genomics of Cannabis and its Close Relatives. Annual Review of Plant Biology. 2020; 71: 713–739.
[313]
Niazian M. Application of genetics and biotechnology for improving medicinal plants. Planta. 2019; 249: 953–973.
[314]
Jensen HM, Korbut R, Kania PW, Buchmann K. Cannabidiol effects on behavior and immune gene expression in zebrafish (Danio rerio). PLoS ONE. 2018; 13: e0200016.
Share
Back to top