You are on page 1of 22

Psychopharmacology (2022) 239:2713–2734

https://doi.org/10.1007/s00213-022-06196-4

REVIEW

Cannabidiol for the treatment of autism spectrum disorder: hope


or hype?
João F. C. Pedrazzi1 · Frederico R. Ferreira2 · Danyelle Silva‑Amaral3 · Daniel A. Lima1 · Jaime E. C. Hallak1 ·
Antônio W. Zuardi1 · Elaine A. Del‑Bel1,4 · Francisco S. Guimarães5 · Karla C. M. Costa5 · Alline C. Campos5 ·
Ana C. S. Crippa6 · José A. S. Crippa1

Received: 15 November 2021 / Accepted: 18 July 2022 / Published online: 29 July 2022
© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2022

Abstract
Rationale Autism spectrum disorder (ASD) is defined as a group of neurodevelopmental disorders whose symptoms include
impaired communication and social interaction, restricted and repetitive patterns of behavior, and varying levels of intellec‑
tual disability. ASD is observed in early childhood and is one of the most severe chronic childhood disorders in prevalence,
morbidity, and impact on society. It is usually accompanied by attention deficit hyperactivity disorder, anxiety, depression,
sleep disorders, and epilepsy. The treatment of ASD has low efficacy, possibly because it has a heterogeneous nature, and its
neurobiological basis is not clearly understood. Drugs such as risperidone and aripiprazole are the only two drugs available
that are recognized by the Food and Drug Administration, primarily for treating the behavioral symptoms of this disorder.
These drugs have limited efficacy and a high potential for inducing undesirable effects, compromising treatment adherence.
Therefore, there is great interest in exploring the endocannabinoid system, which modulates the activity of other neurotrans‑
mitters, has actions in social behavior and seems to be altered in patients with ASD. Thus, cannabidiol (CBD) emerges as a
possible strategy for treating ASD symptoms since it has relevant pharmacological actions on the endocannabinoid system
and shows promising results in studies related to disorders in the central nervous system.
Objectives Review the preclinical and clinical data supporting CBD’s potential as a treatment for the symptoms and comor‑
bidities associated with ASD, as well as discuss and provide information with the purpose of not trivializing the use of this
drug.

Keywords Cannabidiol · Autism spectrum disorder · Treatment autism spectrum disorder · Endocannabinoid system ·
Pharmacological targets · Animal models · Clinical trials · Medical Cannabis

Introduction and stereotyped, repetitive, and restrictive behavior pat‑


terns (American Psychiatric Association 2013). Although
Autism spectrum disorder (ASD) is a set of heterogene‑ defined by these significant symptoms, the phenotype in
ous disorders of early neurodevelopment characterized ASD patients may vary, ranging from individuals with
by impaired social interaction, deficits in communication, severe intellectual disability and poor performance in

4
* João F. C. Pedrazzi Department of Morphology, Physiology, and Basic
joaofranciscopedrazzi@usp.br Pathology, School of Dentistry of Ribeirão Preto, University
of São Paulo, Ribeirão Preto, São Paulo, Brazil
1
Department of Neurosciences and Behavioral Sciences, 5
Department of Pharmacology, School of Medicine
School of Medicine of Ribeirão Preto, University of São
of Ribeirão Preto, University of São Paulo,
Paulo, Ribeirão Preto, São Paulo, Brazil
Ribeirão Preto, São Paulo, Brazil
2
Oswaldo Cruz Institute, Oswaldo Cruz Foundation, 6
Graduate Program in Child and Adolescent Health,
Rio de Janeiro 21040‑900, Brazil
Neuropediatric Center of the Hospital of Clinics (CENEP),
3
Department of Physiology, School of Medicine of Ribeirão Federal University of Paraná, Curitiba, Paraná, Brazil
Preto, University of São Paulo, Ribeirão Preto, São Paulo,
Brazil

13
Vol.:(0123456789)
2714 Psychopharmacology (2022) 239:2713–2734

adaptive behavioral skills to individuals with normal intel‑ effects must be balanced against the common adverse effects
ligence quotient (IQ) who lead independent lives. Patients and low compliance caused by these drugs. It is essential
with this disorder may also have other comorbidities that to emphasize that early intervention could improve the
can cause a delay in diagnosis (Lai et al. 2014), such as clinical outcome of children with ASD (Warren et al. 2012;
hyperactivity, gastrointestinal and sleep disorders, epilepsy, Zwaigenbaum et al. 2015). Since there is an increase in
psychosis, aggressiveness, self-injury, and social anxiety prevalence and high economic and social costs of ASD,
(South and Rodgers 2017). The emergence of ASD symp‑ new pharmacological approaches and molecular targets are
toms appears in the first three years of life, with a higher needed to elucidate and optimize the treatment of ASD,
prevalence in males (4:1) (Brugha et al. 2011; Levy et al. which is often limited. Consequently, there is an increasing
2011). In a recent study conducted in the USA based on interest in investigating CBD for the vast symptomatology
the 5th edition of the Diagnostic and Statistical Manual of of ASD since this compound may present a therapeutic pro‑
Mental Disorders (DSM-5), among 8-year-old children, 1 file for psychiatric conditions that are associated with this
in 59 was shown to have ASD (Baio et al. 2018). Therefore, disorder (for review, see Poleg et al. 2019). Additionally,
the incidence of ASD has significantly increased worldwide, the probable involvement of the endocannabinoid system
ranking third among developmental disorders, ahead of con‑ in the pathophysiology of ASD is the subject of extensive
genital malformations and Down syndrome (Volkmar et al. investigation. Consistent results from endocannabinoid dys‑
2004; Schroeder et al. 2010). Several central nervous sys‑ function in animal models of ASD and clinical studies have
tem (CNS) areas have been associated with ASD neurobiol‑ been shown (Qin et al. 2015; Servadio et al. 2016; Melancia
ogy, including the hippocampus, prefrontal cortex, ventral et al. 2018). Moreover, retrospective and uncontrolled trials
striatum, amygdala, and cerebellum (Bugalho et al. 2006; of CBD- enriched Cannabis in subjects under this condition
Rossignol and Bradstreet 2008; Luna et al. 2015). Brain have been published (Aran et al. 2019a, b; Kuester et al.
structures involved in ASD tend to develop more slowly 2017; Mostafavi and Gaitanis 2020). Despite promising
(Ashwood et al. 2006). Although several genetic and envi‑ results from the use of CBD in patients with ASD, there are
ronmental factors have been identified and associated with few discussions regarding its interactions with other drugs,
this disorder, the etiology of ASD remains unknown (Bölte the side effects and the selection of patients who could ben‑
et al. 2019). Over the past few years, the understanding of efit from its use.
the genetic changes, including de novo mutations, in ASD Meanwhile, anecdotal evidence emerges concomitantly
has expanded (Ramaswami and Geschwind 2018). Several with the search for CBD formulations, which often do not
genes associated with ASD susceptibility have been identi‑ undergo scientific rigor. Given the complexity of ASD, the
fied in approximately 10–20% of the cases (Brugha et al. urgency of developing effective and safe treatments is evi‑
2011). dent while simultaneously conducting responsible and criti‑
Furthermore, epigenetic mechanisms and specific gene- cal approaches to drugs that can have therapeutic effects, as
environment interactions are likely involved in the etiology is the case with CBD.
of ASD (Lai et al. 2014). Environmental influences include The current review will present preclinical and clinical
pathophysiological changes in the CNS caused by oxidative studies that motivate the use of CBD and how the modula‑
stress, neuroinflammation, mitochondrial dysfunction, and tion of the endocannabinoid system can benefit the range
biochemical disorders (Herbert 2010). Clinical and experi‑ of symptoms present in ASD and adequately discuss the
mental evidence indicates that prenatal bacterial and viral challenges of its use from the perspective of its intense
infections, such as rubella and cytomegalovirus, interfere dissemination.
in the intrauterine environment (Yamashita et al. 2003;
Libbey et al. 2005; Atladóttir et al. 2010). These factors The endocannabinoid system and its involvement
may promote behavioral abnormalities with similarities to in the pathophysiology of ASD
autism, including inadequate activation of proinflammatory
cytokines and abnormal immune response (Dalton et al. The Cannabis plant contains many chemical compounds,
2003; Kirsten et al. 2010). In several studies, the immune including flavonoids, terpenes, fatty acids, and phytocan‑
system was associated with neuropsychiatric disorders (for nabinoids. There are more than 100 phytocannabinoids in
review, see Nutma et al. 2019). Cannabis, including delta-9-tetrahydrocannabinol (THC),
Specific compounds to treat the core symptoms of ASD the main psychotomimetic constituent, and CBD, which
or its progression are not yet available. Several conventional lacks the subjective effects caused by THC (for review,
drugs, such as atypical antipsychotics, anxiolytics, stimu‑ see Carlini 2010). In the early 1990s, the discovery of
lants, and selective serotonin reuptake inhibitors (SSRIs), the cannabinoid receptors type 1 (CB1) and type 2 (CB2)
have been used to alleviate some symptoms and commonly significantly advanced the understanding of the effects of
related comorbidities of ASD. These non-specific beneficial chemical compounds in Cannabis (Matsuda et al. 1990;

13
Psychopharmacology (2022) 239:2713–2734 2715

Kaminski et al. 1992; Munro et al. 1993). THC is a partial of lipids, have properties similar to endocannabinoids, act‑
agonist of CB1 receptors. The activation of these receptors ing in many cases on CB1 and CB2 receptors in addition
plays a critical role in the acute anxiogenic and psychotic to GPR55, TRPV1, and PPARs (Di Marzo 2020; Cristino
experiences associated with the use of Cannabis (Huestis et al. 2020). The number of receptors, enzymes, proteins, ion
et al. 2001). The receptors are mainly located presynapti‑ channels, and transporters involved in the spectrum of the
cally, with high densities in the cerebral cortex, olfactory endocannabinoid system has been updated and increased,
bulb, hippocampus, lateral caudate-putamen, substantia including a more expanded system termed the endocan‑
nigra, globus pallidus, and cerebellum. In contrast, a low nabinoidome. Due to their wide distribution in the CNS,
density of these receptors is observed in the thalamus and the endocannabinoidome has been suggested to participate
peripheral organs and tissue (Pertwee, 2008; Russo et al. in the modulation of various brain functions, including
2005; Munro et al. 1993). The CB1 receptors are located in memory and learning, executive functions, and motivational
areas associated with motor control, learning and memory, processes relevant to neurodevelopmental disorders such as
emotional response and cognitive functions, and structures ASD (Di Marzo and Maccarrone 2008).
related to ASD neurobiology (Zamberletti et al. 2017). The More than 100 genes and genomic regions are currently
CB2 receptor is mainly expressed in the immune system. associated with autism. The earliest successes in gene discov‑
However, the receptor is also present in microglia and some ery revealed important general properties, such as most pro‑
postsynaptic locations in the CNS (Sagar et al. 2009). CB2 teins encoded by autism risk genes participate in either synap‑
receptors may also be expressed in neural cells involved in tic structure and function or chromatin modification and
the perception/modulation of pain (Zhang et al. 2003; Elmes regulation of gene expression. Furthermore, in a large-scale
et al. 2004; Clayton et al. 2002; Nackley et al. 2003). CB1 exome sequencing study, both developmental and functional
and CB2 receptors are coupled to inhibitory protein G that, changes were implicated in the neurobiology of autism (Sat‑
when activated, inhibits the enzyme adenylate cyclase, lead‑ terstrom et al. 2019). Rare genetic variants have been found in
ing to a decrease in cyclic AMP levels and inhibition of genes closely linked to the endocannabinoid system, such as
calcium channels (Breivogel et al. 1997; Piomelli, 2003). cannabinoid receptor 1 (CNR1), CNR2, GPR55, diacylglyc‑
The activation of CB1 receptors inhibits the release of erol lipase alpha (DAGLA), MGL, and FAAH (Smith et al.
other neurotransmitters, such as GABA and glutamate. The 2017). In that study, molecular trials were conducted on 6032
main endogenous agonists of CB1 and CB2 receptors are patients with a broad spectrum of neurological disorders and
derived from arachidonic acid and, similar to the phytocan‑ rare genetic variants observed. The variants were evaluated for
nabinoids in Cannabis sativa, are lipid. The most studied association with phenotypes similar to those observed in the
endocannabinoids are 2-arachidonoyl glycerol (2-AG) and knockout orthologous genes in mice. Rare heterozygous cod‑
anandamide (arachidonoyl ethanolamine, ananda, from the ing variants in CNR1, which encode CB1 receptors, are sig‑
Sanskrit meaning “joy, bliss, delight.” (Devane et al. 1992; nificantly associated with sensitivity to pain (especially
Mechoulam et al. 1995; Sugiura et al. 1995). Unlike classic migraine), sleep, and memory disorders alone or in combina‑
neurotransmitters, endocannabinoids are not synthesized at tion with anxiety when compared with a set of controls without
presynaptic terminals or stored in vesicles but are formed these variants in the CNR1 gene. Similarly, rare heterozygous
based on demand at postsynaptic terminals (Devane 1994; variants in DAGLA, which encode DAGLA, have been found
Iversen 1994). After calcium influx, activation of phospho‑ significantly associated with seizures and neurodevelopmental
lipases converts the phospholipids into endocannabinoids disorders, including ASD and abnormalities of brain morphol‑
(phospholipase D in the case of anandamide and diacylg‑ ogy, compared with healthy controls. As mentioned earlier,
lycerol lipase in the case of 2-AG). These substances act DAGLA synthesizes the endocannabinoid 2-AG in the brain,
on presynaptic CB1 receptors inhibiting neurotransmitter interacting with CB1 receptors (Howlett et al. 2010). The phe‑
release (Beltramo et al. 1997, Egertová et al. 1998, Piomelli notypes associated with rare variants of CNR1 are similar to
2003). The availability of endocannabinoids is regulated by the phenotypes thought to be involved in the clinical syndrome
enzymatic uptake/degradation. The fatty acid amide hydro‑ of endocannabinoid deficiency. The several phenotypes associ‑
lase (FAAH) and monoacylglycerol lipase (MGL) enzymes ated with rare variants of DAGLA emphasize the critical role
efficiently hydrolyze anandamide and 2-AG, respectively of rapid synthesis of 2-AG and the endocannabinoid system in
(Cravatt et al. 1996; Dinh et al. 2002). The concept of the regulating neurological function and development, events
endocannabinoid system has been recently revised to include associated with the neurobiology of ASD (Smith et al. 2017).
other receptors that have an affinity for anandamide, such as It is worth mentioning that genetic variations associated with
the G protein-coupled receptor 55 (GPR55), the transient the endocannabinoid system may be why ASD patients using
receptor potential vanilloid-1 (TRPV1) cation channel, CBD-enriched Cannabis do not benefit equally from treating
and peroxisome proliferator-activated receptors (PPARs). their symptoms, while others may be more susceptible to side
N-acylethanolamines and N-acyldopamines, another class effects. Indeed, it is suggested these patients may be more

