ArticlePDF Available

Anxiolytic Effects of Repeated Cannabidiol Treatment in Teenagers With Social Anxiety Disorders

Frontiers
Frontiers in Psychology
Authors:

Abstract and Figures

Accumulated evidence indicates that cannabidiol (CBD), a nonpsychotomimetic and nonaddictive main component of the Cannabis sativa plant, reverses anxiety-like behavior. The purpose of the present study was to assess the efficacy of CBD treatment for Japanese late teenagers with social anxiety disorder (SAD). Thirty-seven 18–19-year-old Japanese teenagers with SAD and avoidant personality disorder received, in a double-blind study, cannabis oil (n = 17) containing 300 mg CBD or placebo (n = 20) daily for 4 weeks. SAD symptoms were measured at the beginning and end of the treatment period using the Fear of Negative Evaluation Questionnaire and the Liebowitz Social Anxiety Scale. CBD significantly decreased anxiety measured by both scales. The results indicate that CBD could be a useful option to treat social anxiety.
This content is subject to copyright.
Frontiers in Psychology | www.frontiersin.org 1 November 2019 | Volume 10 | Article 2466
ORIGINAL RESEARCH
published: 08 November 2019
doi: 10.3389/fpsyg.2019.02466
Edited by:
Changiz Mohiyeddini,
Northeastern University,
UnitedStates
Reviewed by:
Francisco Silveira Guimaraes,
University of São Paulo, Brazil
Sagnik Bhattacharyya,
King’s College London,
UnitedKingdom
*Correspondence:
Nobuo Masataka
masataka.nobuo.7r@kyoto-u.ac.jp;
masataka@pri.kyoto-u.ac.jp
Specialty section:
This article was submitted to
Psychology for Clinical Settings,
a section of the journal
Frontiers in Psychology
Received: 14 June 2019
Accepted: 18 October 2019
Published: 08 November 2019
Citation:
Masataka N (2019) Anxiolytic
Effects of Repeated Cannabidiol
Treatment in Teenagers With Social
Anxiety Disorders.
Front. Psychol. 10:2466.
doi: 10.3389/fpsyg.2019.02466
Anxiolytic Effects of Repeated
Cannabidiol Treatment in Teenagers
With Social Anxiety Disorders
NobuoMasataka*
Primate Research Institute, Kyoto University, Inuyama, Japan
Accumulated evidence indicates that cannabidiol (CBD), a nonpsychotomimetic and
nonaddictive main component of the Cannabis sativa plant, reverses anxiety-like behavior.
The purpose of the present study was to assess the efcacy of CBD treatment for Japanese
late teenagers with social anxiety disorder (SAD). Thirty-seven 18–19-year-old Japanese
teenagers with SAD and avoidant personality disorder received, in a double-blind study,
cannabis oil (n=17) containing 300mg CBD or placebo (n=20) daily for 4weeks. SAD
symptoms were measured at the beginning and end of the treatment period using the
Fear of Negative Evaluation Questionnaire and the Liebowitz Social Anxiety Scale. CBD
signicantly decreased anxiety measured by both scales. The results indicate that CBD
could bea useful option to treat social anxiety.
Keywords: cannabidiol, social anxiety disorder, cannabis, cannabinoid, social phobia, avoidant personality disorder,
social withdrawal
INTRODUCTION
e primary noneuphorizing and nonaddictive compound of cannabis, cannabidiol (CBD),
has recently been shown to possess considerable therapeutic potential for treating a wide
range of neuropsychiatric disorders (De Gregorio et al., 2019). ey include chronic pain
(Costa et al., 2007), nausea (Parker et al., 2006), epilepsy (Devinsky et al., 2016), psychosis
(McGuire et al., 2018), and anxiety (Scuderi et al., 2009; Whiting et al., 2015). CBD in
therapeutics is used within a large therapeutic window, which ranges from 2.85 to 50 mg/
kg/day (Whiting et al., 2015; Devinsky et al., 2016). While this fact indicates that its
therapeutic dose is still mostly unknown, clinical studies have revealed that CBD could
produce analgesic and anxiolytic eects exerted through its interaction with 5HT1A receptors
(De Gregorio et al., 2019). As a potential anxiolytic treatment, in particular, it has drawn
increasing interest. A review (Blessing etal., 2015) concluded that existing preclinical evidence
strongly supports CBD as a treatment for generalized anxiety disorder, panic disorder,
obsessive-compulsive disorder, and posttraumatic disorder when administered acutely.
Clinical data showing therapeutic eects of CBD in patients with anxiety disorders, however,
are still meager (Bergamaschi et al., 2011; Crippa et al., 2011). e purpose of the present
study was to investigate these eects in patients with social anxiety disorder (SAD).
SAD is characterized by excessive anxiety in situations where a person might feel judged,
such as performance situations, and situations involving interpersonal contact with others (American
Psychiatric Association, 2000). is is the fear of social situations that may cause humiliation
or embarrassment. While it is one of the common anxiety disorders (Craske et al., 2017), it is
a relatively new area of research, and thus, its etiology, eects, and treatment are not clearly
Masataka Cannabidiol and Social Anxiety
Frontiers in Psychology | www.frontiersin.org 2 November 2019 | Volume 10 | Article 2466
understood. Even prevalence rates reported in the literature vary
across studies (Antony and Rowa, 2008). For instance, lifetime
prevalence estimates for SAD based on large community samples
in the United States range from 3 to 13%. In addition, some
studies report that SAD has a higher incidence in females than
in males (Kessler et al., 2005).
Developmentally, SAD is likely to not only begin in adolescence
(mid to late teens) but can also occur earlier in childhood
(Somers et al., 2006). A signicant number of adults report
that they have had problems with social anxiety for their entire
lives or as long as they can remember (Brown et al., 2001;
Masataka, 2003). A large-scale study of individuals presenting
at an anxiety clinic found a mean age of onset of 15.7 years,
a number that was younger than the onset of other anxiety
disorders (Merikangas et al., 2011).
SAD is best treated with psychotropic medication and
cognitive behavioral therapies (CBT), and the most eective
treatments are a combination of both (Nordahl et al., 2016).
ey consist of monoamine oxidase inhibitors, the serotonin
reuptake inhibitors, benzodiazepines, and individual cognitive
behavioral therapy. CBT typically includes 10–15 weekly sessions
and consists of a variety of strategies, such as self-monitoring,
psychoeducation, cognitive therapy, exposure-based techniques,
and social skills training. While this method has been proven
to be eective for SAD if it is executed, it is also true that
people with the disorder quite oen show unwillingness (Ryan
and Warner, 2012) to receive CBT. In fact, a study reported
that 92% of individuals with SAD expressed concerns about
starting treatment and that is the biggest barrier to treatment
that should be overcome (Kessler et al., 2005).
In this regard, preliminary ndings reported by a study
(Bergamaschi et al., 2011) that investigated the ecacy of CBD
with patients with SAD are noteworthy. In that study, 12 patients
with SAD were provided with a single dose of CBD (600 mg).
When the anxiety induced by simulated public speaking was
compared between pretreatment and posttreatment, its level
showed a signicant decrease aer the treatment, whereas no
such change was observed in the placebo group of 12 other patients.
e purpose of the current study was to pursue this issue
further and to investigate the possible ecacy of CBD as at
least an adjunctive option for intervention in people with SAD.
