Skip to main content
  • Research article
  • Open access
  • Published:

Medicinal cannabis for psychiatric disorders: a clinically-focused systematic review

Abstract

Background

Medicinal cannabis has received increased research attention over recent years due to loosening global regulatory changes. Medicinal cannabis has been reported to have potential efficacy in reducing pain, muscle spasticity, chemotherapy-induced nausea and vomiting, and intractable childhood epilepsy. Yet its potential application in the field of psychiatry is lesser known.

Methods

The first clinically-focused systematic review on the emerging medical application of cannabis across all major psychiatric disorders was conducted. Current evidence regarding whole plant formulations and plant-derived cannabinoid isolates in mood, anxiety, sleep, psychotic disorders and attention deficit/hyperactivity disorder (ADHD) is discussed; while also detailing clinical prescription considerations (including pharmacogenomics), occupational and public health elements, and future research recommendations. The systematic review of the literature was conducted during 2019, assessing the data from all case studies and clinical trials involving medicinal cannabis or plant-derived isolates for all major psychiatric disorders (neurological conditions and pain were omitted).

Results

The present evidence in the emerging field of cannabinoid therapeutics in psychiatry is nascent, and thereby it is currently premature to recommend cannabinoid-based interventions. Isolated positive studies have, however, revealed tentative support for cannabinoids (namely cannabidiol; CBD) for reducing social anxiety; with mixed (mainly positive) evidence for adjunctive use in schizophrenia. Case studies suggest that medicinal cannabis may be beneficial for improving sleep and post-traumatic stress disorder, however evidence is currently weak. Preliminary research findings indicate no benefit for depression from high delta-9 tetrahydrocannabinol (THC) therapeutics, or for CBD in mania. One isolated study indicates some potential efficacy for an oral cannabinoid/terpene combination in ADHD. Clinical prescriptive consideration involves caution in the use of high-THC formulations (avoidance in youth, and in people with anxiety or psychotic disorders), gradual titration, regular assessment, and caution in cardiovascular and respiratory disorders, pregnancy and breast-feeding.

Conclusions

There is currently encouraging, albeit embryonic, evidence for medicinal cannabis in the treatment of a range of psychiatric disorders. Supportive findings are emerging for some key isolates, however, clinicians need to be mindful of a range of prescriptive and occupational safety considerations, especially if initiating higher dose THC formulas.

Peer Review reports

Introduction

The Cannabaceae family is a comparatively small family of flowering plants encompassing 11 genera and approximately 170 different species, a small number of which elicit a range of varying psychoactive effects [1]. Several medical applications have been studied over the past decades, with the National Academies of Sciences, Engineering and Medicine (NASEM) recently holding the position that cannabis and cannabinoids demonstrate conclusive or substantial evidence for chronic pain in adults, chemotherapy-induced nausea and vomiting and spasticity in multiple sclerosis, with limited evidence for use in increasing appetite in HIV/AIDS patients and improving symptoms of post-traumatic stress disorder (PTSD) [2].

While there is increasing psychiatric interest (and debate) regarding the potential mental health applications (in concert with concerns over the potential for triggering latent psychosis), historical evidence for the use of cannabis in mental health conditions is remarkably ancient. For instance, the Shen-nung Pen-tsao Ching (Divine Husbandman’s Materia Medica) described its benefit as an anti-senility agent [3, 4], while in the Assyrian culture, cannabis was indicated as a drug for grief and sorrow [5, 6]. Sections of the Indian Atharva Veda (1500 BCE) suggest bhanga (Cannabis) exerted anxiolytic effects [5, 7], while in 1563, Da Orta [8] described cannabis as allaying anxiety and engendering laughter. With respect to modern use, contemporary consumers of cannabis report (as assessed via meta-analysis of patient usage data) that pain (64%), anxiety (50%), and depression/mood (34%) are the most common reasons [9].

Increasing scientific research, conducted over recent years, has seen the regulatory pendulum swinging away from the United Nations Single Convention on Narcotic Drugs in 1961 (which recommends enforcement of cannabis use as illegal) [10], towards consideration of its potential use in medical conditions. Recent scientific evidence ascribes anxiolytic, neuroprotective, antioxidant, anti-inflammatory, antidepressant, anti-psychotic and hypnotic pharmacological actions due to several phytochemicals commonly found in the cannabis genus [11, 12].

While Δ9-tetrahydrocannabinol (THC) is considered the main psychoactive constituent, other cannabinoids have also revealed less potent psychotropic effects. These include cannabidiol (CBD) [13], Δ8-tetrahydrocannabinol [14], and other less-studied cannabinoids including cannabinol (CBN) and Δ8 tetrahydrocannabivarin (THCV) [15]. Further, many other constituents such as the terpenes (i.e. volatile organic compounds found mainly as essential oils in many plants), also provoke a range of biological effects, and produce the characteristic aroma of the plant [16]. The hundreds of cannabis chemovars or varieties (commonly referred to as strains) developed over millennia have unique and complex constituent profiles, of which each may provide targeted therapeutic usage due to the unique synergistic combination of plant chemicals. Some pharmaceutical preparations have attempted to isolate the key constituents (there are over 140 phytocannabinoids [17]) to provide standardised formulas that may harness this ‘entourage effect’ [16, 18], while being able to provide batch-to-batch assurance of the medicine.