13
2716 Psychopharmacology (2022) 239:2713–2734

prone to the broadside effect profile relative to other patient this behavior, such as the prefrontal cortex, basolateral amyg‑
populations (Mostafavi and Gaitanis 2020). Thus, preclinical dala, hippocampus and ventral striatum (Wei et al. 2017; Man‑
studies, pharmacogenetics, pharmacogenomics, and double- duca et al. 2021). In two widely used mouse models of ASD,
blind, randomized placebo-controlled clinical trials to evaluate the BTBR and fmr1−/− (model of fragile X syndrome), the
gene expression and post-translational mechanisms are critical FAAH blockade by URB597 completely reversed the social
to elucidate this challenge. To date, clinical studies suggest that impairment in mice (Wei et al. 2016). Furthermore, the CB1
CBD-enriched Cannabis may benefit epilepsy, aggression, receptor blockade, by AM251, prevented the prosocial effect
improved mood, anxiety, and social interaction in ASD of FAAH inhibition in BTBR mice, indicating that increasing
patients. These approaches will be discussed throughout the anandamide activity at these receptors improves ASD-related
review. Changes in the functioning of the immune system have social impairment (Qin et al. 2015). 2-AG also seems to play
been associated with the etiology of ASD (Lyall et al. 2014; an important role in the regulation of social reward in rodents,
Siniscalco et al. 2012). In agreement with these findings, based as its selective decrease in the forebrain of male transgenic
on an analysis of mononuclear cells extracted from peripheral mice (MGL-Tg mice) impaired their performance in the
blood, patients with ASD have excessive production of IL-1β, socially induced place conditioning (Wei et al. 2016). Further‑
which can result in long-term immunological changes more, CB1 receptor knockout mice exhibit more aggressive
(Enstrom et al. 2010). As already discussed, CB2 receptors are behaviors, as well as active and passive defensive coping
closely associated with the immune response. In a pioneering behaviors such as avoidance, freezing, and risk assessment,
study, the mononuclear cells in the peripheral blood of patients suggesting these receptors play an essential role in decreasing
with ASD were compared with healthy controls (Siniscalco social stress and consequently in the regulation of social
et al. 2013). The authors reported significantly high mRNA reward (Rodriguez-Arias et al. 2013; Litvin et al. 2013). Dur‑
and protein levels in CB2 receptors in the ASD group, indicat‑ ing development, CB1 receptors drive axon guidance and are
ing modulation of the endocannabinoid system through these associated with synaptogenesis (Harkany et al. 2008; Watson
receptors could be a therapeutic target in the search for drugs et al. 2008; Fride et al. 2009). Children with ASD have abnor‑
that can treat the various symptoms present in ASD (Sinis‑ mal brain connectivity, which could be due to a lack of CB1
calco et al. 2013). Furthermore, reduced concentration of the axon guidance (McFadden and Minshew, 2013). In addition,
endocannabinoid anandamide (Karhson et al. 2018; Aran et al. reduced expression of CB1 receptors was found in the post‑
2019a, b) and lower levels of oleoyl ethanolamide and palmi‑ mortem brains of individuals with ASD (Purcell et al. 2001).
toylethanolamide (Aran et al. 2019a, b) have been described In preclinical models of ASD induced by valproic acid (VPA),
in ASD patients. In addition, reduced plasma anandamide lev‑ CB1 receptors displayed altered phosphorylation in different
els (Karhson et al. 2018) and associations between polymor‑ brain areas associated with anandamide metabolism changes
phisms in the gene encoding CB1 receptor and emotional in a sex-dependent manner (Servadio et al. 2016; Melancia
processing (Chakrabarti et al. 2006; 2011; Domschke et al. et al. 2018). In addition, the increased anandamide caused by
2008) have been reported in children with ASD. A better inhibition of its degradation enzyme (FAAH) with the com‑
understanding of neurochemical mechanisms that mediate pound URB597 corrected social and emotional impairments
emotional processing like the social reward is essential for induced by VPA exposure during the neurodevelopmental
developing novel treatments, especially for disorders in which period in both studies. Furthermore, in a recent study, the
these processes are impaired. Social reward applies to a variety in vivo optogenetics technique showed that activating the baso‑
of positive stimuli and experiences. Humans and animals have lateral amygdala-nucleus accumbens (BLA-NAc) glutamater‑
a strong motivation to seek social relationships. The need to gic circuit reduced interaction and increased social avoidance
be part of a group causes us to act prosocially, evidencing the in mice (Folkes et al. 2020). The increased endocannabinoid
satisfactory nature of sociability (Rademacher et al. 2017). 2-AG signaling caused by inhibiting its degradation enzyme
These events are critical to maintaining health and survival. (MGL) with the compound JZL184 reduced the glutamatergic
Several approaches suggest that adequate social stimuli early activity of the BLA-NAc. It prevented the impairment in social
in life are fundamental for developing socio-emotional and interaction associated with the stimulation of this pathway
cognitive skills, while adverse social experiences negatively ­ hank3B−/− mice, an ASD model
in vivo. In addition, in the S
affect neurodevelopment and behavior, culminating in which displays significant impairment in social interaction, the
increased susceptibility to neuropsychiatric conditions (Man‑ optogenetic inhibition of the BLA-NAc circuit markedly
duca et al. 2021). It is proposed that impairments in social increased social interaction in the mice. Thus, systemic injec‑
reward in patients with ASD may result from early deficits in tion of JZL184 or direct microinjection into the NAc also nor‑
social motivation (Chevallier et al. 2012). There is evidence ­ hank3B−/− mice.
malized deficits in the social interaction of S
that endocannabinoid neurotransmission has a critical role in However, ex vivo administration of JZL184 corrected aberrant
mediating social reward in different animal groups. Moreover, excitatory and inhibitory neurotransmission in the NAc and
CB1 receptors are widely expressed in areas implicated with reduced feed-forward inhibition induced by BLA-NAc in NAc

13
Psychopharmacology (2022) 239:2713–2734 2717

neurons in Shank3B −/− mice. These data reveal neuromodula‑ Bhattacharyya et al. 2010; Englund et al. 2013). This
tory mechanisms at the level of neuronal circuitry, which regu‑ hypothesis was further investigated in several laboratory
lates the social function relevant to ASD, indicating that animal and clinical studies. More than 65 potential pharma‑
increased signaling by 2-AG can reduce social deficits by cological targets associated with several promising thera‑
modulating excitatory and inhibitory neurotransmission in the peutic applications of CBD are described. Unlike THC,
NAc (Folkes et al. 2020). Evidence also shows the endocan‑ in vitro assay results indicated CBD has a low affinity or can
nabinoid system interacts with oxytocin-mediated social even function as an antagonist or inverse agonist of CB1
reward. Oxytocin mobilizes anandamide in the NAc, which receptors (Thomas et al. 2007; Laprairie et al. 2015). How‑
may be associated with social impairment in ASD (Wei et al. ever, CBD can also block the metabolism and/or uptake of
2015). The involvement of oxytocin in the pathophysiology of anandamide (Bisogno et al. 2001; Schubart et al. 2014). This
ASD has been previously investigated (Young and Barrett last action is the primary effect of CBD on the CB1- or CB2-
2015). Intranasal administration of the ‘‘prosocial” neuropep‑ mediated neurotransmission complex. CBD does not cause
tide oxytocin was shown to have potential therapeutic effects the cannabinoid tetrad (hypolocomotion, catalepsy, hypo‑
on ASD (Anagnostou et al. 2012; Bernaerts et al. 2020). Fur‑ thermia, and antinociception) that is considered a good pre‑
thermore, the involvement of oxytocin in the pathophysiology dictor of THC-like pharmacological activity, thus confirm‑
of ASD has been confirmed (Young and Barrett 2015). Oxy‑ ing that CBD lacks direct CB1 agonist properties (Martin
tocin is produced in the paraventricular and supraoptic nuclei et al. 1991). However, some of the CBD effects (e.g.,
of the hypothalamus (Swanson and Sawchenko 1983) released increased hippocampal neurogenesis) are prevented by CB1
by magnocellular neurons in the bloodstream and is crucial for or CB2 receptor antagonists, indicating they depend on the
stimulating the contraction of the smooth muscle of the uterus, facilitation of endocannabinoid signaling (Campos et al.
facilitating labor, induction of lactation, and mechanisms 2013; Fogaça et al. 2018). Regarding the endocannabinoi‑
involved with the reproduction and care of offspring, in addi‑ dome, CBD presents pharmacological targets that may be
tion to intimate participation in social and reward behavior relevant to ASD. CBD can activate TRPV1 receptors
(Carter 2014). Notably, in several studies, an interaction (Bisogno et al. 2001), facilitating glutamate release (Musella
between the endocannabinoid system and oxytocin mediating et al. 2010; Ho et al. 2012). This compound can facilitate
social reward has been reported. Oxytocin induces mobiliza‑ 5-HT1A-mediated neurotransmission causing anxiolytic,
tion of anandamide to the NAc, which may be associated the antidepressant, and pro-cognitive effects. Moreover, consid‑
social impairment in ASD (Wei et al. 2015). Endocannabi‑ erable evidence in animal models that address fear memory
noids could modify the inflammatory process associated with processing indicates that CBD can decrease learned fear in
ASD (Knuesel et al. 2014). In the maternal immune activation parameters relevant to the translational study of phobias and
(MIA) model, intense production of the proinflammatory post-traumatic stress disorder (Campos and Guimarães
cytokine interleukin (IL)-17 is associated with abnormal corti‑ 2008; Linge et al. 2016; Russo et al. 2005; Lee et al. 2017).
cal development and social deficits in the offspring occurs In addition, the receptors in glycinergic signaling are altered
(Choi et al. 2016). In a mouse model of immune hypersensitiv‑ in subjects with ASD (Pilorge et al. 2016). Therefore, CBD
ity, anandamide decreased IL-17 production and increased the can directly or indirectly act on spinal glycine receptors,
expression of the anti-inflammatory cytokine IL-10 (Jackson especially in the a1 and a3 subunits, causing an analgesic
et al. 2014). Treatment with the innate immunostimulant effect on inflammatory pain (Lu et al. 2018). Due to its poly‑
lipopolysaccharide (LPS) during adolescence increased anan‑ phenolic structure, CBD displays important antioxidant
damide level and FAAH activity in the amygdala and properties. In addition, CBD has anti-inflammatory effects,
decreased social behavior. These alterations are attenuated by which may be associated with its neuroprotective activities,
the FAAH inhibitor PF-04457845 (Doenni et al. 2016). increasing the therapeutic properties of CBD in ASD patho‑
physiology (Chadwick et al. 2013; Franco and Perucca
CBD structure, pharmacological targets, and studies 2019). Furthermore, CBD blocks the orphan GPR55, a
with therapeutic potential relevant to ASD mechanism associated with its anticonvulsant and antitu‑
moral effects (Morales et al. 2017; Laezza et al. 2020). The
CBD is the main non-psychotomimetic phytocannabinoid GPRs are widely expressed in the brain. In studies with mice
present in the Cannabis plant and was isolated in the early subjected to acute stress, activation of these receptors report‑
1940s by Adams et al. 1940. However, the chemical struc‑ edly produced an anxiolytic and antidepressant effect. How‑
ture was elucidated only in 1963 by Mechoulam and Shvo ever, whether CBD exerts similar actions on GPR55 remains
(1963). The observation that CBD antagonized some of the unclear (Sawzdargo et al. 1999; Shi et al. 2017). The endo‑
main pharmacological effects of THC led to the hypothesis cannabinoid system with phytocannabinoids can modulate
that CBD could have anxiolytic and antipsychotic properties mesocorticolimbic dopaminergic signaling, a pathway asso‑
(Zuardi et al. 1982; 1993; Guimarães et al. 1990; ciated with the emergence of psychosis (Hudson et al. 2018).

13
2718 Psychopharmacology (2022) 239:2713–2734

Previous in vitro binding assay results indicated CBD et al. 2019a, b). CBD administration in rodents prevented
mighty function as a partial agonist of D2-type dopaminer‑ apomorphine-induced stereotypes (Zuardi et al. 1991) and
gic receptors in the rat striatum, reinforcing its antipsychotic amphetamine-induced hyperlocomotion (Moreira and Gui‑
properties shared with clinically used drugs such as aripipra‑ marães 2005; Valvassori et al. 2011). When CBD was
zole (Seeman 2016). Furthermore, in a neurodevelopment directly injected into the NAc, CBD prevented prepulse inhi‑
model in which methylazoxymethanol acetate (MAM) is bition impairment caused by amphetamine (Pedrazzi et al.
used in pregnant rats to generate offspring that mimic symp‑ 2015). In a rodent model of schizophrenia based on chronic
toms of schizophrenia in adulthood, such as cognitive defi‑ blockade of glutamate N-methyl-D-aspartate (NMDA)
cits, dopaminergic dysfunction, and physical and behavioral receptors, CBD increased social interaction, improved rec‑
abnormalities, the action of CBD via partial agonism of D3 ognition of a new object, improved social withdrawal, pre‑
receptors was suggested to improve these disruptions (Stark vented information processing impairment, and decreased
et al. 2020). Finally, CBD could interact with many other glial reactivity (Gururajan et al. 2011; Gomes et al. 2015a,
signaling systems such as the opioid, adenosine, transient b; Silva et al. 2020). In addition, beneficial effects of CBD
channel, and PPARs (Carrier et al. 2006; Linge et al. 2016; have been demonstrated against repetitive behaviors in the
Sonego et al. 2018). PPARs are a family of nuclear receptors marble-burying test (Casarotto et al. 2010; Nardo et al.
widely distributed in various organs and tissues (O’Sullivan 2014) and social isolation-induced aggression in rodents
2016) and may contribute to the pathophysiology of ASD. (Hartmann et al. 2019). Furthermore, CBD could induce
In several studies, PPAR expression and their endogenous rapid and sustained antidepressant-like effects in animal
modulators are downregulated in ASD because the admin‑ models of stress through increased brain-derived neuro‑
istration of these endogenous or synthetic ligands can trophic factor signaling and synaptogenesis (Sales et al.
improve behaviors associated with mood and cognition 2019). Notably, CBD can act on signaling cascades associ‑
(Capano et al. 2018; Mirza and Sharma 2019). As already ated with the mechanistic target of rapamycin (mTOR) in
mentioned, ASD may have a strong neuroinflammatory animal models that may be associated with symptoms pre‑
activity, supporting the hypothesis that CBD actions via sent in ASD (Renard et al. 2016; Sales et al. 2019). It has
PPARs could reduce behaviors associated with the ASD been shown that CBD can contain amphetamine-induced
phenotype, as well as reduce inflammation and oxidative neuronal and behavioral sensitization of the mesolimbic
stress. ASD patients present several clinical manifestations dopamine pathway, controlling downstream phosphorylation
observed in other psychiatric disorders, such as psychosis, of the mTOR/p70S6kinasee signaling pathways directly
anxiety, stereotypical behavior, hyperactivity, aggressive‑ within the NAc shell. Blocking the mTOR pathway with
ness, sleep disorders, epilepsy, allergies, and autoimmune rapamycin can prevent the antidepressant effect of CBD in
and gastrointestinal conditions (for review, see Crippa et al. an animal model of stress. Dysregulation of the mTOR and
2018). The therapeutic potential of CBD in several of these protein kinase B cascade has been identified in multiple dis‑
disorders has been described since the early 1970s. In a pio‑ orders, indicating a potential mechanism associated with
neering study, Carlini and coworkers (1975) demonstrated neurodevelopmental disorders such as ASD (Sahin and Sur
the protective role of CBD derivatives against agents that 2015). The mTOR pathway is a conserved serine/threonine
promote seizures in rodents. Later, Cunha and coworkers kinase in the phosphoinositide-3-kinase (PI3K) family that
(1980) showed this anticonvulsant property in humans. In integrates multiple intracellular and extracellular signals and
normal subjects, CBD reduces the anxiety provoked by high coordinates multiple cellular responses, including protein
doses of delta 9-THC (Zuardi et al. 1982). The anxiolytic synthesis, growth, and proliferation (Winden et al. 2018).
effects of CBD have been described in several preclinical An imbalance may exist between excitatory and inhibi‑
and clinical studies (Guimarães et al. 1990; Batalla et al. tory neurotransmissions in ASD, mainly represented by
2021) and shown associated with regional cerebral blood glutamate and γ-aminobutyric acid (GABA), respectively
flow (rCBF) changes in limbic and paralimbic areas of the (Rubenstein and Merzenich 2003; Gogolla et al. 2009;
brain (Crippa et al. 2011). In individuals with social anxiety Coghlan et al. 2012). The isoforms of glutamic acid decar‑
disorder, CBD reduced anxiety, cognitive impairment, and boxylase (GAD65 and GAD67), a rate-limiting step in con‑
discomfort in a simulated public speaking test (Bergamaschi verting glutamate to GABA, are altered in brain structures
et al. 2011). CBD also showed antipsychotic properties in associated with ASD (Erlander and Tobin, 1991; Fatemi
preclinical and clinical studies. The increased serum anan‑ et al. 2002). In addition, the expression of parvalbumin
damide levels in patients with schizophrenia who used CBD (PV), a calcium-binding protein expressed in a subclass of
correlated with a reduction in psychotic symptoms (Leweke rapidly increasing GABAergic interneurons, which partici‑
et al. 2012), which is encouraging in the context of ASD pates in the regulation of excitatory and inhibitory stimuli in
because, in several aforementioned trials, patients with ASD the brain, is increased after chronic treatment with NMDA
had decreased anandamide levels (Karhson et al. 2018; Aran receptor antagonists in rodents (Abdul-Monim et al. 2007).