While SAD has been classied into several subtypes so far (Antony
and Rowa, 2008), here, the author concentrated the research on
that with avoidant personality disorder because this subtype is
the most commonly diagnosed and is becoming a serious social
problem in Japan, where the current study was conducted (Ogino,
2004; Saito, 2007; Teo, 2010). e author attempted to systematically
assess the ecacy of CBD in a total of 37 18–19-year-old Japanese
with SAD, all of whom had been naive to any form of treatment,
by measuring the level of the symptoms of SAD with both the
Fear of Negative Evaluation Questionnaire (FNE; Watson and
Friend, 1969) and the Liebowitz Social Anxiety Scale (LSAS;
Liebowitz, 1987) using an exploratory double-blind parallel-group
trial experimental paradigm.
FNE is a 30-item measure of apprehension and anxiety over
anticipated social evaluations. is measure uses a true-false scale
and is known to show good internal consistency and test-retest
reliability (Watson and Friend, 1969). FNE has a range from 0
to 30, with high scores indicating higher levels of social anxiety.
LSAS is a short questionnaire to assess the range of social
interaction and performance situations feared by a person in
order to assist in the diagnosis of SAD (Liebowitz, 1987). It has
been commonly used to investigate outcomes in clinical trials
and, more recently, to evaluate the eectiveness of psychological
treatments (De Gregorio etal., 2019). e scale features 24 items,
which are divided into two subscales. irteen questions relate
to performance, and 11 relate to situations.
e author attempted to assess the ecacy of CBD by
comparing the FNE and the LSAS scores measured before the
commencement of the 4-week-long intervention (preintervention)
and the scores measured aer the completion of the intervention
(postintervention) in the group of participants who were provided
with CBD (the CBD group) and in the group whose participants
were provided with placebo (the placebo group). e author
hypothesized that CBD would signicantly decrease anxiety
measured by both of the two scales employed.
MATERIALS AND METHODS
Ethics Statement
is investigation was conducted according to the principles
expressed in the Declaration of Helsinki. All experimental
protocols were consistent with the Guide for Experimentation
with Humans and were approved by the Institutional Ethics
Committee of Kyoto University (#2018-150), the regional
committee for medical and health research ethics (#2018/1783),
and the Japanese Data Inspectorate for clinical trials
(JCT0018004564). e author obtained written informed consent
from all of the participants involved in the study. Written
informed consent was additionally obtained from the parent/
legal guardian of all participants who were younger than the
age of consent at the time of the study.
Design and Participants
A randomized, placebo-controlled, comparative study with a
total of 37 Japanese adolescents was undertaken. Double masking
was conducted, and the participants and the investigator were
blinded regarding which condition (CBD oil or placebo) under
which each participant was studied.
At the commencement of the study, 40 teenagers with SAD
participated, and 20 of them were assigned to the CBD group
and the other 20 to the placebo group. is sample size was
determined because the current study had been approved by the
ethics committee on the condition that, as a pilot study, no more
than 20 teenagers take CBD oil. Of the 40 participants, three in
the CBD group declined daily treatment with CBD oil during
the study because they disliked the smell and the taste of the oil.
In all, 26 males and 11 females 18–19-year olds were included
in the study (12 males and 5 females for the CBD group and
14 males and 6 females for the placebo group). ey were all
naive to cannabis and diagnosed by psychiatrists in several
hospitals located in the vicinity of Osaka Prefecture, Japan, using
Masataka Cannabidiol and Social Anxiety
Frontiers in Psychology | www.frontiersin.org 3 November 2019 | Volume 10 | Article 2466
the Structured Clinical Interview for DSM-IV Axis I Disorders
(SCID-I/P; Di Nardo etal., 1994) and Axis II Personality Disorders
(First et al., 1997). For all of the participants, symptoms had
lasted for at least 6 months at the commencement of the study.
Exclusion criteria were experience of receiving previous or
concurrent psychological or drug treatment, any form of psychotic
or organic illness, diagnosis of cluster A or B personality disorder,
acute suicidality, and drug and alcohol dependence. ey have
not had a comorbid diagnosis of other anxiety or mood disorders.
Assessment
e participants were invited to attend an assessment interview
for possible participation in the present study. None of them
were under CBT. Assessment of all of them was undertaken by
psychiatrists who were trained in administering the Structured
Clinical Interview for DSM-IV (SCID I and II; Liebowitz, 1987;
Di Nardo etal., 1994; First etal., 1997). e participants completed
the rst battery of self-report measures before attending the
assessment interview. e baseline period for them was a minimum
of 3weeks showing stable FNE scores (Watson and Friend, 1969;
the primary outcome measure). Subsequently, aer the assessment
interview, they rated themselves on the FNE and LSAS (Liebowitz,
1987) over the succeeding weeks (preintervention). All of the
participants had a stable FNE score over the 3 consecutive weeks
and were therefore scheduled for intervention within a week
aer the third-baseline measuring point.
When the 4-week-intervention ended, the FNE and LSAS
were completed again by each participant (postintervention),
and their scores were compared with those recorded at
preintervention. en, the SCID I and II were administered
again by the same psychiatrists who met the participants
at preintervention.
Aer the completion of the intervention, at the follow-up,
the clinical psychologists who had been responsible for the
intervention visited the participants briey at their home once
a week to check for any eect of it on their health. is
follow-up continued for up to a 6-month-period.
Intervention
All of the participants were randomly assigned into either
the CBD group, in which they were to daily receive 300 mg
of CBD administered in a single dose in the aernoon, or
the placebo group, in which they were to daily receive a
matching placebo. e CBD dose was based on that used in
the rst study showing acute anxiolytic eects in healthy
subjects exposed to a simulated public speaking test (Zuardi
etal., 1993). is assignment was conducted by an independent
statistician who did not know about the purpose of the present
research. e CBD used was RSHO-X Hemp Oil (the product
of HempMeds, USA) that was produced from the stalk of
hemp plants. A 236-ml bottle of the product that was for
sale by the company contained 5,000 mg of CBD (21.4 mg/
ml) but no delta-9-tetrahydrocannabinol (THC). It did not
contain other cannabinoids or terpenes.
e placebo contained olive oil. e CBD oil containing
300 mg of CBD or the equivalent amount of the placebo
was administered orally to each of the participants of the
CBD group and each of the participants of the placebo group,
respectively. For each participant, roughly 420 ml of the
CBD oil or the same amount of the placebo was rebottled
in a container that was dierent from that in which it had
been originally bottled and that was identical in size and
color as well as appearance to the oil administered to the
other group.
e container was provided to a clinical psychologist who
was employed by the principal investigator and was
predetermined to be responsible for each participant. e
psychologist who did not know whether the bottle contained
CBD or not visited the home of the participant with the
container every aernoon and administered the necessary
amount of the prepared oil to the participant, using a syringe,
during the 4-week intervention period.
While CBD oil had a characteristic smell and taste, all of
the psychologists and the participants had been naive to the
CBD oil as well as the placebo. e interview with them that
was conducted aer the completion of the study revealed that
none of them noticed the dierence between the two.