While other reviews have covered cannabis’ use in a range of conditions (cf. Whiting et al. 2015 [10] for a general review of evidence for medicinal cannabis), none to date have provided both a systematic and ‘clinically-focused’ review on psychiatric disorders. As the focus was on emerging data for the use of mental health disorders, we omitted addiction and neurological disorders, which have been extensively covered elsewhere [10, 19], cf. pain [20,21,22], cf. epilepsy [23, 24], cf. movement disorders [25]. A further motive for this paper focusing solely on psychiatric disorders, concerns cannabis users noting that self-reported anxiety, insomnia, and depression symptoms are amongst the most common reasons for usage [26].

Thus, the primary purpose of this paper is to provide a systematic review of the current state of evidence in the emerging field of cannabinoid therapies for psychiatric disorders (PTSD, generalised anxiety disorder, social anxiety, insomnia, psychotic disorders, and attention-deficit hyperactivity disorder: ADHD). In addition, this review provides clinical prescriptive guidelines and consideration of both safety and occupational public health issues. We also provide discussion on considerations for future research in the field. Our intention was to provide a review of the extant literature to inform a discussion with clinical context and appropriate recommendations.

Methods

Due to the field still being in its infancy, a broad inclusion criteria was applied to the available data. The purpose was to locate human studies involving whole cannabis plant medicines and cannabis-derived isolates (singularly or in combinations) for the treatment of major psychiatric disorders or mental health symptoms. Synthetic cannabinoid analogues (e.g. nabilone) and THC isomers (e.g. dronabinol) were omitted as these fall under the auspices of a pharmaceutical-focused review (as these are classified as pharmaceutical ‘drugs’).

Major electronic databases including OVID MEDLINE, Cochrane Central Register of Controlled Trials, Health Technology Assessment Database, Allied and Complementary Medicine and PsychINFO were accessed for data up to July 2019. Initially, data were sought for meta-analytic or systematic review level epidemiological evidence (as there is sufficient data available) on the cross-sectional or longitudinal association of cannabis use and individual psychiatric disorders or symptoms. This was undertaken to assess any deleterious relationship between cannabis and psychiatric disorders. We then specifically sought any literature involving interventional human trials and observational studies, including case studies (due to deficient randomised controlled trials [RCTs] in this emerging area). We included studies with any sample size or age or gender, which used either inhalant, oral, or transdermal administration of medicinal cannabis or cannabis-derived isolates. All studies in English were assessed for inclusion (see supplementary data for the PRISMA flow chart for the number of human clinical trials or case studies excluded/included). The results are presented to firstly cover the major current epidemiological evidence, and then next all available clinical trial or case study data.

The following search terms were used to locate human studies or case report publications:

  • TITLE: cannab* OR THC OR tetrahydrocannabinol OR canab*

  • AND

  • TITLE: depression or depressive or mental illness* or mental disorder* or mental health or mood disorder* or affective disorder* or anxi* or panic disorder or obsessive compulsive or adhd or attention deficit or phobi* or bipolar or psychiat* or psychological or psychosis or psychotic or schizophr* severe mental* or serious mental* or antidepress* or antipsychotic* or post traumatic* or personality disorder* or stress

In summation, 481 articles were located, which was reduced to 310 after duplicates were removed. Of these, 13 studies fitted the eligibility criteria as clinical studies of cannabis-based treatments for symptoms of psychiatric disorders. The full search and screening process is displayed in the supplementary data. There were insufficient homogenous studies to perform a meta-analysis.

Affective disorders

Anxiety

The endocannabinoid system has been found to be a modulator of anxiety and mood, with recent data showing that cannabinoids or substances which target this system may interact with specific brain regions, including the medial prefrontal cortex, amygdaloid complex, bed nucleus of stria terminalis, and hippocampus [27]. Interaction with the CB1 receptor has a modulating effect on GABAergic and Glutamatergic transmission [28], while also influencing the hypothalamic pituitary adrenal (HPA) axis, immune system activation, and neuroplastic mechanisms. In respect to specific psychotropic mechanisms of action, the anxiolytic (and antidepressant effects) may also in part be mediated via CBD’s serotonergic effects via 5-HT1A receptor activation [29], and THC’s CB1 receptor agonism [30, 31]. It is worth noting that studies have demonstrated that CBD may partially inhibit the psychoactive effects of THC, with CBD and THC having demonstrated differing symptomatic and behavioural effects on regional brain function [32,33,34,35].

As in the case of certain other psychiatric symptoms and disorders, epidemiological evidence indicates that there is a relationship between cannabis use and anxiety symptom levels. This association (assessed by Kelzior and colleagues [36] via meta-analysis of 31 studies) has to date only been found to be weak, and based largely on cross-sectional data. Thus, it may be that those with anxiety seek cannabis treatment, rather than a causal effect occurring from cannabis use. Longitudinal data is also not convincing due to the bias of one study with a large odds ratio included in their meta-analysis. However, a stronger positive association was revealed between anxiety and cannabis use disorder. Other longitudinal data involving the USA-based National Epidemiologic survey on Alcohol and Related Conditions [37] confirms there is no obvious causal inference. The study included individuals with a diagnosis of any anxiety disorder during the initial 4-year data collection period, comparing cannabis nonusers to users, and also individuals with cannabis use disorder at a later time point on a range of psychosocial measures. Results revealed that, when controlling for baseline confounders, no significant relationship was found with cannabis use and a greater frequency of anxiety.