13
Psychopharmacology (2022) 239:2713–2734 2719

This finding was confirmed by Gomes et al. (2015a). They an increase in neuroinflammation and the number of acti‑
showed that CBD protects interneurons positively labeled for vated microglial cells (Vargas et al. 2005; Morgan et al. 2010;
PV protein in the medial prefrontal cortex in mice chroni‑ Suzuki et al. 2013). These results indicate that microglia and
cally treated with dizocilpine, also known as MK-801, a neuroinflammation could play a significant role in the patho‑
noncompetitive antagonist of NMDA. Any damage to PV- physiology of ASD (Lenz and Nelson 2018). Also, clinical
positive interneurons results in loss of control of neurons trials have shown increased pro-inflammatory cytokines in
that release the excitatory neurotransmitter glutamate, the brain and peripheral immune cells of autistic patients
with a consequent increase in the activity of neurons that at different ages (Baranova et al. 2021; Toscano et al. 2021;
release dopamine, culminating in psychosis-like conditions Carbone et al. 2021). However, it is still unclear if neuroin‑
(Schwartz et al. 2012). flammation triggers ASD or whether this event is part of it
Furthermore, gestational injection of poly I: C in rodents (Matta et al. 2019). Furthermore, in cell culture, CBD can
decreased GAD67 expression in the hippocampus of male prevent microglial activation induced by LPSs, inhibiting the
but not female offspring. Long-term treatment with CBD release of proinflammatory cytokines (tumor necrosis fac‑
increased hippocampal GAD67 expression regardless of sex tor-alpha (TNF-α) and IL-1β) and glutamate, a noncytokine
or gestational manipulation, reestablishing the control levels mediator of inflammation, ultimately decreasing neuroin‑
in male offspring and increasing GAD67 expression above flammation and subsequent glial activation both in vitro and
control levels in females (Osborne et al. 2019a, 2019b). Fur‑ in vivo (Kozela et al. 2017; Dos-Santos-Pereira et al. 2020).
thermore, CBD can increase GABAergic neurotransmission In a previous review regarding the use of CBD in psychia‑
by antagonizing GPR55, mainly in the basal ganglia nuclei try for psychosis, anxiety, addictive behavior, mood, cogni‑
(Kaplan et al. 2017). Conversely, CBD facilitates 5-HT1A- tive disorders, sleep disorders, attention-deficit/hyperactiv‑
mediated neurotransmission in the prefrontal cortex, modu‑ ity disorder (ADHD), and aggressiveness, the role of the
lating glutamate and GABA function (Santana et al. 2004; endocannabinoid system in social behavior, neurodevelop‑
Russo et al. 2005). Corroborating a possible association of ment, mental disorders, and ASD, the authors concluded
this effect with the potential therapeutic effect of CBD in that CBD is a potential candidate for the treatment of ASD
ASD subjects, proton magnetic resonance (MRS) assays (Poleg et al. 2019). In addition to the various comorbidi‑
of postmortem brains of patients with ASD as well as liv‑ ties present in ASD, 20–30% of patients may have epilepsy
ing ASD patients showed abnormalities in GABAergic and (Besag 2018). The US Food and Drug Administration (FDA)
glutamatergic pathways (Ajram et al. 2017; Horder et al. has recently approved the commercialization of an oral CBD
2014; Cochran et al. 2015). In addition, CBD inhibits the extract (Epidiolex, GW Pharmaceuticals) for the treatment
balanced nucleoside transporter (ENT1) responsible for of drug-resistant epilepsy (Sekar and Pack 2019), especially
synaptic adenosine uptake, thus increasing extracellular for two rare and severe forms, Lennox-Gastaut and Dravet
adenosine levels. Therefore, extracellular adenosine posi‑ syndromes. In addition, in a genetic animal model of Dravet
tively regulated by CBD can trigger a decrease in neuronal syndrome, CBD effectively reduced spontaneous seizures
hyperexcitability (Carrier et al. 2006). and social deficits in mice. This effect may be associated
As described above, CBD is a multitarget drug (Fig. 1). with restoring neuronal excitability of inhibitory interneu‑
Regarding ASD, in addition to the anti-epileptic properties rons in the hippocampal dentate gyrus (Kaplan et al. 2017).
of CBD, several other mechanisms could be associated with In a recent study, CBD administered in the S ­ cn1a+/− mouse
its potential therapeutic effects. Recently, researchers have model of Dravet syndrome prevented premature mortality
hypothesized a link between the onset of the main behavio‑ and improved several behavioral comorbidities, including
ral symptoms of ASD and the chronic neuroinflammatory impaired cognition and social interaction, without trigger‑
condition of the autistic brain. Cannabinoids might represent ing adverse motor effects associated with current pharmaco‑
a promising pharmacological tool for treating ASD neuro‑ therapy (Patra et al. 2020). Furthermore, approximately two-
inflammation. Specifically, CBD, which lacks psychotomi‑ thirds of the patients with Dravet syndrome show decreased
metic side effects, could be an effective and safe medication function in the neuronal voltage-gated sodium channel α1
for treating ASD (Carbone et al. 2021). Microglia are the subunit gene (SCN1A) due to mutations in 80% of Dravet
primary cells responsible for the immune responses in the syndrome patients (Nabbout et al. 2003).
CNS and maintain homeostasis and neuronal functions in the In a review of clinical trials designed to study the effects
brain, constantly surveying against infectious agents and inju‑ of CBD treatment on different forms of epilepsy in infants,
ries (Araujo et al. 2019). Children born from mothers who children, and teenagers affected by epilepsy resistant to
had infections or autoimmune diseases during pregnancy common antiseizure drugs, the authors concluded that CBD
are more likely to develop ASD (Jiang et al. 2016; Careaga should be effective and safe for Lennox-Gastaut syndrome,
et al. 2017). Furthermore, postmortem brains of ASD patients Dravet syndrome, and drug-resistant epilepsy (Silvestro
studied using positron-emission tomography (PET) showed et al. 2019).

13
2720 Psychopharmacology (2022) 239:2713–2734

Fig. 1  Chemical structure of CBD and potential targets for its inhibits the balanced nucleoside transporter (ENT1) responsible for
therapeutic effect on ASD. CBD acts as an agonist of the receptors synaptic adenosine uptake, increasing extracellular adenosine levels.
TRPV1, PPARγ, and 5-HT1A and as an antagonist of the receptor CBD can directly or indirectly activate Gly receptors. CBD is also
GPR55. CBD acts as an inverse agonist of the D2 and D3 receptors. capable of inhibiting sodium channels. TRPV1, transient receptor
CBD is suggested to act as an inverse agonist and a negative allos‑ potential vanilloid type 1; PPARγ, peroxisome proliferator-activated
teric modulator of CB1 and CB2 receptors. CBD inhibits the enzyme receptor gamma; 5-HT1A, serotonin receptor 1A; GPR55, G-protein-
FAAH, with a consequent increase in anandamide (AEA) levels. This coupled receptor 55; D2, dopaminergic receptor type 2; D3, dopa‑
endocannabinoid ligand can activate CB1, CB2, and TRPV1 recep‑ minergic receptor type 3; CB1, cannabinoid receptor type 1; CB2,
tors. CBD increases the activity of mitochondrial complexes and cannabinoid receptor type 2; FAAH, fatty acid amide hydrolase; A1,
displays antioxidant and anti-inflammatory effects that are partially adenosine type 1 receptor; Gly, glycine receptors
mediated by its actions on TRPV1, mitochondria, and PPARγ. CBD

Trials with CBD‑enriched Cannabis extract attended special education programs for children with ASD
in patients with ASD and met the DSM-5 criteria for this disorder at the time of
the treatment. All the children had severe behavioral prob‑
The first study was a retrospective assessment of the tolera‑ lems based on a Clinical Global Impression Scale-Severity
bility and efficacy of CBD-rich Cannabis in 60 children with score of 6 or 7. Initially, the ratio of CBD to THC was 20:1;
ASD and severe behavioral problems (Aran et al. 2019a, b). however, in 29 patients who presented insufficient responses
The children were between 5 and 18 years of age, and 83% based on CGI-S scores ≥ 5, a new strain was attempted. This
of the sample consisted of males. In this trial, the patients strain consisted of a 6:1 ratio of CBD/THC, with a maximum

13
Psychopharmacology (2022) 239:2713–2734 2721

dose of 5 mg/kg of CBD daily. In addition, 49 children were THC), three times a day. The THC:CBD ratio was 1:20 and
treated with other drugs concomitant with Cannabis oil, individually titrated. Symptom assessment and structured
including antipsychotics, mood stabilizers, benzodiazepines, questionnaires assessed side effects after 6 months of CBD-
SSRIs, and stimulants. According to their parent’s reports, enriched Cannabis use. After this period of the 155 patients
twenty children in this group showed some improvement, six on active treatment, 93 were evaluated. Within this sam‑
did not improve, and three showed worsening the condition. ple, 28 patients (30.1%) reported significant improvement,
At the end of the study, 73% of the children continued using 50 (53.7%) moderate, 6 (6.4%) mild, and 8 (8.6%) with‑
Cannabis oil. The other children stopped the treatment due out change in the condition. Twenty-three patients (25.2%)
to marked irritability, unsuccessful attempts to administer reported at least one side effect, with restlessness being the
the oil, low effectiveness, and adverse effects combined with most prevalent (6.6%). Therefore, CBD-enriched Cannabis
low efficacy. Although the results could be considered prom‑ treatment in ASD patients was safe and well-tolerated. The
ising, several limitations include a low number of patients, results presented in this approach are encouraging. However,
lack of a placebo group, the use of several different Can- similar to the other trials, a control group was not included
nabis strains, and the wide variation in the CBD/THC dose in this study, and the Cannabis dose was not adjusted to the
ratio (Table 1). patient´s weight. Symptoms such as sleep disorders and/or
In another study, oral CBD was evaluated in 53 children seizures, sensory difficulties, and food acceptance showed
diagnosed with ASD (Barchel et al. 2019). The study group significant improvement. However, two patients showed
had a median age of 11 years, and the average duration of increased agitation and irritability, which were resolved by
treatment was 66 days. In addition, quality control of CBD changing the strain.
was performed by measuring CBD plasma concentrations. A study conducted in Chile showed promising effects
Information regarding symptoms and treatment safety was of using Cannabis oil in patients with ASD (Kuester et al.
assessed twice a week through interviews with parents. 2017). Clinical changes were estimated using the CGI
Adverse effects, such as drowsiness and appetite changes, Improvement scale and the Autism Parenting Stress Index.
were moderate. Overall improvement occurred in 74.5% of At the end of the treatment, 66.7% of patients showed sig‑
the patients when all comorbidities were evaluated. The dif‑ nificant improvement in ASD symptoms, including social
ference was not reported in 21.6% of cases, and symptom communication, language, or repetitive behaviors. In gen‑
worsening occurred in 3.9% of the children. The lack of eral, symptoms such as sleep disorders and/or seizures, sen‑
information on the history of patient ASD symptoms and sory difficulties, and food acceptance showed significant
the absence of an assessment tool and a control group limit improvement, and the tolerability was good when using oral
the interpretation of the findings. In an observational study Cannabis extracts.
conducted in Brazil, 18 patients with ASD ranging in age In a recently published case report, the use of CBD-
from 6 to 17 years received 4.6 mg/kg CBD with 0.06 mg/ enriched Cannabis extract was described in a 15-year-old
kg THC (Fleury-Teixeira et al. 2019). Among the remaining patient with ASD, selective mutism, anxiety, and controlled
15 patients, 5 were epileptic patients. The parents or caregiv‑ epilepsy. After 3 months of using CBD-enriched Canna-
ers were responsible for monthly evaluating and answering bis, a mild improvement in anxiety symptoms was noticed.
questionnaires related to the symptoms of ADHD, behav‑ After 9 months of this treatment, social anxiety, irritability,
ioral disorders, motor deficits, autonomy deficits, commu‑ aggressiveness, focus, and eloquence improved. Surpris‑
nication, social interaction deficits, cognitive deficits, sleep ingly, when the dose of Cannabis extract was decreased, his
disorders, and seizures. Some improvement was reported in aggressiveness increased.
motor development, communication and social interaction, Recently, a retrospective analysis of patients with ASD
and cognitive performance. However, the small cohort size who have used Cannabis to treat related symptoms was con‑
and the critical adverse effects (e.g., irritability, increased ducted (Mostafavi and Gaitanis 2020). The study sample
appetite, conjunctival hyperemia) limit the generalization consisted of 32 patients diagnosed with ASD who used med‑
of the findings. ical marijuana or hemp-based products taken orally to treat
Schleider and coworkers (2019) conducted a prospec‑ aggression and co-morbid epilepsy. Among the study, eight
tive cohort study in Israel between 2015 and 2017 to evalu‑ patients used Cannabis mainly for epilepsy, nine patients for
ate the effects of CBD-enriched Cannabis in 188 patients aggressive behavior, and 15 patients for aggressive behav‑
diagnosed with ASD. The initial treatment for 94.7% of ior and epilepsy. Overall, 20 of 22 patients with epilepsy
patients was Cannabis oil given sublingually, seven patients reported some improvement in seizure control. In addition,
(3.7%) were allowed to buy oil and inflorescence, and three 12 of 20 patients treated for aggressive behavior reported
patients (1.5%) were allowed to buy only inflorescence. improvement. Adverse treatment effects were observed in
Most patients consumed oil consisting of 30% CBD and 4 patients, including increased obsessive–compulsive and
1.5% THC (average 79.5 ± 61.5 mg CBD and 4.0 ± 3.0 mg repetitive behavior and insomnia. Core symptoms of ASD,

13
Table 1  A summary of clinical studies examining effects of CBD in patients with ASD
2722

References Study sample Treatment schedule/dosages Study design Results Adverse events