RESULTS
e results of the measurements with FNE of the level of
the symptoms that were associated with SAD are shown in
Figure1. When the collected data were analyzed by a 2 (period
of measurements: preintervention versus postintervention,
MEASUREMENT) × 2 (participant group: the CBD group
versus the Placebo group, PARTICIPANT) repeated-measures
ANOVA (analysis of variance), the main eect was statistically
signicant for MEASUREMENT (F1,35 = 10.35, p = 0.003,
h
p
2=0.0228) but not for PARTICIPANT (F1,35=2.69, p=0.11,
h
p
2 = 0.071). e interaction between these factors was
signicant (F1,35 = 44.81, p < 0.001,
h
p
2 = 0.561). e mean
FNE score (SD) of the CBD group was 24.4 (2.7) in the
preintervention measurement and 19.1 (2.1) in the
postintervention measurement and that of the placebo group
was 23.5 (2.1) in the preintervention measurement and 23.3
(2.9) in the postintervention measurement.
Subsequent analyses of simple main effects (using
Bonferroni correction), which were performed because of
the significant interactions between MEASUREMENT and
PARTICIPANT, revealed that the mean score of the CBD
group was lower in the postintervention measurement than
in the preintervention measurement (p = 0.02), while no
such difference was found in the placebo group (p =0.29).
Scores of the participants in the CBD group were lower
than those of the placebo group in the postintervention
measurement (p=0.0002), but the scores were not statistically
significantly different from one another in the preintervention
measurement (p = 0.71).
Figure 2 presents the results of the measurements with LSAS
of the level of the symptoms that are associated with SAD. e
results were strikingly similar to those shown in Figure 1. e
main eect was statistically signicant for MEASUREMENT
Masataka Cannabidiol and Social Anxiety
Frontiers in Psychology | www.frontiersin.org 4 November 2019 | Volume 10 | Article 2466
(F1,35= 10.35, p= 0.003,
h
p
2=0.023) but not for PARTICIPANT
(F1,35 = 0.45, p = 0.57,
h
p
2 = 0.011). e interaction between
these factors was signicant (F1,35=39.16, p<0.001,
h
p
2= 0.528).
e mean LSAS score (SD) of the CBD group was 74.2 (7.5)
in the preintervention measurement and 62.1 (8.7) in the
postintervention measurement and that of the placebo group
was 69.9 (10.3) in the preintervention measurement and 66.8
(11.2) in the postintervention measurement.
Another post hoc test revealed that the mean LSAS score
of the CBD group was lower in the postintervention measurement
than in the preintervention measurement (p = 0.03), but no
such dierence was found in the placebo group (p = 0.42).
FIGURE 1 | Scores of Fear of Negative Evaluation Questionnaire (FNE) in the participants who received the intervention with cannabidiol (CBD; n=17) and in the
participants who received the intervention with placebo (Placebo; n=20). The participants were evaluated before (Pre) and after (Post) treatment. Error bars
represent SDs. * indicates signicant difference from pretreatment measurement.
FIGURE 2 | Scores of Liebowitz Social Anxiety Scale (LSAS) in the participants who received the intervention with cannabidiol (CBD; n=17) and in the participants
who received the intervention with placebo (Placebo; n=20). The participants were evaluated before (Pre) and after (Post) treatment. Error bars represent SDs.
*indicates signicant difference from pretreatment measurement.
Masataka Cannabidiol and Social Anxiety
Frontiers in Psychology | www.frontiersin.org 5 November 2019 | Volume 10 | Article 2466
Scores of the participants in the CBD group were smaller
than those of the placebo group in the postintervention
measurement (p = 0.0018), but the scores in the two groups
were not statistically signicantly dierent from one another
in the preintervention measurement (p = 0.66).
At the follow-up conducted aer the completion of the
intervention, none of the participants had any signicant
health complaint, although no systematic evaluation of side
eects was conducted. At that time, among the 17 participants
included in the CBD group, nine reported that they decided
to receive some form of treatment (medication and CBT)
by regularly visiting hospitals, while none of the 20 participants
in the control group was found to make such a decision.
e participants of the CBD group were more likely to make
such decision than those of the placebo group were
(χ2(1) = 13.99, p < 0.001).
DISCUSSION
e anxiolytic eects of CBD have been extensively demonstrated
in animal studies and in healthy volunteers subjected to anxiety
induced by several procedures, including the simulation of public
speaking (Zuardi etal., 1993; Blessing etal., 2015). A pioneering
study that investigated the eects on SAD patients showed that
CBD reduces anticipatory anxiety (Crippa etal., 2011). Moreover,
CBD was found to exert a signicant eect on increased brain
activity in the right posterior cingulated cortex that is thought
to be involved in the processing of emotional information. A
subsequent study (Bergamaschi et al., 2011) experimentally
demonstrated a reduction in the anxiety provoked by simulated
public speaking by a single dose of CBD in patients with SAD,
although the ndings were preliminary. Based on these ndings,
the current study was conceived to extend the published research
into a more systematic study on the eect of CBD on teenagers
with SAD with avoidant personality disorder for a longer period.
Its results are consistent with those obtained by the previous
research and indicate that intervention with CBD for a 4-week
period reduced the level of symptoms in teenagers with SAD,
as measured by FSE and LSAS.
As an option for medication treatment for SAD, so far, the
use of paroxetine has been reported to be most eective
(Nordahl et al., 2016). at study reported that a 26-week
daily treatment with paroxetine alone produced a 5.2-point
decrease in the FNE score and a 10.2-point decrease in the
LSAS score. ose reported decreases in symptoms were almost
equivalent to the observed decreases induced by CBD here,
although the treatment groups studied in the two studies were
not closely compatible.
In children and adolescents, SAD is known to be among
the most common mental disorders (De Gregorio et al.,
2019). A survey conducted in the United States showed
that the disorder starts as early as age 5 and peaks around
age 12 (Merikangas et al., 2011). When untreated, it runs
a chronic course into adolescence and eventually adulthood.
In Japan, notably, the population of such teenagers with
avoidant personality disorder who “seclude themselves for
more than six months at home” (Saito, 2007) and “typically
withdraw from most social activities and retreat into their
living spaces” (Teo, 2010) is estimated to have reached
1,000,000 (Ogino, 2004), and this has become a serious
social problem. When they are provided with a higher level
of social support, their quality of life (QOL) is likely to
increase, whereas it deteriorates with poor support. The
teenagers with SAD in Japan who have higher levels of
social withdrawal along with such poor support are likely
to develop a stronger sense of loneliness and to suffer from
poorer QOL (Teo, 2010).
Despite such negative impacts of the disorder, the majority
of teenagers with SAD are likely to beuntreated. Psychotropic
medication and CBT are the most common therapeutic
options for SAD. However, socially anxious teenagers rarely
seek help due to the potential stigma associated with mental
issues and fear of interacting therapists and psychiatrists
(Ogino, 2004; Teo, 2010). As revealed by the follow-up
conducted in the current study, many of the participants
treated with CBD became positive in their attitude toward
seeking treatment. To overcome the dilemma of teenagers
with SAD described above, delivering interventions with CBD
could be an eective option for reducing the barriers facing
SAD patients in need of treatment.
In all, the results of the current study provide evidence for
anxiolytic eects of repeated CBD administration in teenagers
with SAD. At the same time, however, the author acknowledges
several limitations of the current study. No assay of the blood
level of CBD was undertaken. A more detailed baseline
sociodemographic evaluation could have been performed to
ensure the pretreatment similarity of the treatment groups.