While to date no human trials could be located for treatment of Generalised Anxiety Disorder using whole cannabis plant extracts or combined isolates, there was one study identified testing CBD (Table 1) for Social Anxiety Disorder. One small preliminary double-blind RCT compared the effects of a simulated public speaking test on treatment-naïve patients with social anxiety (n = 24) versus healthy control participants (n = 12) [38]. Each group received a single acute oral dose of CBD (600 mg) 1.5 h before the test, or matching placebo. Results revealed that pre-treatment with CBD significantly reduced anxiety, cognitive impairment and discomfort in the social anxiety group’s speech performance, and significantly decreased hyper-alertness in their anticipatory speech compared to the placebo group (which presented higher anxiety, cognitive impairment, discomfort, and higher alertness levels). Neuroimaging research has also revealed that in individuals diagnosed with social anxiety, cerebral blood flow may be altered via CBD. One study employed fMRI in 10 treatment-naïve patients with social anxiety who were given 400 mg of oral CBD or placebo in a double-blinded crossover manner. Relative to placebo, 400 mg of CBD was associated with significantly decreased subjective anxiety, with blood flow being modulated in the left parahippocampal gyrus, hippocampus, and inferior temporal gyrus, and the right posterior cingulate gyrus [39]. This suggests that CBD’s activity may occur via interaction with the limbic and paralimbic brain areas.

Table 1 Medicinal cannabis trials in mental disorders

Due to the small sample sizes, the above data needs to be considered with caution. Further, appropriate and considered treatment of anxiety disorders with cannabinoid therapies is crucial due to the complex relationship with substance use disorders, often requiring a more complex biopsychosocial approach [40]. With this context in mind, CBD (being a non-intoxicant compared to THC) may be a more preferable option, having also shown anxiolytic effects in preclinical studies [41].

In respect to planned or ongoing research, one study in Colorado USA has just commenced and is exploring the anxiolytic effects of vaporised or ingested THC/CBD in differing ratios (1:0, 1:1, 0:1) in people with mild-moderate anxiety [42]. Another study is assessing the effect of CBD on reducing symptoms of anxiety disorders in a youth cohort (12–25 years old). The Australian-based study is a 12-week open-label pilot, which aims to see if 200 mg–800 mg of oral CBD (titrated depending on age, tolerability, and efficacy) is safe and effective for a youth population [43].

Post-traumatic stress disorder (PTSD)

Whole plant cannabis use for the management of PTSD symptoms has been identified in usage analyses [44], and in particular in returned armed services veterans [45]. The proposed neurobiological mechanisms by which medicinal cannabis may assist with PTSD are varied and mostly derived from animal research. There are high concentrations of endocannabinoid receptors in the prefrontal cortex, amygdala and hippocampus [46], having a role in fear acquisition and extinction [47]. There is strong evidence revealing that a disruption of the endocannabinoid system impairs fear extinction in CB1 knockout mice, suggestive of a critical role of CB1 receptors (and thereby potentially THC) being related to the extinction of fear [48,49,50].

One survey involving a convenience sample of 170 patients via a medical cannabis dispensary in California evaluated a range of health elements, the frequency of cannabis use, and general mental health [51]. Results revealed that those with high PTSD scores (assessed via The PTSD Checklist-Civilian Version) were more likely to use cannabis to assist with mental health coping, in addition to improving sleep, when compared with those with low PTSD scores. In particular, cannabis use frequency was greater among those with high PTSD scores who often used this for improving sleep. While there is increased use of cannabis in those with PTSD, there is currently no firmly supportive epidemiological data. A cross-sectional case control study of veterans showed that regular users do not have lower PTSD symptoms than non-users [52].

A recent open label retrospective analysis of case study data from 11 adults with PTSD assessed the patients over 8 weeks of CBD treatment (capsule or spray; mean dosage at week-8 of 49 mg) [53]. Results revealed that a reduction in mean PTSD symptoms occurred in 28% of the sample, as assessed on the PTSD checklist for DSM-5 (PCL-5). Statistical data analysis was not conducted, however, and thereby it is not possible to draw firm conclusions. Another retrospective study analysing PTSD symptoms collected during 80 psychiatric evaluations of patients applying to the New Mexico Medical Cannabis Program during 2009 to 2011 [54], revealed more supportive findings. The data identified a greater than 75% reduction in Clinician Administered Posttraumatic Scale for DSM-IV (CAPS) symptom scores when patients with PTSD were using cannabis compared to when they were not. While this study had a small sample, and is a retrospective analysis that has some methodological weaknesses, a 75% reduction on the CAPS is a compelling result, and has spurred recent RCTs which are currently in recruitment [55, 56].

Depression

Phytocannabinoids and terpenes have a potential application for modulation of the endocannabinoid system and the 5HT1A receptor to provide an antidepressant effect [16]. No RCTs to date have been conducted on the primary outcome of depression. Three studies assessing oral-administered nabiximols (i.e. botanically derived preparation containing standardised levels of THC, CBD, terpenes and flavonoids from cannabis) for other conditions (multiple sclerosis and cannabis withdrawal) found no significant effect on the secondary outcome of depression [57,58,59]. It is worth noting that one study involving cancer patients using nabiximols showed a significant reduction in mood occurred for those who used the highest dose (11–16 sprays per day) compared to the placebo [60]. Further, some epidemiological evidence has revealed a greater level of depressive symptoms in heavy cannabis users compared to light-users and non-users [61]. Due to this, higher dose THC should be avoided in people with major depressive disorder (MDD) or low mood. However, a cross-sectional survey on patterns of use and perceived efficacy suggested that in over 1429 participants identified as medical cannabis users, over 50% reported using medicinal cannabis specifically for depression [62].