13
Aran et al. 2019a, b Children with ASD and severe Cannabidiol-Rich Cannabis Retrospective feasibility study Behavioral outbreaks improvement Sleep disturbances, irritability, loss
behavioral problems 20:1 or 6:1 ratio CBD/THC in 61% of patients of appetite, and a serious psychotic
N = 60 (83% M) Mean total daily dose: 3.8 ± 2.6 mg/ event in a patient girl
Mean age = 11.8 ± 3.5 years kg/day CBD and 0.29 ± 0.22 mg/
Age range = 5–18 years kg/day THC for children who
received three daily doses (n = 44)
and 1.8 ± 1.6 mg/kg/day CBD and
0.22 ± 0.14 mg/kg/day THC for
children who received two daily
doses (n = 16)
Mean treatment duration:
10.9 ± 2.3 months
Barchel et al. 2019 Children with ASD-related comor‑ Cannabidiol-rich Cannabis Case reports CBD improved: Moderate somnolence and changes in
bidities 20:1 ratio CBD/THC Self-injury in 67.6% of cases appetite
N = 53 (85% M) CBD mean total daily dose: 16 mg/ Hyperactivity in 68.4% of cases
Mean age = 11 years kg (maximal daily dose 600 mg) Sleep problems in 71.4% of cases
Age range = 4–22 years THC mean total daily dose: 0.8 mg/ Anxiety in 47.1% of cases
kg Overall improvement in 74.5% of
Mean treatment duration: 66 days cases
(30–588 days)
Fleury-Teixeira et al. 2019 Cohort of patients with ASD Cannabidiol-Rich Cannabis Observational study Improvement in multiple autistic Mild and/or transient like sleepiness,
N = 18 (72% M) 75:1 ratio CBD/THC symptoms like sleep disorders, moderate irritability, diarrhea,
Mean age = 10 years CBD mean total daily dose: seizures and behavioral crisis, increased appetite, conjunctival
Age range = 6–17 years 4.6 mg/kg social interaction and cognitive hyperemia, and increased body
THC mean total daily dose: performance temperature
0.06 mg/kg
Children received two daily doses
Mean treatment duration:
6–9 months
Schleider et al. 2019 Children with ASD Cannabis oil 20:1 CBD/THC Prospective cohort study Hyperactivity: Improvement: 68.4% Sleepiness, moderate irritability, diar‑
N = 188 (81.9% M) Mean total daily dose: 30% CBD of cases rhea, increased appetite, conjunctival
Mean age = 12.9 ± 7.0 years and 1.5% THC, on aver‑ Self-injury: improvement: 67.6% hyperemia, increased body tempera‑
Age range = younger than the age of age 79.5 ± 61.5 mg CBD and of cases ture, and restlessness
5 to18 years 4.0 ± 3.0 mg THC, three daily Sleep Problems: Improvement:
doses. In case of insomnia addi‑ 71.4% of cases
tional 5.0 ± 4.5 mg THC daily was Anxiety: improvement: 47.1% of
prescribed cases
Mean treatment duration: 6 months Overall: improvement: 74.5% of
cases
Kuester et al. 2017 Children with ASD Balanced CBD: THC extracts in Retrospective case series (abstract From the CGI-I and APSI rating One patient had greater irritability and
N = 20 (15 M) 71.5% of patients; high-CBD in only) scales, 66.7% of patients had sig‑ two patients more agitation; these
Mean age = 9 years and 10 months 19% of patients; and high-THC nificant improvement. At least one effects were solved with the change
Age range = 2–22 years extracts in 9.5% of patients. Daily of the core symptoms improved of the strain
dose was not reported In most cases, sensory difficulties,
Mean treatment duration: 7.6 months food acceptance, feeding and sleep
disorders, and/or seizures have
improved
Psychopharmacology (2022) 239:2713–2734
Table 1  (continued)
References Study sample Treatment schedule/dosages Study design Results Adverse events

Ponton et al. 2020 15-year-old boy Cannabidiol-rich Cannabis 20:1 ratio Case report Improvement in social anxiety, No significant adverse effects
CBD/THC. First 3 months: 2 mg/ irritability, aggressiveness, focus,
kg of CBD and 0.1 mg/kg of THC, eloquence and social interaction
two times a day in the volume of
0.1 ml. After this period, the dose
was increased to 0.2 ml twice a
day (4 mg/kg CBD and 0.2 mg/kg
THC). Mean treatment duration:
9 months
Mostafavi and Gaitanis 2020 32 patients with ASD diagnosis No mentioned Retrospective analyses 20 of 22 patients with epilepsy (91%) Increased obsessive–compulsive and
showed some improvement in sei‑ repetitive behavior, mania, and
zure control. 12 out of 20 patients insomnia
Psychopharmacology (2022) 239:2713–2734

treated for aggressive behavior


(60%) exhibited improvement
Aran et al. 2021 Children and adolescents with ASD The study was divided into 3 Placebo-controlled trial The changes in the total scores of the Mild or moderate drowsiness,
N = 150 (80% M) groups:—whole-plant Cannabis HSQ-ASD (primary outcome) and decreased appetite, weight loss,
Mean age = 11.8 ± 4.1 years extract containing cannabidiol and APSI (secondary outcome) did not tiredness, euphoria, and anxiety
Age range = 5–21 years Δ9-tetrahydrocannabinol in a 20: differ between groups. Disruptive
1 ratio,—purified cannabidiol, and behavior in CGI-I (co-primary
Δ9-tetrahydrocannabinol in the outcome) improved or greatly
same proportion and the placebo improved in 49% of patients who
group. To assess the effectiveness received the whole plant extract
of treatments, patients received (n = 45) when compared to the
placebo or cannabinoids for placebo group (21% of patients,
4 months followed by a 4-week n = 47). The SRS mean (secondary
washout and cross-over designed result) improved by 14.9 points in
and predetermined for an addi‑ the group that received the extract
tional 12 weeks to better assess of the whole plant (n = 34) com‑
patients' tolerability to treatment. pared to the placebo group that had
Initially, patients received 1 mg/kg a score of 3.6 points (n = 36)
of CBD and 0.05 mg/kg of THC
daily. On alternate days the doses
were increased by 1 mg/kg for
CBD and 0.05 mg for THC until
reaching 10 mg/kg of body weight
per day of CBD and 0.5 mg/kg of
THC for children with a weight
between 20 and 40 kg or 7.5 mg/
kg of CBD per day and 0.375 mg/
kg per day of THC for patients
weighing more than 40 kg. The
treatments were administered
orally divided into 3 daily doses
Bilge and Ekici, 2021 Children with ASD Cannabidiol-rich Cannabis Observational Study Decrease in behavioral problems in No significant side effects
N = 33 (82% M) Full-spectrum CBD and trace THC 10 patients (32.2%). Iincrease in
Mean age = 7.7 ± 5.5 years CBD mean total daily dose: 0.7 mg/ expressive language in 7 patients
kg (22.5%). Improvement in cognition
Children received one sublingual in 4 patients (12.9%). Increase
drop twice a day and one drop in social interaction in 3 patients
every 3 days (9.6%). Decrease in stereotypes in
Mean treatment duration: 6.5 months 1 patient (3.2%)

13
2723
2724 Psychopharmacology (2022) 239:2713–2734

such as language and social development impairments, have

Dizziness, insomnia, colic and weight


not been evaluated in patients.
Aran and coworkers (2021) conducted a placebo-con‑
trolled trial of whole-plant extract and pure cannabinoids in
150 children and adolescents diagnosed with ASD according
to DSM-5 criteria, which was confirmed using the Autism
Adverse events

Diagnostic Observation Schedule (ADOS-2), and moder‑


ate or more significant behavioral problems (rating ≥ 4) on
gain

the CGI-S scale. The participants were 5–21 years of age,


and 20% of the patients were females. The patients were
randomly assigned (1:1:1 ratio) to 1 of 3 treatments for
Improvement in social interaction,

number of meals a day and con‑


anxiety psychomotor agitation

12 weeks. Treatments included oral placebo, whole-plant


Cannabis extract containing CBD and THC at a 20:1 ratio,
and pure CBD and pure THC at the same ratio and concen‑
tration. Two objectives were assessed in the study. First, it
investigated whether whole-plant Cannabis extract signifi‑
centration

cantly improved behavioral assessments compared to pla‑


Results

cebo. The identical CBD:THC ratio was used in the previous


open-label case series. The treatments were administered
Randomized, double-blind and con‑

orally, mostly sublingually. The study started with 50 partici‑


pants randomly chosen with three different treatments. In the
trolled placebo clinical trial

first phase of the study, which lasted 12 weeks, preliminary


results were assessed using the CGI-I scale with disruptive
behavior anchor poibehaviorHome Situation Questionnaire
Study design

for e ASD (HSQ-ASD). After 4 weeks of washout followed


by the second treatment period lasting 12 weeks, the results
obtained were evaluated using the Social Responsiveness
Scale (SRS-2) and APSI (Autism Parenting Stress Index),
CBD-rich Cannabis extract at a con‑

and adverse events were assessed. At the end of the first


centration of 0.5% (5 mg/mL)

Treatment duration: 12 months

period, 45 patients who received whole-plant and pure can‑


Treatment schedule/dosages

nabinoids and 47 subjects in the placebo group completed


the study. At the end of the second study period, 44 subjects
CBD-Rich Cannabis
9:1 ratio CBD/THC

N, number of subjects; CBD, cannabidiol; THC, tetrahydrocannabinol; M, male


6–70 drops daily

th the experiment and placebo groups completed the study.


Cannabinoid treatment for behavioral problems was assessed
using the HSQ-ASD and CGI-I scores (co-primary outcome
measures). The APSI was used to measure the second study
period results and evaluate the child’s behavior. The HSQ-
ASD and APSI total scores were not significantly different
Control group (N = 29; 27 M)

between the patients who received cannabinoids and those


Mean age = 7.68 ± 1.74 years
CBD group (N = 31; 29 M)

who received placebo. The CGI-I scores of 45 participants


Age range = 5–11 years

(49%) who received whole-plant cannabinoids and answered


Children with ASD

the questionnaire were equal to or higher than 21% of the


N = 60 (52 M)
Study sample

47 subjects who received a placebo. Among the 45 partici‑


pants who received pure cannabinoids, 38% of the respond‑
ent CGI-I scores were not significantly higher than subjects
who received a placebo. The HSQ-ASD, CGI-I, and APSI
scores were not significantly different between participants
who received whole-plant extract versus pure cannabinoids.
Table 1  (continued)

Furthermore, the ASD symptoms in the second study period


were assessed using SRS-2. The total SRS-2 scores signifi‑
Silva et al. 2022

cantly improved after treatment with whole-plant extract


References

compared with placebo. In addition, treatment with the


whole plant extract was significantly associated with CGI-I

13
Psychopharmacology (2022) 239:2713–2734 2725

and SRS-2 scores after controlling for sleepiness with the GABA + (GABA + macromolecules) levels after receiving
concomitant use of medications during treatment. The most a single oral dose of CBD (600 mg/kg) or placebo. CBD
common side effects, when they occurred, were drowsi‑ altered the main excitatory and inhibitory neurotransmitters
ness, decreased appetite, weight loss, tiredness, euphoria, in adults with and without ASD. CBD increased Glx in the
and anxiety. Although the initial study was designed to be basal ganglia in both groups and decreased Glx in the dor‑
a crossover assay, the first analyses revealed the treatment somedial prefrontal cortex. Conversely, CBD had opposite
results would affect the data from the second treatment effects on GABA + levels in each group. Specifically, both
period. Thus, the sample size was limited. The question‑ in the prefrontal and subcortical regions, CBD increased
naires showed several items were inapplicable to some low- GABA + levels in controls but decreased the levels in ASD
functioning participants, which resulted in invalid scores and patients.
a decrease in the statistical power of these measures. The In another study published in 2019 by Pretzcsh et al. using
study did not conduct genetic or intelligence quotient assess‑ the same study cohort and similar methodology, the effects
ments, and the effects of gene history or cognitive level on of CBD on low-frequency activity and functional connectiv‑
treatment response could not be assessed. Collected data ity in the brain were evaluated. This approach measured the
about the use of concomitant medications were collected. fractional amplitude of low-frequency fluctuations (fALFF)
However, the authors did not mention methods to detect in the whole brain. CBD significantly increased fALFF in
factors that may affect the treatment response or adverse cerebellar vermis VI and the right fusiform gyrus of patients
events. In addition, pharmacokinetics and interventions were with ASD. Furthermore, the change in fALFF in the cerebel‑
not performed, and information regarding other drugs, liver lum (but not in the fusiform gyrus) was accompanied by
enzyme tests, and complete blood count were not collected. generalized differences in the functional vermal connectivity
The authors stated they detected no clinical evidence of with several of its subcortical and cortical targets in ASD
hepatic or hematological dysfunction. patients but not in controls.
In a 2-year observational study conducted in Turkey,
33 children with a mean age of 7.7 years diagnosed with
ASD received CBD-enriched Cannabis daily. According to Discussion
reports from parents and caregivers, there was considerable
improvement in the behavior of patients. Unlike other tri‑ CBD has several potential neuropsychiatric therapeutic
als, this study used lower doses of CBD and traces of THC, effects on important manifestations of ASD (Fig. 2). Fur‑
which could explain the lack of side effects in the sample thermore, several of its multiple molecular mechanisms tar‑
evaluated. get brain changes involved in the pathophysiology of ASD.
Recently, a randomized, double-blind, placebo-controlled Because CBD is generally well-tolerated, with no abuse
clinical trial conducted in Brazil evaluated the efficacy and potential and low toxicity in humans and other species, it
safety of a CBD-enriched Cannabis extract in autistic chil‑ could be a practical single or add-on alternative therapy for
dren. Sixty children, aged between 5 and 11 years, were ASD (Scuderi et al. 2009). Some preclinical study results
selected and divided into two groups: the treatment group, support this possibility. However, the clinical investigations
which received the CBD-rich cannabis extract, and the con‑ of CBD effects on ASD are limited. Positive results have
trol group, which received the placebo for 12 weeks. The been reported in open-label trials and case reports, often
Sociodemographic Questionnaire and Childhood Autism accompanied by decreased dosage and medication with‑
Rating Scale (CARS) was used. In this study, a wide drawal. Side effects are mild or moderate when they appear.
improvement in social interaction was observed; the side They are usually diarrhea, sleep and appetite disturbances,
effects, when present, were dizziness, insomnia, colic, and drowsiness, and restlessness, which generally disappear with
weight gain. decreasing dosages or treatment discontinuation. However,
In addition to these papers in which the effects of CBD proper double-blind, placebo-controlled trials are needed to
on ASD symptomatology are described, the effects of a explore this possibility further. The results obtained with
single oral dose of CBD in ASD patients on brain excita‑ CBD-enriched Cannabis in patients with ASD are encourag‑
tion and inhibition systems and functional connectivity in ing for patients with and without epileptic conditions. How‑
structures associated with this disorder are addressed in ever, some concerns should be highlighted.
two papers. In a randomized placebo-controlled single- The gender disparity in the samples evaluated is apparent
dose trial using MRS, the CBD effects on excitatory and in clinical trials previously discussed. Although the preva‑
inhibitory systems were investigated in male adults with lence of ASD is higher in males, it is essential to note that
and without ASD (Pretzsch et al. 2019a, b). The sample persistent impairments in communication and social interac‑
consisted of 34 males (17 controls and 17 with ASD). MRS tion are very relevant features in ASD. It is suggested that
measured glutamate (Glx = glutamate + glutamine) and late or missed diagnosis in females may be related to marked