Measurements need to beperformed at additional times between
the baseline and the end of the study. ese measures would
be essential to show, for example, if CBD could produce rapid
improvement of social anxiety (a putative advantage over
paroxetine). Moreover, possible side eects should be evaluated
systematically. Clearly, these are issues for future research that
should also be long-term studies with a positive control (e.g.,
paroxetine) to better assess the potential usefulness of CBD
in the therapy of SAD.
DATA AVAILABILITY STATEMENT
All datasets generated for this study are included in the article/
supplementary material.
ETHICS STATEMENT
e studies involving human participants were reviewed and
approved by the Institutional Ethics Committee of Kyoto
University (#2018-150), the Regional committee for medical
and health research ethics (#2018/1783), and the Japanese Data
Inspectorate for clinical trials (JCT0018004564). e patients/
participants provided their written informed consent to
participate in this study.
Masataka Cannabidiol and Social Anxiety
Frontiers in Psychology | www.frontiersin.org 6 November 2019 | Volume 10 | Article 2466
AUTHOR CONTRIBUTIONS
NM conceived the study, collected and analyzed the data, and
draed the manuscript.
FUNDING
is research was supported by a grant-in-aid (JSPS#25285201)
as well as by the Grants for Excellent Graduate Schools program
from the Ministry of Education, Science, Sports, and Culture,
Japanese Government. e funder had no role in study design,
data collection and analysis, decision to publish, or preparation
of the manuscript.
ACKNOWLEDGMENTS
e author is grateful to Dr. Satomi Yamada, Yoshiyuki Nagai,
Atsushi Ishige, Ryohei Tatsumi, and Koji Maki for their assistance
in conducting experimentation and Elizabeth Nakajima for
making comments on an earlier version of this manuscript.
REFERENCES
American Psychiatric Association (2000). Diagnostic and statistical manual of
mental disorders. 4th Edn. (Washington, DC: American Psychiatric Association,
Text Revision), 1–302.
Antony, M. M., and Rowa, K. (2008). Social anxiety disorder. (Cambridge, MA:
Hogrefe), 1–177.
Bergamaschi, M. M., Queiroz, R. H. C., Chagas, M. H. N., de Oliveira, D. C. G.,
De Martinis, B. S., Kapczinski, F., et al. (2011). Cannabidiol reduces the
anxiety induced by simulated public speaking in treatment-naïve social phobia
patients. Neuropsychopharmcology 36, 1219–1226. doi: 10.1038/npp.2011.6
Blessing, E. M., Steenkamp, M. M., Manzanares, J., and Marmar, C. R. (2015).
Cannabidiol as a potential treatment for anxiety disorders. Neurotherapeutics
12, 825–836. doi: 10.1007/s13311-015-0387-1
Brown, T. A., Campbell, L. A., Lehman, C. L., Grisham, J. R., and Mancill,
R. B. (2001). Current and lifetime comorbidity of the DSM-IV anxiety and
mood disorders in a large clinical sample. J. Abnorm. Psychol. 110, 49–58.
doi: 10.1037//0021-843x.110.4.585
Costa, B., Trovato, A. E., Comelli, F., Giagnoni, G., and Colleoni, M. (2007).
e non-psychoactive cannabis constituent cannabidiol is an orally eective
therapeutic agent in rat chronic inammatory and neuropathic pain. Eur.
J. Pharmacol. 556, 75–83. doi: 10.1016/j.ejphar.2006.11.006
Craske, M. G., Stein, M. B., Eley, T. C., Milad, M. R., Holmes, A., Rapes, R. M.,
et al. (2017). Anxiety disorders. Nat. Rev. Dis. Prim. 3:17024. doi: 10.1038/
nrdp.2017.24
Crippa, J. A., Derenusson, G. N., Ferrari, T. B., Wichert-Ana, L., Duran, F. L.,
Martin-Santos, R., et al. (2011). Neural basis of anxiolytic eects of cannabidiol
(CBD) in generalized social anxiety disorder: a preliminary report.
J. Psychopharmacol. 25, 121–130. doi: 10.1177/0269881110379283
De Gregorio, D., McLaughlin, R. J., Posa, L., Ochoa-Sanchez, R., Enns, J.,
Lopez-Canul, M., et al. (2019). Cannabidiol modulates serotonergic transmission
and reverses both allodynia and anxiety-like behavior in a model of neuropathic
pain. Pain 160, 136–150. doi: 10.1097/j.pain.0000000000001386
Devinsky, O., Marsh, E., Friedman, D., iele, E., Laux, L., Sullivan, J., et al.
(2016). Cannabidiol in patients with treatment-resistant epilepsy: an open-
label interventional trial. Lancet Neurol. 15, 270–278. doi: 10.1016/
S1474-4422(15)00379-8
Di Nardo, P. A., Brown, T. A., and Barlow, D. H. (1994). Anxiety disorders
interview schedule for DSM-IV (ADIS-IV). Albany, NY: Graywind, 1–195.
First, M. B., Gibbon, M., Spitzer, R. L., Williams, J. B. W., and Benjamin, L.
S. (1997). User’s guide for the structured clinical interview for DSM-IV Axis
II personality disorders (SCD-II). (Washington, DC: American Psychiatric
Press), 1–176.
Kessler, R. C., Berglund, P., Demier, O., Jin, R., Merikangas, K. R., and Walters,
E. E. (2005). Lifetime prevalence and age-of-onset distribution of DSM-IV-R
psychiatric disorders in the National Comorbidity Survey Replication. Arch.
Gen. Psychiatry 49, 273–281. doi: 10.1001/archpsyc.62.6.593
Liebowitz, M. R. (1987). Social phobia. Mod. Probl. Pharm. Psychiatry 22,
141–173.
Masataka, N. (2003). e onset of language. (Cambridge: Cambridge University
Press), 1–281.
McGuire, P., Robson, P., Cubala, W. J., Vasile, D., Morrison, P. D., Barron, R.,
et al. (2018). Cannabidiol (CBD) as an adjunctive therapy in schizophrenia:
a multicenter randomized control trial. Am. J. Psychiatry 175, 225–231. doi:
10.1176/appi.ajp.2017.17030325
Merikangas, K. R., He, J. P., Burstein, M., Svendsen, J., Avenevoll, S.,
Case, B., et al. (2011). Service utilization for lifetime mental disorders
in U.S. adolescents: results of the National Comorbidity Survey-Adolescent
Supplement (NCS-A). J. Am. Acad. Child Adolesc. Psychiatry 50, 32–45.
doi: 10.1016/j.jaac.2010.10.006
Nordahl, H. M., Vogel, P. A., Morken, G., Stiles, T. C., Sandvik, P., and Wells,
A. (2016). Paraxetine, cognitive therapy or their combination in the treatment
of social anxiety disorder with and without avoidant personality disorder:
a randomized clinical trial. Psychother. Psychosom. 85, 346–356. doi:
10.1159/000447013
Ogino, T. (2004). Managing categorization and social withdrawal in Japan:
rehabilitation process in a private support group for hikikomorians. Int. J.