Insomnia

Anecdotal survey evidence abounds for the soporific effect of cannabis, with sufferers of a range of conditions including pain, anxiety and PTSD reporting that it assists in the management of insomnia [51, 63,64,65,66,67,68]. While this may commonly take the form of whole plant cannabis being administered via vaporised inhalation, isolated CBD may also be of benefit. An example case study detailed in the literature concerns a 10-year-old girl with prior early childhood trauma [69]. A trial of oral CBD oil (25 mg) resulted in a decrease in this patient’s anxiety, and improvement in the quality and quantity of her sleep. A more substantial retrospective case series of 72 adults given CBD for anxiety and sleep complaints at a psychiatric clinic (as an adjunct to usual treatment) assessed patient data monthly over 12 weeks [70]. Anxiety scores on the Hamilton Anxiety rating Scale (HAMA) decreased within the first month in 79% of the sample and remained low during the study duration. The Pittsburgh Sleep Quality Index score also improved within the first month in 67% of the sample, but fluctuated over time. It should be noted that the data were not analysed for statistical significance, and it appeared that the sub-sample presenting primarily for anxiety treatment did not fare as well as the cohort presenting primarily with sleep issues.

A study by Johnson et al. [71] tested the long-term safety and tolerability of a THC/CBD spray and a THC spray in relieving pain in patients with advanced cancer. A total of 43 patients were continued on a previous three-arm RCT involving an open label administration of a self-titrated THC/CBD spray (n = 39) or THC spray (n = 4) (2.7 mg) assessed over a 5-week period. While results revealed a consistent reduction in perceived pain, participants also reported a decrease in their insomnia, which also reflected less fatigue. Cannabinoids may have a dual effect of lessening pain (which makes it easier to sleep), in addition to their direct soporific and anxiolytic effects being mediated in part via serotonergic activity.

As detailed above, the evidence for this use is currently very weak, and to date no RCTs were located in the literature specifically assessing cannabinoid isolates or whole plant formulas. As of late 2018, there is however, a clinical trial taking place in Australia assessing cannabinoid treatment in chronic insomnia [72]. The study, based in Western Australia, is aiming to enrol 24 participants aged (25–70 years) who have insomnia (defined as difficulty initiating or maintaining sleep for 3 or more nights per week for at least 3 months). The intervention involves an oral MC extract (ZTL-101) or placebo given in a cross-over manner for a study period of approximately 2 months. Participants will be assessed via the clinically-validated insomnia scales, an actigraph watch, and will be assessed in a sleep centre after 2-weeks.

Psychotic disorders

Schizophrenia

Consistent evidence has shown that there is a relationship between schizophrenia and cannabis use [73,74,75]. Heavy cannabis use may proceed to a diagnosis of the disorder, however, increased use may also result from ‘self-medication’. Cannabis use is cross-sectionally associated with more severe symptoms of psychosis in young people who do not meet the threshold for schizophrenia, and appears to be one high-risk component for the tumescence of the disorder [76]. More importantly, there is also longitudinal data to support a causal relationship [77,78,79]. A 2016 meta-analysis showed that while general lifetime use is not cross-sectionally associated with increased risk of psychosis, there is a robust relationship demonstrated in recent or current use in ultra-high-risk (UHR) adolescents with a DSM-diagnosed cannabis use disorder [80]. A recent prominent study has corroborated this finding. Data from 11 sites across Europe and Brazil involving patients with first-episode psychosis versus population controls, revealed that daily cannabis use was associated with increased odds of a psychotic disorder occurring compared with never-users, with nearly five-times increased odds for daily use of high-potency THC types of cannabis [81]. Several academics [82,83,84,85,86] have disputed these findings and comment that while there is a relationship, cannabis use is not causally related to increased psychosis risk (potentially due to a range of confounders e.g. correlated genetic liabilities or indirect and bidirectional processes). However, di Forti and colleagues (the study authors) [87] maintain that the data does indeed support this causal association, and that other research has flawed elements (e.g. previous Mendelian Randomisation studies using imprecise measurements of cannabis use).

It is of note that schizophrenia risk alleles are linked to cannabis use in a general population [78]. Regardless, the transition rates from a general population of cannabis users to schizophrenia is very low and can be considered to be part of a constellation of various potential gene-environment interactions. Several key genes have been implicated as potentially modulating the risk of developing schizophrenia after early cannabis use: BDNF, CNR1, COMT, AKT1, and DRD2 genes [88, 89]. There is also a likely increased susceptibility when a combination of these at-risk alleles from these single nucleotide polymorphisms (SNPs) are combined with childhood trauma [90, 91].

The apprehension regarding the promotion of psychotic symptoms are primarily based on the THC constituent of cannabis, a CB1 receptor agonist, which is the primary psychoactive phytochemical. This effect has been shown to be more prominent in users of high-THC cannabis, or in chronic heavy users [92]. Thus, THC should be avoided in people with or at risk of schizophrenia. Exposure to THC increases extracellular dopamine and glutamate and decreases GABA concentrations in the prefrontal cortex [93]. A recent double-blind crossover RCT investigated whether altered striatal glutamate (measured via proton magnetic resonance spectroscopy) was a mediating biomarker from intravenously administered THC in 16 healthy participants [94]. Results revealed that that an increase in striatal glutamate levels may underlie acute cannabis-induced psychosis, while lower baseline levels may provide a valid biomarker of greater sensitivity to its acute psychotomimetic effects.