13
2726 Psychopharmacology (2022) 239:2713–2734

Fig. 2  The following image


suggests the therapeutic profile
of CBD in treating the core
symptoms of ASD, such as
improving social interaction and
communication, and decreas‑
ing stereotyped behaviors, in
addition to its neuroprotective
profile. CBD may also have
therapeutic effects on ASD-
associated comorbidities such as
psychosis, aggression, epilepsy,
conditions related to anxiety
and depression, and obsessive–
compulsive disorder (OCD).
CBD may offer a promising
treatment for the range of symp‑
toms of ASD. However, due to
the complexity of ASD, it is still
a challenge to select patients
who would benefit from the
therapeutic effects of CBD and
individuals who are more prone
to side effects

sex differences in the phenotypic presentation of these symp‑ enzymes such as Cytochrome P450, highlighting the iso‑
toms (Rivet and Matson 2011; Lai and Baron-Cohen 2015; forms CYP3A4 and CYP2C19 (Jiang et al. 2011). CBD
Bargiela et al. 2016). Thus, sexual dimorphism in the expres‑ can inhibit members of the CYP3 family, where 60% of
sion of genes is related to ASD, where boys, in general, are clinically prescribed medications are also metabolized
more susceptible than girls to genetic alterations involved through it (Jiang et al. 2013; Zendulka et al. 2016). In addi‑
in synaptic plasticity and brain development, where the tion, this compound inhibits the liver enzyme CYP2D6
endocannabinoid system plays a crucial role (Maccarrone and may increase serum concentrations of SSRIs, tricy‑
et al. 2014). It is important to note that sexually dimorphic clic antidepressants, antipsychotics, beta-blockers, and
neural pathways are intimately involved in synaptic struc‑ opioids. CBD can interact with various antiepileptics by
ture, function, and plasticity. Moreover, sex hormones can inhibiting CYP2C19 and CYP3A4, which catalyze the
modulate brain endocannabinoid activity (Fattore and Fratta metabolism of N-desmethylclobazam (nCLB), an active
2010). A higher male-to-female ratio in autism may arise metabolite of clobazam (Giraud et al. 2004; Walzer et al.
because males have a lower threshold than females for aber‑ 2012). The inhibition of these enzymes by CBD leads to
rant changes in synaptic dynamics during neurodevelopment the accumulation of nCBL, which is about 20–100% as
following genetic or environmental insults (Mottron et al. potent as clobazam. A study by Gaston and Szaflarski
2015). (2018) showed that the effectiveness of antiepileptics can
The great popularity of CBD-enriched Cannabis-based be modulated by CBD; however, its antiepileptic profile
drugs available on the market, combined with the fact that is not affected by this class of drugs. Great attention is
patients with ASD use a range of drugs to treat the vari‑ recommended in the interactions of CBD with Warfarin
ety of symptoms present, makes it imperative to expand and with acetaminophen since, respectively, the interac‑
the knowledge of drug-drug interactions (Balachandran tions can culminate in increased bleeding (Damkier et al.
et al. 2021). CBD is extensively metabolized by liver 2019) and liver damage (Brown and Winterstein 2019).

13
Psychopharmacology (2022) 239:2713–2734 2727

Therefore, in these cases, it is recommended to reduce the Ethical and legal concerns
dose of CBD. Besides, CBD can interact with alcohol, but
these effects are still poorly understood. However, this Most of the trials evaluated in the present review raise vari‑
drug can decrease alcohol consumption and blood levels. ous legal and ethical concerns because the children with
In addition, CBD promotes protection against the harm‑ ASD received non-pure-grade CBD and even a mixture of
ful effects of alcohol on the liver and brain and reduces CBD and THC. To date, THC:CBD combinations are not
oxidative stress (Nona et al. 2019; Yang et al. 2014; Con‑ acceptable in any dose range for non-compassionate use
sroe et al. 1979). On the other hand, this interaction pro‑ in children. ASD children were chronically exposed (up
moted motor and psychomotor disruptions (Robinson and to 6 months) to THC (up to 21 mg/day). For comparison,
Berridge 1993). Thus, as discussed, further double-blind, in previous studies, an oral dose of 10 mg of THC acutely
controlled, randomized studies are essential to assess the induced psychosis in healthy adults (Martin-Santos et al.
interactions of CBD with other drugs, as well as bioavail‑ 2012; Bhattacharyya et al. 2009). The THC exposure dur‑
ability, pharmacokinetics, and pharmacodynamics assess‑ ing the neurodevelopmental phase may increase the risk of
ments in these contexts. later behavioral sequelae, including cognition, motivation,
The endocannabinoid system is widely involved in impulsivity, mood, anxiety, psychosis, intelligence, and psy‑
embryonic and adult neurogenesis (Jiang et al. 2005; Trazzi chosocial functioning (Sorkhou et al. 2021). The evidence
et al. 2010). During neurodevelopment, synapse formation, shows that frequency and amount of THC content, early
cell migration, axonal orientation, and cell survival are age of use, and cumulative exposure can contribute to these
modulated. In addition, changes in these processes during adverse outcomes even in people without past medical con‑
development are primarily involved with the emergence of ditions or psychiatric disorders (Sorkhou et al. 2021). The
psychiatric disorders (Maccarrone et al. 2014). The manipu‑ adverse outcomes may even be worse in the ASD population
lation of the endocannabinoid system, especially during the because they are more prone to these comorbidities. Because
CNS maturation period, should be approached with caution, the plasma level of the different cannabinoids varies widely,
especially with the use of THC, which at the molecular level especially in ASD children taking other drugs that cause
acts as a partial CB1 receptor agonist and produces its psy‑ drug-drug interactions, dosing both the cannabinoids and the
choactive effects through the receptors. Furthermore, the other medicines is strongly recommended. General medical
recreational use of Cannabis is associated with an increased examinations should be given and include measurements
risk of developing psychosis in a dose-dependent manner of hepatic enzymes and sexual hormones. Non-GMP/GLP
(De Souza Crippa et al. 2004). Clinical trial results have drugs were given to ASD children in studies without the
shown that for patients with psychosis, as in the general ideal aging down, block study design. Therefore, it is crucial
population, the use of this drug is associated with a higher that in clinical studies involving children, the CBD adminis‑
incidence of psychotic disorders and cognitive impairment tered should be a pure and pharmaceutical-grade compound,
(Meier et al. 2012). In addition, the use of Cannabis is asso‑ and the study population should be advised regarding the
ciated with an earlier onset of psychosis and severe cognitive possible hazards of consuming unregulated products labeled
impairment (Bagot et al. 2015). A strong relationship exists as CBD to minimize possible risks.
between patients who have psychosis and individuals who
routinely use Cannabis showing a higher risk of relapse due
to the disorder, exacerbation of symptoms, and longer-term Conclusions
hospitalization (for review, see Koskinen et al. 2010; Large
et al. 2011; Marconi et al. 2016; Schoeler et al. 2016; Bogaty ASD is generally a debilitating condition, and available
et al. 2018; Hunt et al. 2018; Murrie et al. 2020). treatments have limited efficacy and are associated with
In a pioneering meta-analysis study, evidence of the significant adverse reactions. The endocannabinoid system
relationship between episodes of psychosis and ASD was is involved in the pathophysiology of ASD, indicating that
observed (Kiyono et al. 2020). The study results indicated CBD could have therapeutic effects on the main and associ‑
the prevalence of psychotic episodes in ASD could reach ated symptoms. However, only a few randomized controlled
24%, 2–fivefold higher than in the general population. Cor‑ trials involved the administration of CBD to ASD patients,
roborating the findings of this study, in a recent review by and highly purified GMP/GLP compounds were not used in
Zheng et al. (2018), the authors reported that individuals any of the studies. The main concerns with the use of CBD
with ASD are 3.55 times more likely to have schizophre‑ as a therapeutic for ASD include the potential for long-term
nia than healthy people and a high incidence of episodes adverse effects, drug-drug interactions, and lack of regula‑
of psychosis. Based on this evidence, patients with ASD tory oversight of retail CBD-enriched products. Therefore,
have a higher prevalence of schizophrenia and episodes of an adequate therapeutic window for CBD should be deter‑
psychosis than the general population. mined to achieve satisfactory effects. In all the studies in

13
2728 Psychopharmacology (2022) 239:2713–2734

which CBD use was evaluated for the treatment of ASD, Adams R, Hunt M, Clark JH (1940) Structure of cannabidiol, a product
erratic CBD-enriched Cannabis extract oils with other phy‑ isolated from the Marihuana extract of Minnesota Wild Hemp I.
J Am Chem Soc 62:196–200
tocannabinoid molecules (such as THC) were used. Thus, Ajram LA, Horder J, Mendez MA, Galanopoulos A, Brennan LP,
CBD safety requires further investigation, and an adequate Wichers RH et al (2017) Shifting brain inhibitory balance and
therapeutic window must be determined for the main ASD connectivity of the prefrontal cortex of adults with autism spec‑
symptoms and comorbid conditions. Finally, further clinical trum disorder Transl. Psychiatry 7:e1137
American Psychiatric Association (2013) Diagnostic and statistical
long-term, placebo-controlled trials using pharmaceutical- manual of mental disorders (5th ed.) American Psychiatric Asso‑
grade CBD, involving different doses and neurodevelop‑ ciation press, Washington, DC
mental treatment periods, are necessary to prove the effec‑ Anagnostou E, Soorya L, Chaplin W, Bartz J, Halpern D, Wasserman S
tiveness, safety, and absence of acute and severe long-term et al (2012) Intranasal oxytocin versus placebo in the treatment of
adults with autism spectrum disorders: a randomized controlled
adverse effects in ASD patients. trial. Mol Autism 3:1–9
Aran A, Cassuto H, Lubotzky A, Wattad N, Hazan E (2019) Brief
Acknowledgements The authors thank Lucas Prota Crippa for prepar‑ report: cannabidiol-rich cannabis in children with autism spec‑
ing the art in Fig. 2 trum disorder and severe behavioral problems—a retrospective
feasibility study. J Autism Dev Disord 49:1284–1288
Author contribution All authors have contributed to the writing of Aran A, Eylon M, Harel M, Polianski L, Nemirovski A, Tepper S et al
this review. (2019) Lower circulating endocannabinoid levels in children with
autism spectrum disorder. Mol Autism 10:1–11
Funding This work was supported by the Fundação de Amparo à Pes‑ Aran A, Harel M, Cassuto H, Polyansky L, Schnapp A, Wattad N,
quisa do Estado de São Paulo (FAPESP) and by the Instituto Nacional Shmueli D, Golan D, Castellanos FX (2021) Cannabinoid treat‑
de Ciência e Tecnologia Translacional em Medicina (INCT-TM; CNPq/ ment for autism: a proof-of-concept randomized trial. Mol
FAPESP; 2008/09009–2). JAC received a grant from the University Autism 12:1–11
Global Partnership Network (UGPN) – Global Priorities in Cannabi‑ Araujo DJ, Tjoa K, Saijo K (2019) The endocannabinoid system as a
noid Research Excellence Program. ACC, FSG, EADB, JAC, JEH, and window into microglial biology and its relationship to autism.
AWZ are recipients of CNPq research fellowships. The authors appreci‑ Front Cell Neurosci 13:1–7
ate the financial support from Fapesp, Capes, and CNPq. Ashwood P, Wills S, Van de Water J (2006) The immune response in
autism: a new frontier for autism research. J Leukoc Biol 80:1–15
Atladóttir HÓ, Thorsen P, Østergaard L, Schendel DE, Lemcke S,
Declarations Abdalla M et al (2010) Maternal infection requiring hospitaliza‑
tion during pregnancy and autism spectrum disorders. J Autism
Conflict of interest JAC is a member of the International Advisory Dev Disord 40:1423–1430
Board of the Australian Centre for Cannabinoid Clinical and Re‑ Bagot KS, Mili R, Kaminer Y (2015) Adolescent initiation of cannabis
search Excellence (ACRE) – National Health and Medical Research use and early-onset psychosis. Subst Abus 36:524–533
Council (NHMRC). JAC and JEH have received travel support to at‑ Baio J, Wiggins L, Christensen DL, Maenner MJ, Daniels J, Warren
tend scientific meetings and personal consultation fees from BSPG- Z et al (2018) Prevalence of autism spectrum disorder among
Pharm. JAC, JEH, FSG, and AWZ are co-inventors of the patent children aged 8 Years - Autism and developmental disabilities
“Fluorinated CBD compounds, compositions and uses thereof. monitoring network, 11 Sites, United States, 2014 MMWR. Sur‑
Pub. No.: W.O./2014/108899. International Application No.: PCT/ veill Summ 67:1–23
IL2014/050023,” Def. U.S. number Reg. 62193296; July 29, 2015; Balachandran P, Elsohly M, Hill KP (2021) Cannabidiol interactions
INPI on August 19, 2015 (BR1120150164927; Mechoulam R, Zuardi with medications, illicit substances, and alcohol: a comprehen‑
AW, Kapczinski F, Hallak JEC, Guimarâes FS, Crippa JAS, Breuer A). sive review. J Gen Intern Med 36:2074–2084
Universidade de São Paulo (USP) has licensed this patent to Phytecs Baranova J, Dragunas G, Botellho MCS, Ayub ALP, Bueno-Alves
Pharm (USP Resolution No. 15.1.130002.1.1) and has an agreement R, Alencar RR, Papaiz DD, Sogayar MC, Ulrich H, Correa RG
with Prati-Donaduzzi to “develop a pharmaceutical product contain‑ (2021) Autism spectrum disorder: signaling pathways and pro‑
ing synthetic CBD and prove its safety and therapeutic efficacy in the spective therapeutic targets. Cell Mol Neurobiol 41:619–649
treatment of epilepsy, schizophrenia, Parkinson’s disease, and anxiety Barchel D, Stolar O, De-Haan T, Ziv-Baran T, Saban N, Fuchs DO,
disorders.” JAC, JEH, FSG, and AWZ are co-inventors of the patent Koren G, Berkovitch M (2019) Oral cannabidiol use in children
“Cannabinoid-containing oral pharmaceutical composition, method with autism spectrum disorder to treat related symptoms and
for preparing and using same,” INPI on September 16, 2016 (BR co-morbidities. Front Pharmacol 9:1521
112018005423–2). JAC and JECH have received personal consultation Bargiela S, Steward R, Mandy W (2016) The experiences of late-
fees from BSPG-Pharm, and PurMed Global in the past. JAC received diagnosed women with autism spectrum conditions: an inves‑
speaking fees from Torrent, Green Care Store and Janssen. The other tigation of the female autism phenotype. J Autism Dev Disord
authors declare no competing interests. 46:3281–3294
Batalla A, Bos J, Postma A, Bossong MG (2021) The impact of can‑
nabidiol on human brain function: a systematic review. Front in
Pharmacol 11:618184
Beltramo M, Stella N, Calignano A, Lin SY, Makriyannis A, Piomelli
References D (1997) Functional role of high-affinity anandamide transport,
as revealed by selective inhibition. Science 277:1094–1097
Abdul-Monim Z, Neill JC, Reynolds GP (2007) Sub-chronic psychoto‑ Bergamaschi MM, Queiroz RH, Chagas MH, de Oliveira DC, De
mimetic phencyclidine induces deficits in reversal learning and Martinis BS, Kapczinski F, Quevedo J, Roesler R, Schröder N,
alterations in parvalbumin-immunoreactive expression in the rat. Nardi AE, Martín-Santos R, Hallak JE, Zuardi AW, Crippa JA
J Psychopharmacol 21:198–205 (2011) Cannabidiol reduces the anxiety induced by simulated