Soc. Psychiatry 13, 120–133. doi: 10.1111/j.1475-6781.2004.00057.x
Parker, L. A., Kwiatokowska, M., and Mechoulam, R. (2006). Delta-9-
tetrahydrocannabinol and cannabidiol, but not ondansetron, interfere with
conditioned retching reactions elicited by a lithium-paired context in Suncus
murinus: an animal model of anticipatory nausea and vomiting. Physiol.
Behav. 87, 66–71. doi: 10.1016/j.physbeh.2005.08.045
Ryan, J. L., and Warner, C. M. (2012). Treating adolescents with social anxiety
disorders in schools. Child Adolesc. Psychiatr. Clin. N. Am. 21:1050118. doi:
10.1016/j.chc.2011.08.011
Saito, T. (2007). Why hikikomori is cured? Tokyo: Chuo Hoki Press, 1–138.
Scuderi, C., Filippis, D. D., Iuvone, T., Blasio, A., Steardo, A., and
Esposito, G. (2009). Cannabidiol in medicine: a review of its therapeutic
potential in CNS disorders. Psychother. Res. 23, 597–602. doi: 10.1002/
ptr.2625
Somers, J. M., Goldner, E. M., Waraich, P., and Hsu, L. (2006). Prevalence
and incidence studies of anxiety disorders: a systematic review of the literature.
Can. J. Psychiatry 51, 110–113. doi: 10.1177/070674370605100206
Teo, A. R. (2010). A new form of social withdrawal in Japan: a review of
Hikikomori. Int. J. Soc. Psychiatry 56, 175–185. doi: 10.1177/
0020764008100629
Watson, D., and Friend, R. (1969). Measurement of social-evaluative anxiety.
J. Consult. Clin. Psychol. 33, 448–457. doi: 10.1037/h0027806
Whiting, P. F., Wol, R. F., Deshepande, S., Di Nisio, M., Duy, S., Hemandez,
A. V., et al. (2015). Cannabinoids for medical use: a systematic review and
meta-analysis. JAMA 313, 2456–2473. doi: 10.1001/jama.2015.6358
Zuardi, A. W., Cosme, R. A., Grae, F. G., and Guimaraes, F. S. (1993). Eects
of ipsapirone and cannabidiol on human experimental anxiety. J. Psychopharmacol.
7, 82–88. doi: 10.1177/026988119300700112
Conict of Interest: e author declares that the research was conducted in
the absence of any commercial or nancial relationships that could beconstrued
as a potential conict of interest.
Copyright © 2019 Masataka. is is an open-access article distributed under the
terms of the Creative Commons Attribution License (CC BY). e use, distribution
or reproduction in other forums is permitted, provided the original author(s) and
the copyright owner(s) are credited and that the original publication in this journal
is cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
... Participants, both male and female, had experienced anxiety disorders for at least six months. Over a four-week period, they received daily doses of hemp oil containing 300 mg of CBD (17 participants) or a placebo (20 participants) [14]. ...
... A clinical psychologist, unaware of whether the participants received CBD or the placebo, visited their homes daily in the afternoon to administer 420 ml of the oil via a syringe throughout the four-week intervention. After the intervention, during a follow-up phase, the clinical psychologists monitored the participants' health and assessed any effects of the administered substance once a week for up to six months [14]. ...
... Similarly, average LSAS scores in the CBD group were significantly reduced after the intervention compared to before (p = 0.03), but no significant reduction in LSAS scores was found in the placebo group (p = 0.42). Postintervention LSAS scores in the CBD group were also significantly lower than those in the placebo group (p = 0.0018), while pre-intervention differences between the two groups were not statistically significant (p = 0.66) [14]. ...
Article
Full-text available
Anxiety disorders are the most commonly diagnosed mental disorders worldwide, and their prevalence continues to rise, leading to significant social and economic burdens. Recent interest has focused on CBD as a potential treatment. This systematic review assesses the current evidence on the efficacy of CBD in treating anxiety and related disorders, including post-traumatic stress disorder (PTSD), social anxiety disorder (SAD), and autism spectrum disorder (ASD). Unlike open-label trials prone to placebo bias, this study reviews randomized, double-blind, placebo-controlled trials. A review of literature was conducted using the Google Scholar database to gather information on "cannabidiol," specifically focusing on its relationship with "anxiety," "social anxiety disorder", "post-traumatic stress disorder” and ,,human trials." Positive outcomes were observed with 300 mg CBD doses, reducing anxiety in SAD patients, tremor in Parkinson’s disease, and anxiety in ASD and opioid-dependent individuals. Minimal improvements were noted in PTSD patients with sexual trauma. Further research is needed to clarify CBD’s therapeutic potential in diverse clinical contexts.
... None of these trials showed effects of CBD in the dose range < 300 mg/day (4.3 mg/kg bw/day) on anxiety (Table 6). By contrast, all included studies investigating higher doses of 300 mg CBD/day (4.3 mg/kg bw/day) [72,106,194,195,197,198], 350 mg CBD/day (5 mg/kg bw/day) [199], or 400 mg CBD/day (5.7 mg/kg bw/day [57,74,200] reported positive effects on anxiety. However, some of these studies were conducted in patients with social anxiety disorders [198] or heroin use disorders [74], which may not be directly transferrable to healthy subjects, e.g., consumers of food supplements. ...
... By contrast, all included studies investigating higher doses of 300 mg CBD/day (4.3 mg/kg bw/day) [72,106,194,195,197,198], 350 mg CBD/day (5 mg/kg bw/day) [199], or 400 mg CBD/day (5.7 mg/kg bw/day [57,74,200] reported positive effects on anxiety. However, some of these studies were conducted in patients with social anxiety disorders [198] or heroin use disorders [74], which may not be directly transferrable to healthy subjects, e.g., consumers of food supplements. Other studies, not included in the review by Arnold et al. [193], also showed positive effects at 300 mg CBD/day (4.3 mg/kg bw/day) [201], 600 mg CBD/day (8.6 mg/kg bw/day) ( [115,202,203], and 800 mg CBD/day (11.4 mg/kg bw/day) [204], while other studies found no effects at 600 mg CBD/day (8.6 mg/kg bw/day) [112,195,205,206], 150-600 mg (2.1-8.6 mg/kg bw/day) [207], or even 900 mg CBD/day (13 mg/kg bw/day) [194]. ...
Article
Full-text available
Background: Cannabidiol (CBD) is a cannabinoid present in the hemp plant (Cannabis sativa L.). Non-medicinal CBD oils with typically 5–40% CBD are advertised for various alleged positive health effects. While such foodstuffs containing cannabinoids are covered by the Novel Food Regulation in the European Union (EU), none of these products have yet been authorized. Nevertheless, they continue to be available on the European market. Methods: The Permanent Senate Commission on Food Safety (SKLM) of the German Research Foundation (DFG) reviewed the currently available data on adverse and potential beneficial effects of CBD in the dose range relevant for foods. Results: Increased liver enzyme activities were observed in healthy volunteers following administration of 4.3 mg CBD/kg bw/day and higher for 3–4 weeks. As lower doses were not tested, a no observed adverse effect level (NOAEL) could not be derived, and the dose of 4.3 mg/kg bw/day was identified as the lowest observed adverse effect level (LOAEL). Based on the CBD content and dose recommendations of CBD products on the market, the SKLM considered several exposure scenarios and concluded that the LOAEL for liver toxicity may be easily reached, e.g., via consumption of 30 drops of an oil containing 20% CBD, or even exceeded. A critical evaluation of the available data on potential beneficial health effects of CBD in the dose range at or below the LOAEL of 4.3 mg/kg bw/day revealed no scientific evidence that would substantiate health claims, e.g., in relation to physical performance, the cardiovascular, immune, and nervous system, anxiety, relaxation, stress, sleep, pain, or menstrual health. Conclusions: The SKLM concluded that consumption of CBD-containing foods/food supplements may not provide substantiated health benefits and may even pose a health risk to consumers.