The psychotropic effects of THC may mimic the presentation of psychotic symptoms, including paranoia, sensory alteration, euphoria, and hallucinations [95]. In laboratory-based research, people with schizophrenia appear to be even more sensitive to the psychosis-inducing effects of THC than healthy controls [96]. In contrast to THC, as mentioned in the introduction, CBD may in fact provide an opposing effect to THC albeit more research into this mechanism is required. Additional effects include the inhibition of anandamide breakdown via fatty acid amide hydrolase (FAAH) blocking effects, and anti-inflammatory effects [97, 98].

CBD is well-tolerated with minimal deleterious psychoactive effects (although some psychological effects are evident due to modulation of the 5HT1A receptor and enhanced anandamide signalling) [99]. Due to this, studies have primarily employed isolated CBD, however this work could potentially be extended to formulas from whole-plant strains which are high in CBD (> 10 mg/g) and lower in THC (< 4 mg/g). These preparations may also contain other yet-to-be-studied compounds from the plant which may be beneficial for the positive or negative symptoms of schizophrenia.

In respect to current research, aside from an initial index case study conducted by Zuardi, Morais [100] in 1995, who showed that 1500 mg of CBD administered for 26 days was beneficial for treatment-resistant schizophrenia, three clinical studies exist to date. A study by Leweke, Piomelli [99] tested in a double-blind, RCT design 600–800 mg/day of oral CBD vs the antipsychotic amisulpride over 4 weeks in 42 patients. While both treatments were safe and led to significant non-differential clinical improvements, the CBD arm had a superior side-effect profile. CBD also significantly increased anandamide levels, which was associated with clinical improvement. Another double-blind parallel-group trial, involving 88 patients with schizophrenia who were given either oral CBD (1000 mg/day) or placebo adjunctively to existing antipsychotic medication revealed after 6 weeks of treatment that the CBD group had lower levels of positive psychotic symptoms on the Positive and Negative Syndrome Scale (PANSS), and were more likely to have been rated as improved via clinician-ratings [101]. While these studies were supportive of CBD, a recent double-blind RCT by Boggs, Surti [102] found no benefit for 600 mg/day of CBD in comparison to placebo. The 6-week study involving 36 patients with schizophrenia revealed that both placebo and CBD PANSS scores improved, but no Group × Time effect was evident. The CBD was well-tolerated, however, and more sedation was evident in the CBD group compared to placebo.

Furthermore, CBD may confer some protective effects in young people at clinical high-risk for psychosis (n = 33), as a recent single-dose RCT found that 600 mg of CBD temporarily normalised aberrant brain activity in the parahippocampal, striatal, and midbrain areas, which is associated with increased psychosis risk [103]. Currently, an ongoing clinical trial in the United Kingdom is assessing the efficacy of 600 mg of CBD per day for reducing symptoms of psychosis in young people at clinical high-risk for psychosis [104].

Bipolar disorder

To date no clinical trial has assessed cannabinoids for the treatment of bipolar disorder (in respect to maintaining euthymia, or as a treatment of hypo/mania or depression), although there is a potential role of the endocannabinoid system in the disorder, as detailed above. Initial case reports contend this approach may not however be of benefit [105]. Two patients diagnosed with DSM-IV Bipolar type I disorder, and presenting with mania, were provided adjunctive CBD (titrated to 1200 mg per day) after receiving placebo for an initial five-day period. On Day 31, CBD treatment was discontinued and replaced by placebo for five days. While the first patient showed symptom improvement while on olanzapine plus CBD, she showed no additional improvement during CBD monotherapy, while the second patient had no symptom improvement with any dose of CBD during the trial. Both patients tolerated CBD very well and no side-effects were reported, despite no obvious effect on reducing mania.

ADHD

Evidence has revealed that adults with ADHD may self-medicate with cannabis as a coping strategy for a range of potential effects [106]. Off-label use in the US for this application has been noted despite a relative deficit of evidence for this use [107]. One study was located, the “Experimental Medicine in ADHD-Cannabinoids” pilot RCT, using nabiximol (cannabinoid/terpene combination) oromucosal spray in 30 adults with ADHD for 6 weeks [108]. The primary outcome was cognitive performance and activity level (as measured by head movements) using the Quantitative Behavioural Test. Secondary outcomes included ADHD and emotional lability symptoms. While a trend towards significance occurred in favour of nabiximols, no significant difference was revealed on the primary outcomes. Notably, the use of nabiximols did not impair cognition. For secondary outcomes, the combination of note was associated with a nominally significant improvement in hyperactivity/impulsivity scores on the investigator-rated Conners Adult ADHD Rating Scale. The combination was well-tolerated, however, a serious adverse event involving muscular seizures/spasms occurred in the active group. While not definitive, this study provides preliminary evidence supporting the self-medication theory of cannabis use in ADHD and the need for further studies of the endocannabinoid system in ADHD. Results, however, did not meet significance following adjustment for multiple testing, and it should be recognised that the sample size was small, thus a more robust sample would be better placed to determine the true effect.