13
Psychopharmacology (2022) 239:2713–2734 2729

public speaking in treatment-naïve social phobia patients. Neu‑ Carlini EA, Mechoulam R, Lander N (1975) Anticonvulsant activ‑
ropsychopharmacology 36:1219–1226 ity of four oxygenated cannabidiol derivatives. Res Commun
Bernaerts S, Boets B, Bosmans G, Steyaert J, Alaerts K (2020) Chem Pathol Pharmacol 12:1–15
Behavioral effects of multiple-dose oxytocin treatment in Carrier EJ, Auchampach JA, Hillard CJ (2006) Inhibition of an
autism: a randomized, placebo-controlled trial with long-term equilibrative nucleoside transporter by cannabidiol: a mecha‑
follow-up. Mol Autism 11:1–14 nism of cannabinoid immunosuppression. Proc Natl Acad Sci
Besag FMC (2018) Epilepsy in patients with autism: links, risks 103:7895–7900
and treatment challenges. Neuropsychiatr Dis Treat 14:1–10 Carter CS (2014) Oxytocin pathways and the evolution of human
Bhattacharyya S, Fusar-Poli P, Borgwardt S, Martin-Santos R, Nosarti C, behavior. Annu Rev Psychol 65:17–39
O’Carroll C, Allen P, Seal ML, Fletcher PC, Crippa JA, Giampi‑ Casarotto PC, Resstel Gomes FV, LBM, Guimarães, F S, (2010)
etro V, Mechelli A, Atakan Z, McGuire P (2009) Modulation of Cannabidiol inhibitory effect on marble-burying behavior:
mediotemporal and ventrostriatal function in humans by delta9- involvement of CB1 receptors. Behav Pharmacol 21:353–358
tetrahydrocannabinol: a neural basis for the effects of Cannabis Chadwick B, Miller ML, Hurd YL (2013) Cannabis use during ado‑
sativa on learning and psychosis. Arch Gen Psychiatry 66:442–451 lescent development: susceptibility to psychiatric illness. Front
Bhattacharyya S, Morrison PD, Fusar-Poli P, Martin-Santos R, Borg‑ Psychiatry 4:1–8
wardt S, Winton-Brown T et al (2010) Opposite effects of δ-9- Chakrabarti B, Baron-Cohen S (2011) Variation in the human can‑
tetrahydrocannabinol and cannabidiol on human brain function nabinoid receptor CNR1 gene modulates gaze duration for
and psychopathology. Neuropsychopharmacology 35:764–774 happy faces. Mol Autism 2:1–7
Bilge S, Ekici B (2021) CBD-enriched cannabis for autism spec‑ Chakrabarti B, Kent L, Suckling J, Bullmore E, Baron-Cohen S
trum disorder: an experience of a single center in Turkey and (2006) Variations in the human cannabinoid receptor (CNR1)
reviews of the literature. J Cannabis Res 3:53 gene modulate striatal responses to happy faces. Eur J Neurosci
Bisogno T, Hanuš L, De Petrocellis L, Tchilibon S, Ponde DE, 23:1944–1948
Brandi I et al (2001) Molecular targets for cannabidiol and its Chevallier C, Kohls G, Troiani V, Brodkin ES, Schultz RT (2012)
synthetic analogues: effect on vanilloid VR1 receptors and on The social motivation theory of autism. Trends Cogn Sci
the cellular uptake and enzymatic hydrolysis of anandamide. 16:231–239
Br J Pharmacol 134:845–852 Choi GB, Won H, Yim YS, Kim SV, Cai Y, Hoeffer CA et al (2016)
Bogaty SER, Lee RSC, Hickie IB, Hermens DF (2018) Meta-analysis The maternal ROR γ t / IL-17a pathway promotes autism-like
of neurocognition in young psychosis patients with current phenotypes in offspring. Science 351:933–940
cannabis use. J Psychiatr Res 99:22–32 Clayton N, Marshall FH, Bountra C, O’Shaughnessy CT (2002) CB1
Bölte S, Girdler S, Marschik PB (2019) The contribution of environ‑ and CB2 cannabinoid receptors are implicated in inflammatory
mental exposure to the etiology of autism spectrum disorder. pain. Pain 96:253–260
Cell Mol Life Sci 76:1275–1297 Cochran DM, Sikoglu EM, Hodge SM, Edden RAE, Foley A,
Breivogel CS, Sim LJ, Childers SR (1997) Regional differences in Kennedy DN et al (2015) Relationship among glutamine,
cannabinoid receptor/G-protein coupling in rat brain. J Phar‑ γ-aminobutyric acid, and social cognition in autism spectrum
macol Exp Ther 282:1632–1642 disorders. J Child Adolesc Psychopharmacol 25:314–322
Brown JD, Winterstein AG (2019) Potential adverse drug events Coghlan S, Horder J, Inkster B, Mendez MA, Murphy DG, Nutt
and drug–drug interactions with medical and consumer Can‑ DJ (2012) GABA system dysfunction in autism and related
nabidiol (CBD) use. J Clin Med 8:989 disorders: from synapse to symptoms. Neurosci Biobehav Rev
Brugha TS, McManus S, Bankart J, Scott F, Purdon S, Smith J et al 36:2044–2055
(2011) Epidemiology of autism spectrum disorders in adults in Consroe P, Carlini EA, Zwicker AP, Lacerda LA (1979) Interaction of
the community in England. Arch Gen Psychiatry 68:459–465 cannabidiol and alcohol in humans. Psychopharmacology 66:45–50
Bugalho P, Correa B, Viana-Baptista M (2006) Role of the cerebel‑ Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gil‑
lum in cognitive and behavioral control: scientific basis and ula NB (1996) Molecular characterization of an enzyme that
investigation models. Acta Med Port 19:257–267 degrades neuromodulatory fatty-acid amides. Nature 7:83–87
Campos AC, Guimarães FS (2008) Involvement of 5HT1A receptors Crippa JAS, Nogueira Derenusson G, Borduqui Ferrari T, Wichert-
in the anxiolytic-like effects of cannabidiol injected into the Ana L, Duran FLS, Martin-Santos R et al (2011) Neural basis
dorsolateral periaqueductal gray of rats. Psychopharmacology of anxiolytic effects of cannabidiol (CBD) in generalized social
199:223–230 anxiety disorder: a preliminary report. J Psychopharmacol
Campos AC, Ortega Z, Palazuelos J, Fogaça MV, Aguiar DC, Díaz- 25:121–130
Alonso J, Ortega-Gutiérrez S, Vázquez-Villa H, Moreira FA, Crippa JA, Guimarães FS, Campos AC, Zuardi AW (2018) Transla‑
Guzmán M, Galve-Roperh I, Guimarães FS (2013) The anxio‑ tional investigation of the therapeutic potential of cannabidiol
lytic effect of cannabidiol on chronically stressed mice depends (CBD): toward a new age. Front Immunol 9:2009
on hippocampal neurogenesis: involvement of the endocan‑ Cristino L, Bisogno T, Di Marzo V (2020) Cannabinoids and the
nabinoid system. Int J Neuropsychopharmacol 16:1407–1419 expanded endocannabinoid system in neurological disorders.
Capano L, Dupuis A, Brian J, Mankad D, Genore L, Adams RH, Nat Rev Neurol 16:9–29
Smile S (2018) A pilot dose finding study of pioglitazone in Cunha JM, Carlini EA, Pereira AE, Ramos OL, Pimentel C, Gagliardi
autistic children. Mol Autism 9:59. https://​pubmed.​ncbi.​nlm.​ R et al (1980) Chronic administration of cannabidiol to healthy
nih.​gov/​30498​564/ volunteers and epileptic patients. Pharmacology 21:175–185
Carbone E, Manduca A, Cacchione C, Vicari S, Trezza V (2021) Dalton P, Deacon R, Blamire A, Pike M, McKinlay I, Stein J et al
Healing autism spectrum disorder with cannabinoids: a neu‑ (2003) Maternal neuronal antibodies associated with autism and
roinflammatory story. Neurosci Biobehav Rev 121:128–143 a language disorder. Ann Neurol 53:533–537
Careaga M, Murai T, Bauman MD (2017) Maternal immune activa‑ Damkier P, Lassen D, Christensen MMH, Madsen KG, Hellfritzsch M,
tion and autism spectrum disorder: from rodents to nonhuman Pottegård A (2019) Interaction between warfarin and cannabis.
and human primates. Biol Psychiatry 81:391–401 Basic Clin Pharmacol Toxicol 124:28–31
Carlini EA (2010) The research on marijuana in Brazil. Braz J Psy‑ De Souza Crippa JA, Zuardi AW, Garrido GEJ, Wichert-Ana L,
chiatry 32:53–54 Guarnieri R, Ferrari L et al (2004) Effects of cannabidiol (CBD)

13
2730 Psychopharmacology (2022) 239:2713–2734

on regional cerebral blood flow. Neuropsychopharmacology basolateral amygdala–nucleus accumbens circuit modulates
29:417–426 sociability. J Clin Invest 130:1728–1742
Devane WA (1994) New dawn of cannabinoid pharmacology. Trends Franco V, Perucca E (2019) Pharmacological and therapeutic proper‑
Pharmacol Sci 15:40–41 ties of cannabidiol for epilepsy. Drugs 79:1435–1454
Devane WA, Hanuš L, Breuer A, Pertwee RG, Stevenson LA et al Fride E, Gobshtis N, Dahan H, Weller A, Giuffrida A, Ben-Shabat S
(1992) Isolation and structure of a brain constituent that binds to (2009) Chapter 6 the endocannabinoid system during develop‑
the cannabinoid receptor. Science 258:1946–1949 ment: emphasis on perinatal events and delayed effects. Vitam
Di Marzo V (2020) The endocannabinoidome as a substrate for none‑ Horm 81:139–158
uphoric phytocannabinoid action and gut microbiome dysfunc‑ Gaston TE, Szaflarski JP (2018) Cannabis for the treatment of epilepsy:
tion in neuropsychiatric disorders. Dialogues Clin Neurosci an update. Curr Neurol Neurosci Rep 18:73
22:259–269 Giraud C, Tran A, Rey E, Vincent J, Tréluyer J-M, Pons G (2004)
Di Marzo V, Maccarrone M (2008) FAAH and anandamide: is 2-AG In vitro characterization of clobazam metabolism by recombi‑
really the odd one out? Trends Pharmacol Sci 29:229–233 nant cytochrome P450 enzymes: importance of CYP2C19. Drug
Dinh TP, Freund TF, Piomelli D (2002) A role for monoglyceride Metab Dispos 32:1279–1286
lipase in 2-arachidonoylglycerol inactivation. Chem Phys Lipids Gogolla N, LeBlanc JJ, Quast KB, Südhof TC, Fagiolini M, Hensch
121:149–158 TK (2009) Common circuit defect of excitatory-inhibitory bal‑
Doenni VM, Gray JM, Song CM, Patel S, Hill MN, Pittman QJ (2016) ance in mouse models of autism. J Neurodev Disord 1:172–181
Deficient adolescent social behavior following early-life inflam‑ Gomes FV, Issy AC, Ferreira FR, Viveros MP, Del Bel EA, Guimarães
mation is ameliorated by augmentation of anandamide signaling. FS (2015) Cannabidiol attenuates sensorimotor gating disruption
Brain Behav Immun 58:237–247 and molecular changes induced by chronic antagonism of NMDA
Domschke K, Dannlowski U, Ohrmann P, Lawford B, Bauer J, Kugel receptors in Mice. Int J Neuropsychopharmacol 18:1–10
H et al (2008) Cannabinoid receptor 1 (CNR1) gene: impact on Gomes FV, Llorente R, Del Bel EA, Viveros MP, López-Gallardo
antidepressant treatment response and emotion processing in M, Guimarães FS (2015) Decreased glial reactivity could be
Major Depression. Eur Neuropsychopharmacol 18:751–759 involved in the antipsychotic-like effect of cannabidiol. Schizo‑
Dos-Santos-Pereira M, Guimarães FS, Del-Bel E, Raisman-Vozari R, phr Res 164:155–163
Michel PP (2020) Cannabidiol prevents LPS-induced microglial Guimarães FS, Chiaretti TM, Graeff FG, Zuardi AW (1990) Antianxi‑
inflammation by inhibiting ROS/NF-κB-dependent signaling and ety effect of cannabidiol in the elevated plus-maze. Psychophar‑
glucose consumption. Glia 68:561–573 macology 100:558–559
Egertová M, Giang DK, Cravatt BF, Elphick MR (1998) A new per‑ Gururajan A, Taylor DA, Malone DT (2011) Effect of cannabidiol in a
spective on cannabinoid signaling: complementary localization MK-801-rodent model ofaspects of schizophrenia. Behav Brain
of fatty acid amide hydrolase and the CB1 receptor in rat brain. Res 222:299–308
Proc R Soc B Biol Sci 265:2081–2085 Harkany T, Keimpema E, Barabás K, Mulder J (2008) Endocannabi‑
Elmes SJR, Jhaveri MD, Smart D, Kendall DA, Chapman V (2004) noid functions controlling neuronal specification during brain
Cannabinoid CB2 receptor activation inhibits mechanically development. Mol Cell Endocrinol 286:84–90
evoked responses of wide dynamic range dorsal horn neurons Hartmann A, Lisboa SF, Sonego AB, Coutinho D, Gomes FV, Guimarães
in naïve rats and in rat models of inflammatory and neuropathic FS (2019) Cannabidiol attenuates aggressive behavior induced by
pain. Eur J Neurosci 20:2311–2320 social isolation in mice: involvement of 5-HT1A and CB1 recep‑
Englund A, Morrison PD, Nottage J, Hague D, Kane F, Bonaccorso S tors. Prog Neuro-Psychopharmacology Biol Psychiatry 94:109637
et al (2013) Cannabidiol inhibits THC-elicited paranoid symp‑ Herbert MR (2010) Contributions of the environment and environmen‑
toms and hippocampal-dependent memory impairment. J Psy‑ tally vulnerable physiology to autism spectrum disorders. Curr
chopharmacol 27:19–27 Opin Neurol 23:103–110
Enstrom AM, Onore CE, Van de Water JA, Ashwood P (2010) Dif‑ Ho KW, Ward NJ, Calkins DJ (2012) TRPV1: a stress response protein
ferential monocyte responses to TLR ligands in children with in the central nervous system. Am J Neurodegener Dis 1:1–14
autism spectrum disorders. Brain Behav Immun 24:64–71 Horder J, Wilson CE, Mendez MA, Murphy DG (2014) Autistic traits
Erlander MG, Tobin AJ (1991) The structural and functional hetero‑ and abnormal sensory experiences in adults. J Autism Dev Dis‑
geneity of glutamic acid decarboxylase: A review. Neurochem ord 44:1461–1469
Res 16:215–226 Howlett AC, Blume LC, Dalton DG (2010) CB 1 Cannabinoid recep‑
Fatemi SH, Halt AR, Stary JM, Kanodia R, Schulz SC, Realmuto GR tors and their associated proteins. Curr Med Chem 17:1382–1393
(2002) Glutamic acid decarboxylase 65 and 67 kDa proteins are Hudson R, Rushlow W, Laviolette RS (2018) Phytocannabinoids mod‑
reduced in autistic parietal and cerebellar cortices. Biol Psychia‑ ulate emotional memory processing through interactions with
try 52:805–810 the ventral hippocampus and mesolimbic dopamine system:
Fattore L, Fratta W (2010) How important are sex differences in can‑ implications for neuropsychiatric pathology. Psychopharmacol‑
nabinoid action? Br J Pharmacol 160:544–548 ogy 235:447–458
Fleury-Teixeira P, Caixeta FV, da Silva LCR, Brasil-Neto JP, Malcher- Huestis MA, Gorelick DA, Heishman SJ, Preston KL, Nelson RA,
Lopes R (2019) Effects of cbd-enriched cannabis sativa extract Moolchan ET et al (2001) Blockade of effects of smoked mari‑
on autism spectrum disorder symptoms: an observational study juana by the CB1-selective cannabinoid receptor antagonist
of 18 participants undergoing compassionate use. Front Neurol SR141716. Arch Gen Psychiatry 58:322–328
10:1–9 Hunt GE, Large MM, Cleary M, Lai HMX, Saunders JB (2018) Preva‑
Fogaça MV, Campos AC, Coelho LD, Duman RS, Guimarães FS lence of comorbid substance use in schizophrenia spectrum dis‑
(2018) The anxiolytic effects of cannabidiol in chronically orders in community and clinical settings, 1990–2017: systematic
stressed mice are mediated by the endocannabinoid system: Role review and meta-analysis. Drug Alcohol Depend 191:234–258
of neurogenesis and dendritic remodeling. Neuropharmacology Iversen L (1994) Endogenous cannabinoids. Nature 372:619–619
135:22–33 Jackson AR, Nagarkatti P, Nagarkatti M (2014) Anandamide attenuates
Folkes MO, Báldi R, Kondev V, Marcus DJ, Hartley ND, Turner Th-17 cell-mediated delayed-type hypersensitivity response by
BD, Ayers JK, Baechle JJ, Misra MP, Altemus M, Grueter CA, triggering IL-10 production and consequent microRNA induc‑
Grueter BA, Patel S (2020) An endocannabinoid-regulated tion. PLoS ONE 9:e93954