... Anxiety and anxiety-related complications are the most common reason for off-label medicinal hemp use [11,48,49]. While empirical demonstrations of CBD s anxiolytic effects are becoming more common [32,50], there is little empirical understanding of independent contributions that prominent monoterpenes or monoterpenoids, found in whole-plant cannabis extracts, contribute to the plant s purported anxiolytic effects. ...
... Anxiety and anxiety-related complications are the most common reason for off-label medicinal hemp use [11,48,49]. While empirical demonstrations of CBD's anxiolytic effects are becoming more common [32,50], there is little empirical understanding of independent contributions that prominent monoterpenes or monoterpenoids, found in whole-plant cannabis extracts, contribute to the plant's purported anxiolytic effects. ...
Article
Full-text available
Volatile organic compounds, colloquially referred to as “terpenes”, have been proposed to impact the therapeutic qualities that are traditionally ascribed to cannabis. However, the contribution of these terpenes in anxiety, at relevant levels and exposure methods common with cannabis use, is lacking empirical assessment. We tested the anxiolytic properties of two prominent cannabis terpenes, linalool and β-myrcene, in male and female mice using short duration vapor pulls to model human inhalation when combusting flower or vaping cannabis oil. We observed sex differences in the locomotor effects in the open field and anxiolytic properties in the elevated plus maze of these terpenes that depended on their exposure characteristics. Both linalool and β-myrcene had anxiolytic effects in female mice when delivered in discrete vapor pulls over the course of 30 min. In male mice, only a single vapor hit containing linalool or β-myrcene had anxiolytic effects. The combination of sub-effective levels of linalool and the phytocannabinoid, cannabidiol (CBD), had synergistic anxiolytic effects in females, but these entourage effects between CBD and terpenes were absent with β-myrcene for females and for either terpene in males. Together, our findings reveal sex differences in the anxiolytic properties of common cannabis terpenes and highlight the potential benefits of unique combinations of CBD and terpenes in expanding the therapeutic dose window.
... Not surprisingly, there is significant interest in pursuing the development of cannabinoid-based products for anxiety, particularly those rich in CBD. While additional work is needed, acute administration studies and clinical trials thus far have reported that CBD appears to be effective for reducing situational anxiety, such as public speaking tasks [45,46], and for reducing anxiety in those with moderate-to-severe anxiety [47], social anxiety disorder [48,49], and treatment-resistant anxiety [50]. In addition, a recent systematic review and meta-analysis [44] assessed eight clinical studies evaluating anxiety symptoms (i.e., anxiety disorders or anxiety induced by other medical conditions such as high paranoia and PTSD) in 316 participants (157 assigned to CBD treatment and 159 to a control group). ...
Article
The relationship between cannabis use and mental health is complex, as studies often report seemingly contradictory findings regarding whether cannabis use results in more positive or negative treatment outcomes. With an increasing number of individuals using cannabis for both recreational (i.e., non-medical) and medical purposes, it is critical to gain a deeper understanding of the ways in which cannabis may be helpful or harmful for those diagnosed with psychiatric disorders. Although cannabis is composed of hundreds of compounds, studies assessing the effects of “cannabis” most often report the impact of delta-9-tetrahydrocannabinol (d9-THC), the primary intoxicating constituent of the plant. While d9-THC has documented therapeutic properties, negative clinical outcomes commonly associated with cannabis are generally related to d9-THC exposure. In contrast, non-intoxicating cannabinoids such as cannabidiol (CBD) show promise as potential treatment options for psychiatric symptoms. In this article, findings from studies and reviews examining the relationship between mental health conditions (mood, anxiety, psychosis, and post-traumatic stress disorder [PTSD]) and cannabis use are summarized to highlight critical variables that are often overlooked, including those associated with cannabis use patterns (e.g., frequency of use, amount used, cannabinoid exposure, product choice, and route of administration). Further, this article explores individual factors (e.g., age, sex, genetics/family history) that likely impact cannabis-related outcomes. Research to date suggests that youth and those with a family history or genetic liability for psychiatric disorders are at higher risk for negative outcomes, while more research is needed to fully understand unique effects related to sex and older age.
... Journal of Clinical and Basic Psychosomatics has been shown to have anxiolytic (anxiety-reducing) effects, which can be particularly beneficial for individuals whose insomnia is driven by anxiety or stress. 127,128 Unlike THC, CBD does not have sedative effects at typical doses, but it can help improve sleep by reducing the cognitive arousal and anxiety that often interfere with sleep onset. Moreover, CBD has been found to modulate the SWC through its effects on the hypothalamus, which plays a crucial role in maintaining circadian rhythms. ...
Article
Full-text available
Insomnia is the most prevalent sleep disorder with significant implications for mental and physical health. While traditionally understood through a biomedical lens, increasing evidence highlights the role of psychosomatic factors in its onset and perpetuation. This narrative review explores the relationship between psychological and somatic influences on insomnia, examining the underlying mechanisms, diagnostic challenges, and treatment strategies. We discuss neurobiological pathways, including the hypothalamic–pituitary–adrenal axis and neurotransmitter imbalances, alongside psychological factors such as anxiety, hyperarousal, and cognitive distortions. Somatic symptoms, particularly chronic pain, and other physical conditions are also regarded as integral contributors to insomnia. Diagnosis of psychosomatic insomnia requires a comprehensive approach that incorporates both psychological and physical assessments, utilizing a combination of clinical interviews, standardized questionnaires, and differential diagnostic techniques. Treatment strategies are discussed in this paper with an emphasis on cognitive-behavioral therapy for insomnia, pharmacological interventions, and integrative approaches that address the multifaceted nature of the disorder. The review also highlights the importance of lifestyle modifications and the potential role of alternative therapies in managing insomnia. A rounded understanding of psychosomatic influences is crucial for effective diagnosis and treatment of insomnia. Future research should focus on personalized therapeutic approaches and further interpreting the complex interconnections between mind and body in the pathophysiology of insomnia. This perspective has the potential to enhance clinical practice and improve outcomes for individuals suffering from this challenging disorder.
Article
Full-text available
A presente revisão destaca o uso do canabidiol (CBD) como alternativa terapêutica para transtornos de ansiedade e depressão, destacando sua interação com o sistema endocanabinoide e os receptores 5HT1A, responsáveis pela regulação do humor e estresse. Materiais e Métodos: trata-se de uma revisão integrativa de literatura de artigos publicados entre 2015 e 2024, utilizando a estratégia PICO para definir a eficácia do CBD em comparação com placebo e tratamentos convencionais. Resultados e Discussão: Foram incluídos 11 estudos que indicaram resultados variados: enquanto alguns demonstraram reduções significativas nos sintomas de ansiedade e depressão, outros mostraram evidências inconclusivas ou benefícios limitados. Conclusão: Embora o CBD tenha se mostrado seguro e potencialmente eficaz, especialmente em formulações específicas e para populações selecionadas, a necessidade de estudos mais robustos é destacada. Conclusivamente, o CBD é apresentado como uma abordagem terapêutica emergente, mas ainda carente de validação científica abrangente para aplicação clínica rotineira.