Discussion

Data synthesis

As the present data indicates, the current field of cannabinoid therapeutics in psychiatry currently provides no convincing evidentiary support for use in any mental health application. More research is urgently needed, and many RCTs are currently being undertaken; thereby the landscape will change rapidly over the next several years. Currently, the most promising (although inconclusive) evidence is for CBD as an adjunctive treatment in schizophrenia, with an additional isolated study showing efficacy in social anxiety, and weak data suggesting a potential effect for ADHD symptoms. The evidence also tentatively suggests that a role exists for cannabinoids in PTSD, and also in reducing insomnia, which may also commonly occur in chronic pain. For other plant-derived cannabinoid therapy applications for psychiatric symptoms/disorders (e.g. several affective disorders) no firm conclusions can currently be drawn.

Clinical prescriptive considerations

It initially should be recognised by clinicians that, as detailed above, weak evidence currently exists in the field, thus this prescriptive advice should be taken in the context of evolving research. The first consideration faced by a clinician (in a legal jurisdiction) with a patient who is interested (or for clinician-initiated prescription) in using cannabis medicinally, is whether this is medically appropriate for them. A thorough screening firstly needs to occur, with Canadian British Columbian Physician guidelines [109] suggesting that clinicians initially assess:

  • Age – higher-dose THC forms not advised in people < 25 years of age;

  • If a personal history or family history of psychosis is present, and if so, no THC is advised;

  • Any current or past drug or alcohol misuse or dependence (avoid especially in individuals with cannabis dependence or misuse);

  • Cardiovascular or respiratory diseases (avoid or use caution);

  • Current medications which may interact with cannabis; and

  • Pregnant or planning or conceive or breastfeeding (avoid).

Next, if no contraindication is apparent, medical consideration can be given to what potential clinical application the MC may present for, and the cannabis formulation or isolate/s that may be appropriate for them. Given the complexity of MC whole-plant formulas (and the current challenge to standardise for batch-to-batch consistency), companies have primarily tested cannabinoid isolates and analogues. The most studied including nabiximols (Sativex), nabilone (Cesamet), and dronabinol (Marinol). While this may provide more pharmacological assurity, such an approach also negates the potential of unique genetic chemovars of cannabis which may provide specific therapeutic activity due to a complex synergistic interaction of constituents (known as the entourage effect). Patient preference may also be towards vaporisation of dried raw material [110]. To this end, specific prescriptive considerations need to be adopted, including:

  • Determining patient preference regarding administration - vaporisation (via specific devices), inhalation via traditional smoking apparatuses, oral dose (i.e. capsule, oil or in some cases food product), sublingual via lozenges or sprays. Note that each has a different onset of action and half-life. Inhalants will provide a more instantaneous effect (due to the alacrity of THC decarboxylation), whereas oral forms will take longer e.g. 45–90 min to take effect. Both forms of administration may be advisable to provide flexible symptom management;

  • Patient’s personality in terms of the effects of higher THC formulas. Avoidance of higher THC formulas should occur in youth and in those with paranoid personalities;

  • Potential for abuse (with greater theoretical potential in vaporised/inhaled forms [which also carries additional general health consequences]);

  • When the application should occur with respect to occupational and carer responsibilities and driving. Note that there is the potential to prescribe different cannabis preparations which contain differing levels of THC and CBD, with higher dose THC applications being applied preferentially in the evening.

  • CBD dosage (based on current evidence) varies according to disorder, age, weight, and potentially pharmacogenetic differences. Most research tends to focus on a range of 200 mg–800 mg per day [111]. In respect to THC-containing formulas, it has been advised to be cautious exceeding 20 mg per day due to potential side effects [112], and people may find a psychotropic effect with as little as 1 mg–2.5 mg per dose.

As mentioned above, there are a myriad of potential cannabis chemovars that can be developed, each with unique medicinal applications. However, to maintain pharmacodynamic/kinetic consistency, at present, the two major constituents commonly standardised for are THC and CBD (in some cases select terpenes are also included). THC provides, as indicated above, the primary psychotropic effect, and higher doses may be preferable for the amelioration of pain and inducement of sleep [63]. Further, it may provide an acute mood elevating effect in some people, however as mentioned, this may also elicit symptoms of paranoia, anxiety, and cognitive impairment (and in higher doses may actually impair mood). This effect may potentially be opposed by CBD (and/or other less studied cannabinoids), however data is mixed as to this effect. Additionally, the findings are not clear cut, with users of cannabis (to treat anxiety) having a statistically significant preference for higher THC/lower CBD containing cannabis cultivars [113].

Safety considerations

Clinicians needs to be aware that cannabinoid therapies may elicit a range of side effects. In respect to potentially expected side-effects from cannabinoid interventions, occasional adverse effects revealed in clinical trials include co-ordination problems, dizziness, disorientation, euphoria, drowsiness or fatigue, dry mouth, nausea and gastrointestinal upsets [10]. Due to this, regular monitoring is advised, especially when commencing treatment in cannabis-naïve patients.

The previously cited report conducted by the National Academy of Sciences [6] on the health effects of cannabis and cannabinoids cites limited evidence that cannabis use increases the rates of initiation of other psychoactive drugs. Additionally, while there are concerns over the relationship with schizophrenia, no firm evidence shows any association between cannabis use and the likelihood of developing bipolar disorder. Further evidence is suggestive that smoking cannabis on a regular basis is associated with cough and phlegm production, while limited evidence exists suggestive of a statistical association between cannabis use and ischaemic stroke and/or acute myocardial infarction. Evidence is noted to exist for the association between increased cannabis use frequency and progression to developing problematic cannabis use [6], as well as potential respiratory infections/disorders (especially in the use of poor-quality raw material). Clinicians need to balance these concerns together with the potential benefits, especially regarding the potential for lesser harm from other prescriptive or illicit options in patients managing psychiatric and pain conditions.