13
Psychopharmacology (2022) 239:2713–2734 2731

Jiang W, Zhang Y, Xiao L, Van Cleemput J, Ji SP, Bai G, Zhang X Levy D, Ronemus M, Yamrom B, Lee Y, Leotta A, Kendall J et al
(2005) Cannabinoids promote embryonic and adult hippocam‑ (2011) Rare de novo and transmitted copy-number variation in
pus neurogenesis and produce anxiolytic- and antidepressant-like autistic spectrum disorders. Neuron 70:886–897
effects. J Clin Invest 115:3104–3116 Leweke FM, Piomelli D, Pahlisch F, Muhl D, Gerth CW, Hoyer C,
Jiang R, Yamaori S, Takeda S, Yamamoto I, Watanabe K (2011) Identi‑ Klosterkötter J, Hellmich M, Koethe D (2012) Cannabidiol
fication of cytochrome P450 enzymes responsible for metabolism enhances anandamide signaling and alleviates psychotic symp‑
of cannabidiol by human liver microsomes. Life Sci 89:165–170 toms of schizophrenia. Transl Psychiatry 2:e94
Jiang R, Yamaori S, Okamoto Y, Yamamoto I, Watanabe K (2013) Libbey JE, Sweeten TL, McMahon WM, Fujinami RS (2005) Autistic
Cannabidiol is a potent inhibitor of the catalytic activity of disorder and viral infections. J Neurovirol 11:1–10
cytochrome P450 2C19. Drug Metab Pharmacokinet 28:332–338 Linge R, Jiménez-Sánchez L, Campa L, Pilar-Cuéllar F, Vidal R, Pazos
Jiang HY, Xu LL, Shao L, Xia RM, Yu ZH, Ling ZX et al (2016) A et al (2016) Cannabidiol induces rapid-acting antidepressant-
Maternal infection during pregnancy and risk of autism spectrum like effects and enhances cortical 5-HT/glutamate neurotransmis‑
disorders: a systematic review and meta-analysis. Brain Behav sion: role of 5-HT1A receptors. Neuropharmacology 103:16–26
Immun 58:165–172 Litvin Y, Phan A, Hill MN, Pfaff DW, McEwen BS (2013) CB1 recep‑
Kaminski NE, Abood ME, Kessler FK, Martin BR, Schatz AR (1992) tor signaling regulates social anxiety and memory. Genes Brain
Identification of a functionally relevant cannabinoid receptor Behav 12:479–489
on mouse spleen cells that is involved in cannabinoid-mediated Lu J, Fan S, Zou G et al (2018) Involvement of glycine receptor a1
immune modulation. Mol Pharmacol 42:736–742 subunits in cannabinoid-induced analgesia. Neuropharmacology
Kaplan JS, Stella N, Catterall WA, Westenbroek RE (2017) Canna‑ 133:224–232
bidiol attenuates seizures and social deficits in a mouse model of Luna B, Marek S, Larsen B, Tervo-Clemmens B, Chahal R (2015) An
Dravet syndrome. Proc of the Natl Acad of Sci 114:11229–11234 integrative model of the maturation of cognitive control. Annu
Karhson DS, Krasinska KM, Dallaire JA, Libove RA, Phillips JM, Rev Neurosci 38:151–170
Chien AS et al (2018) Plasma anandamide concentrations are Lyall K, Ashwood P, Van de Water J, Hertz-Picciotto I (2014) Maternal
lower in children with autism spectrum disorder. Mol Autism immune-mediated conditions, autism spectrum disorders, and
9:1–6 developmental delay. J Autism Dev Disord 44:1546–1555
Kirsten TB, Taricano M, Maiorka PC, Palermo-Neto J, Bernardi MM Maccarrone M, Guzman M, Mackie K, Doherty P, Harkany T (2014)
(2010) Prenatal lipopolysaccharide reduces social behavior in Programming of neural cells by (endo)cannabinoids: from
male offspring. NeuroImmunoModulation 17:240–251 physiological rules to emerging therapies. Nat Rev Neurosci
Kiyono T, Morita M, Morishima R, Fujikawa S, Yamasaki S, Nishida 15:786–801
A, Ando S, Kasai K (2020) The prevalence of psychotic experi‑ Manduca A, Carbone E, Schiavi S, Cacchione C, Buzzelli V, Campo‑
ences in autism spectrum disorder and autistic traits: a systematic longo P, Trezza V (2021) The neurochemistry of social reward
review and meta-analysis. Schizophrenia Bulletin Open 1 during development: what have we learned from rodent models?
Knuesel I, Chicha L, Britschgi M, Schobel SA, Bodmer M, Hellings J Neurochem 157:1408–1435
JA et al (2014) Maternal immune activation and abnormal brain Marconi A, Di Forti M, Lewis CM, Murray RM, Vassos E (2016)
development across CNS disorders. Nat Rev Neurol 10:643–660 Meta-analysis of the association between the level of cannabis
Koskinen J, Löhönen J, Koponen H, Isohanni M, Miettunen J (2010) use and risk of psychosis. Schizophr Bull 42:1262–1269
Rate of cannabis use disorders in clinical samples of patients with Martin BR, Compton DR, Thomas BF, Prescott WR, Little PJ, Razdan
schizophrenia: a meta-analysis. Schizophr Bull 36:1115–11130 RK, Johnson MR, Melvin LS, Mechoulam R, Susan JW (1991)
Kozela E, Juknat A, Vogel Z (2017) Modulation of astrocyte activity Behavioral, biochemical, and molecular modeling evaluations
by cannabidiol, a nonpsychoactive cannabinoid. Int J Mol Sci of cannabinoid analogs. Pharmacol Biochem Behav 40:471–478
18:1669 Martin-Santos R, Crippa JA, Batalla A, Bhattacharyya S, Atakan Z,
Kuester G, Vergara K, Ahumada A, Gazmuri AM (2017) Oral can‑ Borgwardt S, Allen P, Seal M, Langohr K, Farré M, Zuardi AW,
nabis extracts as a promising treatment for the core symptoms McGuire PK (2012) Acute effects of a single, oral dose of d9-tet‑
of autism spectrum disorder: preliminary experience in Chilean rahydrocannabinol (THC) and cannabidiol (CBD) administration
patients. J Neurol Sci 381:932–933 in healthy volunteers. Curr Pharm Des 18:4966–4979
Laezza C, Pagano C, Navarra G, Pastorino O, Proto MC, Fiore D et al Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI (1990)
(2020) The endocannabinoid system: a target for cancer treat‑ Structure of a cannabinoid receptor and functional expression of
ment. Int J Mol Sci 21:747 the cloned cDNA. Nature 346:561–564
Lai MC, Baron-Cohen S (2015) Identifying the lost generation of Matta SM, Hill-Yardin EL, Crack PJ (2019) The influence of neuroin‑
adults with autism spectrum conditions. Lancet Psychiatry flammation in Autism Spectrum Disorder. Brain Behav Immun
2:1013–1027 79:75–90
Lai M, Lombardo M, Baron-Cohen S (2014) Autism. Autism. Lancet McFadden K, Minshew NJ (2013) Evidence for dysregulation of axonal
383:896–910 growth and guidance in the etiology of ASD. Front Hum Neu‑
Laprairie RB, Bagher AM, Kelly ME, Denovan-Wright EM (2015) rosci 7:671
Cannabidiol is a negative allosteric modulator of the cannabinoid Mechoulam R, Shvo Y (1963) Hashish-I the structure of cannabidiol.
CB1 receptor. Br J Pharmacol 172:4790–4805 Tetrahedron 19:2073–2078
Large M, Sharma S, Compton MT, Slade T, Nielssen O (2011) Canna‑ Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky M, Kaminski NE,
bis use and earlier onset of psychosis: a systematic meta-analysis. Schatz AR et al (1995) Identification of an endogenous 2-mono‑
Arch Gen Psychiatry 68:555–651 glyceride, present in canine gut, that binds to cannabinoid recep‑
Lee JLC, Bertoglio LJ, Guimarães FS, Stevenson CW (2017) Can‑ tors. Biochem Pharmacol 50:83–90
nabidiol regulation of emotion and emotional memory process‑ Meier MH, Caspi A, Ambler A, Harrington H, Houts R, Keefe RS,
ing: relevance for treating anxiety-related and substance abuse McDonald K, Ward A, Poulton R, Moffitt TE (2012) Persistent
disorders. Br J Pharmacol 174:3242–3256 cannabis users show neuropsychological decline from childhood
Lenz KM, Nelson LH (2018) Microglia and beyond: Innate immune to midlife. Proc Natl Acad Sci 109:E2657–E2664
cells as regulators of brain development and behavioral function. Melancia F, Schiavi S, Servadio M, Cartocci V, Campolongo P,
Front Immunol 9:698 Palmery M, Pallottini V, Trezza V (2018) Sex-specific autistic

13
2732 Psychopharmacology (2022) 239:2713–2734

endophenotypes induced by prenatal exposure to valproic acid disruption induced by amphetamine. Psychopharmacology
involve anandamide signaling. Br J Pharmacol 175:3699–3712 232:3057–3065
Mirza R, Sharma B (2019) Beneficial effects of pioglitazone, a selec‑ Pertwee RG (2008) Ligands that target cannabinoid receptors in
tive peroxisome proliferator-activated receptor-γ agonist in pre‑ the brain: From THC to anandamide and beyond. Addict Biol
natal valproic acid-induced behavioral and biochemical autistic 13:147–159
like features in Wistar rats. Int J Dev Neurosci 76:6–16 Pilorge M, Fassier C, Le Corronc H, Potey A, Bai J, De Gois S,
Morales P, Hurst DP, Reggio PH (2017) Molecular targets of the phy‑ Delaby E, Assouline B, Guinchat V, Devillard F et al (2016)
tocannabinoids: a complex picture. Prog Chem Org Nat Prod Genetic and functional analyses demonstrate a role for
103:103–131 abnormal glycinergic signaling in autism. Mol Psychiatry
Moreira FA, Guimarães FS (2005) Cannabidiol inhibits the hyper‑ 21:936–945
locomotion induced by psychotomimetic drugs in mice. Eur J Piomelli D (2003) The molecular logic of endocannabinoid signaling.
Pharmacol 512:199–205 Nat Rev Neurosci 4:873–884
Morgan JT, Chana G, Pardo CA, Achim C, Semendeferi K, Buckwal‑ Poleg S, Golubchik P, Offen D, Weizman A (2019) Cannabidiol as a
ter J et al (2010) Microglial activation and increased microglial suggested candidate for treatment of autism spectrum disorder.
density observed in the dorsolateral prefrontal cortex in autism. Prog Neuropsychopharmacol Biol Psychiatry 8:90–96
Biol Psychiatry 68:368–376 Ponton JA, Smyth K, Soumbasis E, Llanos SA, Lewis M, Meerholz
Mostafavi M, Gaitanis J (2020) Autism spectrum disorder and medical WA, Tanguay RL (2020) A pediatric patient with autism spec‑
cannabis: review and clinical experience. Semin Pediatr Neurol trum disorder and epilepsy using cannabinoid extracts as comple‑
35:100833 mentary therapy: a case report. J Med Case Rep 14:1–7
Mottron L, Duret P, Mueller S, Moore RD, Forgeot d’Arc B, Jacque‑ Pretzsch CM, Freyberg J, Voinescu B, Lythgoe D, Horder J, Mendez
mont S et al (2015) Sex differences in brain plasticity: a new MA et al (2019) Effects of cannabidiol on brain excitation and
hypothesis for sex ratio bias in autism. Mol Autism 6:33 inhibition systems; a randomized placebo-controlled single dose
Munro S, Thomas KL, Abu-Shaar M (1993) Molecular characteriza‑ trial during magnetic resonance spectroscopy in adults with and
tion of a peripheral receptor for cannabinoids. Nature 365:61–65 without autism spectrum disorder. Neuropsychopharmacology
Murrie B, Lappin J, Large M, Sara G (2020) Transition of substance- 44:1398–1405
induced, brief, and atypical psychoses to schizophrenia: a sys‑ Pretzsch CM, Voinescu B, Mendez MA, Wichers R, Ajram L, Ivin G
tematic review and meta-analysis. Schizophr Bull 46:505–516 et al (2019) The effect of cannabidiol (CBD) on low-frequency
Musella A, De Chiara V, Rossi S, Cavasinni F, Castelli M, Cantarella activity and functional connectivity in the brain of adults with
C et al (2010) Transient receptor potential vanilloid 1 channels and without autism spectrum disorder (ASD). J Psychopharma‑
control acetylcholine/2-arachidonoylglicerol coupling in the col 33:1141–1148
striatum. Neuroscience 167:864–871 Purcell AE, Jeon OH, Pevsner J (2001) The abnormal regulation of
Nabbout R, Gennaro E, Dalla Bernardina B, Dulac O, Madia F, Bertini gene expression in autistic brain tissue. J Autism Dev Disord
E et al (2003) Spectrum of SCN1A mutations in severe myo‑ 31:545–549
clonic epilepsy of infancy. Neurology 60:1961–1967 Qin M, Zeidler Z, Moulton K, Krych L, Xia Z, Smith CB (2015)
Nackley AG, Makriyannis A, Hohmann AG (2003) Selective activa‑ Endocannabinoid-mediated improvement on a test of aversive
tion of cannabinoid CB2 receptors suppresses spinal Fos protein memory in a mouse model of fragile X syndrome. Behav Brain
expression and pain behavior in a rat model of inflammation. Res 291:164–171
Neuroscience 119:747–757 Rademacher L, Schulte-Ruther M, Hanewald B, Lammertz S (2017)
Nardo M, Casarotto PC, Gomes FV, Guimarães FS (2014) Cannabidiol Reward: from basic reinforcers to anticipation of social cues.
reverses the mCPP-induced increase in marble-burying behavior. Curr Top Behav Neurosci 30:207–221
Fundam Clin Pharmacol 28:544–550 Ramaswami G, Geschwind DH (2018) Genetics of autism spectrum
Nona CN, Hendershot CS, Le Foll B (2019) Effects of cannabidiol disorder. Handb Clin Neurol 147:321–329
on alcohol-related outcomes: a review of preclinical and human Renard J, Loureiro M, Rosen LG, Zunder J, de Oliveira C, Schmid
research. Exp Clin Psychopharmacol 27:359 S, Rushlow WJ, Laviolette SR (2016) Cannabidiol counteracts
Nutma E, Willison H, Martino G, Amor S (2019) Neuroimmunology amphetamine-induced neuronal and behavioral sensitization
– the past, present and future. Clin Exp Immunol 197:278–293 of the mesolimbic dopamine pathway through a novel mTOR/
O’Sullivan SE (2016) An update on PPAR activation by cannabinoids. p70S6 kinase signaling pathway. J Neurosci 36:5160–5169
Br J Pharmacol 173:1899–1910 Rivet TT, Matson JL (2011) Review of gender differences in core
Osborne AL, Solowij N, Babic I, Lum JS, Huang XF, Newell KA, symptomatology in autism spectrum disorders. Res Autism
Weston-Green K (2019) Cannabidiol improves behavioral and Spectr Disord 5:957–976
neurochemical deficits in adult female offspring of the maternal Robinson TE, Berridge KC (1993) The neural basis of drug craving:
immune activation (poly I:C) model of neurodevelopmental dis‑ an incentive-sensitization theory of addiction. Brain Res Rev
orders. Brain Behav Immun 81:574–587 18:247–291
Osborne AL, Solowij N, Babic I, Lum JS, Huang XF, Newell KA, Rodriguez-Arias M, Navarrete F, Daza-Losada M, Navarro D, Aguilar
Weston-Green K (2019) Effect of cannabidiol on endocannabi‑ MA, Berbel P, Miñarro J, Manzanares J (2013) CB1 cannabi‑
noid, glutamatergic and GABAergic signaling markers in male noid receptor-mediated aggressive behavior. Neuropharmacology
offspring of a maternal immune activation (poly I:C) model rel‑ 75:172–180
evant to schizophrenia. Prog Neuropsychopharmacol Biol Psy‑ Rossignol DA, Bradstreet JJ (2008) Evidence of mitochondrial dys‑
chiatry 95:109666 function in autism and implications for treatment. Am J Biochem
Patra PH, Serafeimidou-Pouliou E, Bazelot M, Whalley BJ, Williams Biotechnol 4:208–217
CM, McNeish AJ (2020) Cannabidiol improves survival and Rubenstein JLR, Merzenich MM (2003) Model of autism: increased
behavioral co-morbidities of Dravet syndrome in mice. Br J ratio of excitation/inhibition in key neural systems Genes. Brain
Pharmacol 177:2779–2792 Behav 2:255–267
Pedrazzi JFC, Issy AC, Gomes FV, Guimarães FS, Del-Bel Russo EB, Burnett A, Hall B, Parker KK (2005) Agonistic properties of
EA (2015) Cannabidiol effects in the prepulse inhibition cannabidiol at 5-HT1a receptors. Neurochem Res 30:1037–1043