Article
Artocarpus lacucha Linn is a traditional herbal medicine used for different diseases and they have a good anti-oxidant nature to help treat neuroinflammation and protect the potential of anti-depressants, anti-anxiety. Commonly known as Monkey Jack or Monkey Fruit, is a tropical fruit-bearing tree found in Southeast Asia. This review article aims to explore the potential mechanisms and evidence supporting the role of Artocarpus lacucha in combating antidepressant and antianxiety effects, and increase the GABA, GABAA&B levels and improving mental health, shedding light on its pharmacological properties, and highlighting avenues for further research. The findings indicate that while mitochondrial protection was difficult to achieve, both of the substances studied Artocarpus lacucha Linn —improved cell survival, particularly in relation to ROS and lipid peroxidation. Because of the effectiveness of the redox-sensitive expression of antioxidant enzymes and its pharmacokinetic properties, oral Artocarpus lacucha Linn may offer useful protection against acute neurodegenerative diseases.
Article
O uso dos canabinoides vem ganhando destaque para o tratamento de diversas patologias. Na psiquiatria, seu uso no tratamento de transtornos ansiosos permanece controverso, elevando a necessidade de novos estudos.
Article
Full-text available
Clinical studies indicate that cannabidiol (CBD), the primary non-addictive component of cannabis that interacts with the serotonin (5-HT) 1A receptor, may possess analgesic and anxiolytic effects. However, its effects on 5-HT neuronal activity, as well as its impact in models of neuropathic pain are unknown. First, using in-vivo single unit extracellular recordings in rats, we demonstrated that acute intravenous (i.v.) increasing doses of CBD (0.1-1.0 mg/kg) decreased the firing rate of 5-HT neurons in the dorsal raphe nucleus (DRN), which was prevented by administration of the 5-HT1A antagonist WAY 100635 (0.3 mg/kg, i.v.) and the TRPV1 antagonist capsazepine (1 mg/kg, i.v.) but not by the CB1 receptor antagonist AM 251 (1 mg/kg, i.v.). Repeated treatment with CBD (5 mg/kg/day, subcutaneously, s.c, for 7 days) increased 5-HT firing via desensitization of 5-HT1A receptors. Rats subjected to the spared nerve injury (SNI) model for 24 days showed decreased 5-HT firing activity, mechanical allodynia, and increased anxiety-like behavior in the elevated plus maze (EPMT), open field (OFT), and novelty suppressed feeding tests (NSFT). Seven days of treatment with CBD reduced mechanical allodynia, decreased anxiety-like behavior, and normalized 5-HT activity. Anti-allodynic effects of CBD were fully prevented by capsazepine (10 mg/kg/day, s.c., for 7 days) and partially prevented by WAY 100635 (2 mg/kg/day, s.c., for 7 days), while the anxiolytic effect was blocked only by WAY. Overall, repeated treatment with low-dose CBD induces analgesia predominantly via TRPV1 activation, reduces anxiety via 5-HT1A receptor activation, and rescues impaired 5-HT neurotransmission under neuropathic pain conditions.
Article
Full-text available
Objective: Research in both animals and humans indicates that cannabidiol (CBD) has antipsychotic properties. The authors assessed the safety and effectiveness of CBD in patients with schizophrenia. Method: In an exploratory double-blind parallel-group trial, patients with schizophrenia were randomized in a 1:1 ratio to receive CBD (1000 mg/day; N=43) or placebo (N=45) alongside their existing antipsychotic medication. Participants were assessed before and after treatment using the Positive and Negative Syndrome Scale (PANSS), the Brief Assessment of Cognition in Schizophrenia (BACS), the Global Assessment of Functioning scale (GAF), and the improvement and severity scales of the Clinical Global Impressions Scale (CGI-I and CGI-S). Results: After 6 weeks of treatment, compared with the placebo group, the CBD group had lower levels of positive psychotic symptoms (PANSS: treatment difference=-1.4, 95% CI=-2.5, -0.2) and were more likely to have been rated as improved (CGI-I: treatment difference=-0.5, 95% CI=-0.8, -0.1) and as not severely unwell (CGI-S: treatment difference=-0.3, 95% CI=-0.5, 0.0) by the treating clinician. Patients who received CBD also showed greater improvements that fell short of statistical significance in cognitive performance (BACS: treatment difference=1.31, 95% CI=-0.10, 2.72) and in overall functioning (GAF: treatment difference=3.0, 95% CI=-0.4, 6.4). CBD was well tolerated, and rates of adverse events were similar between the CBD and placebo groups. Conclusions: These findings suggest that CBD has beneficial effects in patients with schizophrenia. As CBD's effects do not appear to depend on dopamine receptor antagonism, this agent may represent a new class of treatment for the disorder.
Article
Full-text available
Background: The most efficacious treatments for social anxiety disorder (SAD) are the SSRIs and cognitive therapy (CT). Combined treatment is advocated for SAD but has not been evaluated in randomized trials using CT and SSRI. Our aim was to evaluate whether one treatment is more effective than the other and whether combined treatment is more effective than the single treatments. Methods: A total of 102 patients were randomly assigned to paroxetine, CT, the combination of CT and paroxetine, or pill placebo. The medication treatment lasted 26 weeks. Of the 102 patients, 54% fulfilled the criteria for an additional diagnosis of avoidant personality disorder. Outcomes were measured at posttreatment and 12-month follow-up assessments. Results: CT was superior to paroxetine alone and to pill placebo at the end of treatment, but it was not superior to the combination treatment. At the 12-month follow-up, the CT group maintained benefits and was significantly better than placebo and paroxetine alone, whereas there were no significant differences among combination treatment, paroxetine alone, and placebo. Recovery rates at 12 months were much higher in the CT group (68%) compared to 40% in the combination group, 24% in the paroxetine group, and 4% in the pill placebo group. Conclusions: CT was the most effective treatment for SAD at both posttreatment and follow-up compared to paroxetine and better than combined treatment at the 12-month follow-up on the Liebowitz Social Anxiety Scale. Combined treatment provided no advantage over single treatments; rather there was less effect of the combined treatment compared to CT alone.
Article
Full-text available
Cannabidiol (CBD), a Cannabis sativa constituent, is a pharmacologically broad-spectrum drug that in recent years has drawn increasing interest as a treatment for a range of neuropsychiatric disorders. The purpose of the current review is to determine CBD's potential as a treatment for anxiety-related disorders, by assessing evidence from preclinical, human experimental, clinical, and epidemiological studies. We found that existing preclinical evidence strongly supports CBD as a treatment for generalized anxiety disorder, panic disorder, social anxiety disorder, obsessive-compulsive disorder, and post-traumatic stress disorder when administered acutely; however, few studies have investigated chronic CBD dosing. Likewise, evidence from human studies supports an anxiolytic role of CBD, but is currently limited to acute dosing, also with few studies in clinical populations. Overall, current evidence indicates CBD has considerable potential as a treatment for multiple anxiety disorders, with need for further study of chronic and therapeutic effects in relevant clinical populations.