Pharmacogenetic considerations

Increased attention to the influence of pharmacogenetics factors is advised, with several genes being identified that may differentially affect cannabinoid pharmacokinetics and pharmacodynamics. A recent review led by Hryhorowicz [114] characterised pertinent genes with relevant interaction with cannabis into three broad categories: Receptor genes (CNR1, CNR2, TRPV1, and GPR55), transporter genes (ABCB1, ABCG2, SLC6A) and pharmacokinetic/metabolism (CYP3A4, CYP2C19, CYP2C9, CYP2A6, CYP1A1, COMT, FAAH, COX2, ABHD6, ABHD12). Research into the pharmacogenomic influence is however nascent, with most of the focus being on the relationship with cannabis dependence (e.g. CNR1 receptor SNPs which shows no obvious association), or schizophrenia (COMT, DRD2 SNPs showing a stronger correlation) [81, 114]. Further exploration of FAAH SNPs differentially affecting people’s response to CBD is also of value (given its important role in inhibiting the degradation of anandamide).

Occupational and public health considerations

Occupational health and safety issues also exist in consideration with medicinal cannabis users. Workplace safety concerns have been raised in relation to the potential for medicinal cannabis use to impair judgement and psychomotor skills, especially in relation to motor vehicle use, operation of fixed and mobile plants particularly heavy industrial machinery, and the potential for risk-taking behaviors and those working in safety sensitive positions [115, 116]. Employers have a ‘duty of care’ to provide safe and healthy workplaces, which includes the management of alcohol and drug use and their potential to create unsafe workplaces or practices. Workplace drug testing (WDT) is common in some industries including mining, transportation and correctional services [117]. Employees in building, transportation, maritime and mining operations cannot use drugs, legal or illegal, if they could impair their ability to safely undertake their duties [118]. However, the presence of a drug, or its metabolite, in a person’s system is not always proportional to cognitive impairment [119]. In addition, WDT does not discriminate between recreational or medicinal use and could place medicinal cannabis patients at risk of discrimination or unfair dismissal. Implementation of WDT should be balanced with a greater knowledge on the dose response relationship between cannabis-based medicines and their potential side effects.

Medicinal cannabis patients may also be subject to mobile drug testing in jurisdictions such as Australia. The salivary testing process is inefficient for assessment of cannabis related driving impairment because the tests can trace THC in saliva for days after consumption, long after any cognitive impairment has abated. The potential impact of medicinal cannabis on function will vary with dose, the length of usage (tolerance), route of administration (oral versus smoking), [120] and saliva THC levels are not direct measures of cognitive status. Further, the concentration of THC in urine does not correlate with cognitive function [120]. Conversely, in the US, a whole blood THC level of 5 ng/mL has been established as a legal limit for driving in states where cannabis has been legalised [121]. Guidelines and strategies for the specific risk management of cannabis in the workplace have been published in North America [120,121,122,123]. However, Australia is yet to publish its own risk management guidelines relating to medicinal cannabis in the workplace, although generic workplace alcohol and drug risk management guidelines could be adapted in the interim [118].

Future research considerations

It should also be noted that the majority of studies assessing the effect of cannabis on cognitive function were undertaken with low potency THC strains (< 4% THC), [120] and further study is required for both high potency THC medicines, as well as THC-free medicines such as CBD [121]. Further, more research is needed on the dosage required (especially of CBD and lesser studied cannabinoids), the potential entourage effect, the pharmacokinetics, and the influence of pharmacogenetics on both metabolism of the cannabinoids and the pharmacodynamics. Novel trial designs are advised in some instances, particularly involving employing high-quality RCTs (or N-of-1 designs), to explore the potential benefits in psychiatric conditions.

While research is rapidly advancing, there is a challenge regarding the adequate blinding of medicinal cannabis studies (due to the obvious psychotropic effect, and lack thereof in cannabinoid-removed controls) [124]. This may be addressed via cannabis naïve participants with psychomimetic controls (e.g. atropine; these however have the innate challenges of being biologically active themselves); adequate assessment of un/blinding; and use of varying levels of THC within the study. It still should, however, be taken in the context of other psychiatric or neurological research, with opioids and benzodiazepines also eliciting an obvious psychophysiological effect, and the acceptance of the research demonstrating analgesic and anxiolytic effects, respectively.

Conclusions

Currently the evidence is nascent and too weak to recommend cannabinoid-based interventions for a range of psychiatric disorders. While encouraging, research is only just beginning to determine whether cannabis or its isolates may or may not be effective for this application, and clinicians need to be mindful of several safety considerations (as articulated above). The most promising (although inconclusive) evidence is for CBD as an adjunctive treatment in schizophrenia, with an additional isolated study showing efficacy in social anxiety, and some data suggesting a potential effect for PTSD and ADHD symptoms. The data also tentatively suggests that a role exists for cannabinoids in reducing insomnia, which may also commonly occur in chronic pain. Given the generally favorable safety profile of cannabinoids observed across the observational studies and clinical trials conducted to date, there is clearly a strong case for encouraging further research.

Availability of data and materials

No data or materials were generated for this narrative review.