13
Psychopharmacology (2022) 239:2713–2734 2733

Sagar DR, Jhaveri M, Chapman V (2009) Targeting the cannabi‑ controlled placebo clinical trial. Trends Psychiatry Psychother‑
noid system to produce Analgesia. Curr Top Behav Neurosci apy 26:44
1:275–287 Silvestro S, Mammana S, Cavalli E, Bramanti P, Mazzon E (2019)
Sahin M, Sur M (2015) Genes, circuits, and precision therapies for Use of cannabidiol in the treatment of epilepsy: efficacy and
autism and related neurodevelopmental disorders. Science security in clinical trials. Molecules 24:1459
350:6263 Siniscalco D, Sapone A, Cirillo A, Giordano C, Maione S, Anto‑
Sales AJ, Fogaça MV, Sartim AG, Pereira VS, Wegener G, Guimarães nucci N (2012) Autism spectrum disorders: is mesenchymal
FS, Joca SRL (2019) Cannabidiol induces rapid and sustained stem cell personalized therapy the future? J Biomed Biotechnol
antidepressant-like effects through increased BDNF signaling 2012:480289
and synaptogenesis in the prefrontal cortex. Mol Neurobiol Siniscalco D, Sapone A, Giordano C, Cirillo A, de Magistris L, Rossi
56:1070–1081 F, Fasano A, Bradstreet JJ, Maione S, Antonucci N (2013)
Santana N, Bortolozzi A, Serrats J, Mengod G, Artigas F (2004) Cannabinoid receptor type 2, but not type 1, is up-regulated
Expression of serotonin1A and serotonin2A receptors in pyrami‑ in peripheral blood mononuclear cells of children affected by
dal and GABAergic neurons of the rat prefrontal cortex. Cereb autistic disorders. J Autism Dev Disord 43:2686–2695
Cortex 14:1100–1109 Smith DR, Stanley CM, Foss T, Boles RG, McKernan K (2017) Rare
Satterstrom FK, Walters RK, Singh T, Wigdor EM, Lescai F et al genetic variants in theendocannabinoid system genes CNR1
(2019) Autism spectrum disorder and attention deficit hyperac‑ and DAGLA are associated with neurological phenotypes in
tivity disorder have a similar burden of rare protein-truncating humans. PLoS One 12:e0187926
variants. Nat Neurosci 22:1961–1965 Sonego AB, Prado DS, Vale GT, Sepulveda-Diaz JE, Cunha TM,
Sawzdargo M, Nguyen T, Lee DK, Lynch KR, Cheng R, Heng HH, Tirapelli CR et al (2018) Cannabidiol prevents haloperidol-
George SR, O’Dowd BF (1999) Identification and cloning of induced vacuos chewing movements and inflammatory changes
three novel human G protein-coupled receptor genes GPR52, in mice via PPARγ receptors. Brain Behav Immun 74:241–251
YGPR53 and GPR55: GPR55 is extensively expressed in human Sorkhou M, Bedder RH, George TP (2021) The behavioral sequelae
brain. Mol Brain Res 64:193–198 of cannabis use in healthy people: a systematic review. Front
Schleider LBL, Mechoulam R, Saban N, Meiri G, Novack V (2019) Psychiatry 12:122
Real life experience of medical cannabis treatment in autism: South M, Rodgers J (2017) Sensory, emotional and cognitive con‑
analysis of safety and efficacy. Sci Rep 9:1–7 tributions to anxiety in autism spectrum disorders. Front Hum
Schoeler T, Monk A, Sami MB, Klamerus E, Foglia E et al (2016) Neurosci 11:20
Continued versus discontinued cannabis use in patients with psy‑ Stark T, Di Bartolomeo M, Di Marco R, Drazanova E, Platania
chosis: a systematic review and meta-analysis. Lancet Psychiatry CBM, Iannotti FA et al (2020) Altered dopamine D3 receptor
3:215–225 gene expression in MAM model of schizophrenia is reversed
Schroeder JH, Desrocher M, Bebko JM, Cappadocia MC (2010) The by peripubertal cannabidiol treatment. Biochem Pharmacol
neurobiology of autism: Theoretical applications. Res Autism 177:114004
Spectr Disord 4:555–564 Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, Itoh K
Schubart CD, Sommer IEC, Fusar-Poli P, de Witte L, Kahn RS, Boks et al (1995) 2-arachidonoylglycerol: a possible endogenous
MPM (2014) Cannabidiol as a potential treatment for psychosis. cannabinoid receptor ligand in brain. Biochem Biophys Res
Eur Neuropsychopharmacol 24:51–64 Commun 215:89–97
Schwartz TL, Sachdeva S, Stahl SM (2012) Glutamate neurocircuitry: Suzuki K, Sugihara G, Ouchi Y, Nakamura K, Futatsubashi M, Take‑
theoretical underpinnings in schizophrenia. Front Pharmacol bayashi K et al (2013) Microglial activation in young adults
3:195 with autism spectrum disorder. Arch Gen Psychiatry 70:49–58
Scuderi C, De Filippis D, Iuvone T, Blasio A, Steardo A, Esposito Swanson LW, Sawchenko PE (1983) Hypothalamic integration:
G (2009) Cannabidiol in medicine: a review of its therapeutic organization of the paraventricular and supraoptic nuclei. Annu
potential in CNS disorders. Phyther Res 23:597–602 Rev Neurosci 6:269–324
Seeman P (2016) Cannabidiol is a partial agonist at dopamine D2High Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee
receptors, predicting its antipsychotic clinical dose. Transl Psy‑ RG (2007) Cannabidiol displays unexpectedly high potency as
chiatry 6:e920 an antagonist of CB 1 and CB 2 receptor agonists in vitro. Br
Sekar K, Pack A (2019) Epidiolex as adjunct therapy for treatment J Pharmacol 150:613–623
of refractory epilepsy: a comprehensive review with a focus on Toscano CVA, Barros L, Lima AB, Nunes T, Carvalho HM, Gaspar
adverse effects. Research 8:F1000 JM (2021) Neuroinflammation in autism spectrum disorders:
Servadio M, Melancia F, Manduca A, Di Masi A, Schiavi S, Cartocci exercise as a “pharmacological” tool. Neurosci Biobehav Ver
V et al (2016) Targeting anandamide metabolism rescues core 129:63–74
and associated autistic-like symptoms in rats prenatally exposed Trazzi S, Steger M, Mitrugno VM, Bartesaghi R, Ciani E (2010) CB1
to valproic acid. Transl Psychiatry 6:e902 cannabinoid receptors increase neuronal precursor prolifera‑
Shi Q, Yang L, Shi W, Wang L, Zhou S, Guan S, Zhao M, Yang Q tion through AKT/glycogen synthase kinase-3beta/beta-catenin
(2017) The novel cannabinoid receptor GPR55 mediates anxio‑ signaling. J Biol Chem 285:10098–10109
lytic-like effects in the medial orbital cortex of mice with acute Valvassori SS, Elias G, De Souza B, Petronilho F, Dal-Pizzol F,
stress. Mol Brain 10:38 Kapczinski F et al (2011) Effects of cannabidiol on ampheta‑
Silva NR, Gomes FV, Sonego AB, Silva NRD, Guimarães FS (2020) mine-induced oxidative stress generation in an animal model
Cannabidiol attenuates behavioral changes in a rodent model of of mania. J Psychopharmacol 25:274–279
schizophrenia through 5-HT1A, but not CB1 and CB2 receptors. Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, Pardo
Pharmacol Res 156:104749 CA (2005) Neuroglial activation and neuroinflammation in the
Silva EADJ, Medeiros WMB, Santos JPMD, Sousa JMM, Costa FBD, brain of patients with autism. Ann Neurol 57:67–81
Pontes KM, Borges TC et al (2022) Evaluation of the efficacy Volkmar FR, Lord C, Bailey A, Schultz RT, Klin A (2004) Autism
and safety of cannabidiol-rich cannabis extract in children and pervasive developmental disorders. J Child Psychol Psy‑
with autism spectrum disorder: randomized, double-blind and chiatry Allied Discip 45:135–170

13
2734 Psychopharmacology (2022) 239:2713–2734

Walzer M, Bekersky I, Blum RA, Tolbert D (2012) Pharmacokinetic Zamberletti E, Gabaglio M, Parolaro D (2017) The endocannabinoid
drug interactions between clobazam and drugs metabolized by system and autism spectrum disorders: insights from animal
cytochrome P450 isoenzymes. Pharmacotherapy 32:340–353 models. Int J Mol Sci 18:1916
Warren Z et al (2012) Therapies for children with autism spectrum Zendulka O, Dovrtelová G, Nosková K, Turjap M, Šulcová A, Hanuš L,
disorders. Comp Eff Rev 85:878–879 Juřica J et al (2016) Cannabinoids and cytochrome P450 interac‑
Watson S, Chambers D, Hobbs C, Doherty P, Graham A (2008) The tions. Curr Drug Metab 17:206–226
endocannabinoid receptor, CB1, is required for normal axonal Zhang J, Hoffert C, Vu HK, Groblewski T, Ahmad S, O’Donnell D
growth and fasciculation. Mol Cell Neurosci 38:89–97 (2003) Induction of CB2 receptor expression in the rat spinal
Wei D, Lee D, Cox CD, Karsten CA, Peñagarikano O, Geschwind DH cord of neuropathic but not inflammatory chronic pain models.
et al (2015) Endocannabinoid signaling mediates oxytocin-driven Eur J Neurosci 17:2750–2754
social reward. Proc Natl Acad Sci U S A 112:14084–14089 Zheng Z, Zheng P, Zou X (2018) Association between schizophrenia
Wei D, Dinh D, Lee D, Li D, Anguren A, Moreno-Sanz G et al (2016) and autism spectrum disorder: a systematic review and meta-
Enhancement of anandamide-mediated endocannabinoid signal‑ analysis. Autism Res 11:1110–1119
ing corrects autism-related social impairment. Cannabis Can‑ Zuardi AW, Shirakawa I, Finkelfarb E, Karniol IG (1982) Action of
nabinoid Res 1:81–89 cannabidiol on the anxiety and other effects produced by δ9-THC
Wei D, Allsop S, Tye K, Piomelli D (2017) endocannabinoid signaling in normal subjects. Psychopharmacology 76:245–250
in the control of social behavior. Trends Neurosci 40:385–396 Zuardi AW, Antunes Rodrigues J, Cunha JM (1991) Effects of can‑
Winden KD, Ebrahimi-Fakhari D, Sahin M (2018) Abnormal mTOR nabidiol in animal models predictive of antipsychotic activity.
activation in autism. Annu Rev Neurosci 41:1–23 Psychopharmacology 104:260–264
Yamashita Y, Fujimoto C, Nakajima E, Isagai T, Matsuishi T (2003) Zuardi AW, Cosme RA, Graeff FG et al (1993) Effects of ipsapirone
Possible association between congenital cytomegalovirus infec‑ and cannabidiol on human experimental anxiety. J Psychophar‑
tion and autistic disorder. J Autism Dev Disord 33:455–459 macoly 7:82–88
Yang L, Rozenfeld R, Wu D, Devi LA, Zhang Z, Cederbaum A (2014) Zwaigenbaum L, Bauman ML, Fein D, Pierce K, Buie T, Davis PA
Cannabidiol protects liver from binge alcohol-induced steato‑ et al (2015) Early screening of autism spectrum disorder: recom‑
sis by mechanisms including inhibition of oxidative stress and mendations for practice and research. Pediatrics 136:S41–S59
increase in autophagy. Free Radic Biol Med 68:260–267
Young LJ, Barrett CE (2015) Can oxytocin treat autism? Science Publisher's note Springer Nature remains neutral with regard to
347:825–826 jurisdictional claims in published maps and institutional affiliations.

13

You might also like