Article
Full-text available
Cannabis and cannabinoid drugs are widely used to treat disease or alleviate symptoms, but their efficacy for specific indications is not clear. To conduct a systematic review of the benefits and adverse events (AEs) of cannabinoids. Twenty-eight databases from inception to April 2015. Randomized clinical trials of cannabinoids for the following indications: nausea and vomiting due to chemotherapy, appetite stimulation in HIV/AIDS, chronic pain, spasticity due to multiple sclerosis or paraplegia, depression, anxiety disorder, sleep disorder, psychosis, glaucoma, or Tourette syndrome. Study quality was assessed using the Cochrane risk of bias tool. All review stages were conducted independently by 2 reviewers. Where possible, data were pooled using random-effects meta-analysis. Patient-relevant/disease-specific outcomes, activities of daily living, quality of life, global impression of change, and AEs. A total of 79 trials (6462 participants) were included; 4 were judged at low risk of bias. Most trials showed improvement in symptoms associated with cannabinoids but these associations did not reach statistical significance in all trials. Compared with placebo, cannabinoids were associated with a greater average number of patients showing a complete nausea and vomiting response (47% vs 20%; odds ratio [OR], 3.82 [95% CI, 1.55-9.42]; 3 trials), reduction in pain (37% vs 31%; OR, 1.41 [95% CI, 0.99-2.00]; 8 trials), a greater average reduction in numerical rating scale pain assessment (on a 0-10-point scale; weighted mean difference [WMD], -0.46 [95% CI, -0.80 to -0.11]; 6 trials), and average reduction in the Ashworth spasticity scale (WMD, -0.36 [95% CI, -0.69 to -0.05]; 7 trials). There was an increased risk of short-term AEs with cannabinoids, including serious AEs. Common AEs included dizziness, dry mouth, nausea, fatigue, somnolence, euphoria, vomiting, disorientation, drowsiness, confusion, loss of balance, and hallucination. There was moderate-quality evidence to support the use of cannabinoids for the treatment of chronic pain and spasticity. There was low-quality evidence suggesting that cannabinoids were associated with improvements in nausea and vomiting due to chemotherapy, weight gain in HIV infection, sleep disorders, and Tourette syndrome. Cannabinoids were associated with an increased risk of short-term AEs.
Article
Anxiety disorders constitute the largest group of mental disorders in most western societies and are a leading cause of disability. The essential features of anxiety disorders are excessive and enduring fear, anxiety or avoidance of perceived threats, and can also include panic attacks. Although the neurobiology of individual anxiety disorders is largely unknown, some generalizations have been identified for most disorders, such as alterations in the limbic system, dysfunction of the hypothalamic-pituitary-adrenal axis and genetic factors. In addition, general risk factors for anxiety disorders include female sex and a family history of anxiety, although disorder-specific risk factors have also been identified. The diagnostic criteria for anxiety disorders varies for the individual disorders, but are generally similar across the two most common classification systems: The Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5) and the International Classification of Diseases, Tenth Edition (ICD-10). Despite their public health significance, the vast majority of anxiety disorders remain undetected and untreated by health care systems, even in economically advanced countries. If untreated, these disorders are usually chronic with waxing and waning symptoms. Impairments associated with anxiety disorders range from limitations in role functioning to severe disabilities, such as the patient being unable to leave their home.
Article
Background: Almost a third of patients with epilepsy have a treatment-resistant form, which is associated with severe morbidity and increased mortality. Cannabis-based treatments for epilepsy have generated much interest, but scientific data are scarce. We aimed to establish whether addition of cannabidiol to existing anti-epileptic regimens would be safe, tolerated, and efficacious in children and young adults with treatment-resistant epilepsy. Methods: In this open-label trial, patients (aged 1-30 years) with severe, intractable, childhood-onset, treatment-resistant epilepsy, who were receiving stable doses of antiepileptic drugs before study entry, were enrolled in an expanded-access programme at 11 epilepsy centres across the USA. Patients were given oral cannabidiol at 2-5 mg/kg per day, up-titrated until intolerance or to a maximum dose of 25 mg/kg or 50 mg/kg per day (dependent on study site). The primary objective was to establish the safety and tolerability of cannabidiol and the primary efficacy endpoint was median percentage change in the mean monthly frequency of motor seizures at 12 weeks. The efficacy analysis was by modified intention to treat. Comparisons of the percentage change in frequency of motor seizures were done with a Mann-Whitney U test. Results: Between Jan 15, 2014, and Jan 15, 2015, 214 patients were enrolled; 162 (76%) patients who had at least 12 weeks of follow-up after the first dose of cannabidiol were included in the safety and tolerability analysis, and 137 (64%) patients were included in the efficacy analysis. In the safety group, 33 (20%) patients had Dravet syndrome and 31 (19%) patients had Lennox-Gastaut syndrome. The remaining patients had intractable epilepsies of different causes and type. Adverse events were reported in 128 (79%) of the 162 patients within the safety group. Adverse events reported in more than 10% of patients were somnolence (n=41 [25%]), decreased appetite (n=31 [19%]), diarrhoea (n=31 [19%]), fatigue (n=21 [13%]), and convulsion (n=18 [11%]). Five (3%) patients discontinued treatment because of an adverse event. Serious adverse events were reported in 48 (30%) patients, including one death-a sudden unexpected death in epilepsy regarded as unrelated to study drug. 20 (12%) patients had severe adverse events possibly related to cannabidiol use, the most common of which was status epilepticus (n=9 [6%]). The median monthly frequency of motor seizures was 30·0 (IQR 11·0-96·0) at baseline and 15·8 (5·6-57·6) over the 12 week treatment period. The median reduction in monthly motor seizures was 36·5% (IQR 0-64·7). Interpretation: Our findings suggest that cannabidiol might reduce seizure frequency and might have an adequate safety profile in children and young adults with highly treatment-resistant epilepsy. Randomised controlled trials are warranted to characterise the safety profile and true efficacy of this compound. Funding: GW Pharmaceuticals, Epilepsy Therapy Project of the Epilepsy Foundation, Finding A Cure for Epilepsy and Seizures.
Article
The comorbidity of current and lifetime DSM-IV anxiety and mood disorders was examined in 1,127 outpatients who were assessed with the Anxiety Disorders Interview Schedule for DSM-IV :Lifetime version (ADIS-IV-L). The current and lifetime prevalence of additional Axis I disorders in principal anxiety and mood disorders was found to be 57% and 81%, respectively. The principal diagnostic categories associated with the highest comorbidity rates were mood disorders, posttraumatic stress disorder (PTSD), and generalized anxiety disorder (GAD). A high rate of lifetime comorbidity was found between the anxiety and mood disorders; the lifetime association with mood disorders was particularly strong for PTSD, GAD, obsessive-compulsive disorder, and social phobia. The findings are discussed in regard to their implications for the classification of emotional disorders.
Article
• While other anxiety disorders have recently become the subjects of increasing investigation, social phobia remains, except among behavior therapists, relatively unstudied. As a result, major uncertainties exist concerning classification, prevalence, severity, etiology, assessment, and treatment of social phobia. Existing findings do suggest that in its own right and as a comparison for other anxiety disorders, social phobia should prove a fertile area for psychobiological and clinical investigation.