Abbreviations

CBD:

cannabidiol

HPA:

hypothalamic pituitary adrenal

MC:

medicinal cannabis

PANSS:

Positive and Negative Syndrome Scale

PCL-5:

PTSD checklist for DSM-5

SNP:

single nucleotide polymorphism

THC:

tetrahydrocannabinol

UHR:

ultra-high-risk

WDT:

workplace drug testing

References

  1. Merlin M. Archaeological evidence for the tradition of psychoactive plant use in the old world. Econ Bot. 2003;57:295–323.

    Article  Google Scholar 

  2. National Academies of Sciences E, Medicine. The health effects of cannabis and cannabinoids: the current state of evidence and recommendations for research. Washington, DC: The National Academies Press; 2017. 486 p

    Google Scholar 

  3. Touwn M. The religious and medicinal uses of Cannabis in China, India and Tibet. J Psychoactive Drugs. 1981;13:23–34.

    Article  Google Scholar 

  4. Zuardi AW. History of cannabis as a medicine: a review. Rev Bras Psiquiatr. 2006;28(2):153–7.

    Article  PubMed  Google Scholar 

  5. Russo E. The pharmacological history of Cannabis. In: Pertwee R, editor. Handbook of cannabis. Oxford: Oxford University Press; 2016. p. 23–43.

    Google Scholar 

  6. Thompson R. A dictionary of Assyrian botany. British Academy: London; 1949.

    Google Scholar 

  7. Grierson G. The hemp plant in Sanskrit and hindi literature. Indian Antiqu. 1894;September:260–2.

    Google Scholar 

  8. Da Orta G. Colloquies on the simples and drugs of India. London: Henry Sotheran; 1913.

    Google Scholar 

  9. Kosiba JD, Maisto SA, Joseph W. Patient-reported use of medical cannabis for pain, anxiety, and depression symptoms: systematic review and meta-analysis. Soc Sci Med. 2019;

  10. Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, Hernandez AV, et al. Cannabinoids for medical use: a systematic review and meta-analysis. JAMA. 2015;313(24):2456–73.

    Article  CAS  PubMed  Google Scholar 

  11. Drysdale AJ, Platt B. Cannabinoids: mechanisms and therapeutic applications in the CNS. (0929–8673 (Print)).

  12. The National Institutes of Health. Marijuana and cannabinoids: a neuroscience research summit. Bethesda; 2016.

  13. Rong C, Lee Y, Carmona NE, Cha DS, Ragguett RM, Rosenblat JD, et al. Cannabidiol in medical marijuana: research vistas and potential opportunities. Pharmacol Res. 2017;121:213–8.

    Article  CAS  PubMed  Google Scholar 

  14. Abrahamov A, Abrahamov A, Mechoulam R. An efficient new cannabinoid antiemetic in pediatric oncology. Life Sci. 1995;56(23–24):2097–102.

    Article  CAS  PubMed  Google Scholar 

  15. ElSohly MA, Radwan MM, Gul W, Chandra S, Galal A. Phytochemistry of Cannabis sativa L. Prog Chem Org Nat Prod. 2017;103:1–36.

    CAS  PubMed  Google Scholar 

  16. Russo EB. Taming THC: potential cannabis synergy and phytocannabinoid-terpenoid entourage effects. Br J Pharmacol. 2011;163(7):1344–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hanus LO, Meyer SM, Munoz E, Taglialatela-Scafati O, Appendino G. Phytocannabinoids: a unified critical inventory. Nat Prod Rep. 2016;33(12):1357–92.

    Article  CAS  PubMed  Google Scholar 

  18. Blasco-Benito S, Seijo-Vila M, Caro-Villalobos M, Tundidor I, Andradas C, Garcia-Taboada E, et al. Appraising the “entourage effect”: antitumor action of a pure cannabinoid versus a botanical drug preparation in preclinical models of breast cancer. Biochem Pharmacol. 2018;

  19. Khoury JM, Neves M, Roque MAV, Queiroz DAB, Correa de Freitas AA, de Fatima A, et al. Is there a role for cannabidiol in psychiatry? World J Biol Psychiatry. 2017:1–16.

  20. Habib G, Artul S. Medical cannabis for the treatment of fibromyalgia. J Clin Rheumatol. 2018;14:14.

    Google Scholar 

  21. Miller RJ, Miller RE. Is cannabis an effective treatment for joint pain? Clin Exp Rheumatol. 2017;35 Suppl 107(5):59–67.

    Article  PubMed  Google Scholar 

  22. Kim PS, Fishman MA. Cannabis for pain and headaches: primer. Curr Pain Headache Rep. 2017;21(4):19.

    Article  PubMed  Google Scholar 

  23. Reddy DS. The utility of cannabidiol in the treatment of refractory epilepsy. Clin Pharmacol Ther. 2017;101(2):182–4.

    Article  CAS  PubMed  Google Scholar 

  24. O’Connell BK, Gloss D, Devinsky O. Cannabinoids in treatment-resistant epilepsy: a review. Epilepsy Behav E&B. 2017;70(Pt B):341–8.

    Article  Google Scholar 

  25. Landa L, Jurica J, Sliva J, Pechackova M, Demlova R. Medical cannabis in the treatment of cancer pain and spastic conditions and options of drug delivery in clinical practice. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2018;19:19.

    Google Scholar 

  26. Lintzeris N, Driels J, Elias N, Arnold JC, McGregor IS, Allsop DJ. Medicinal cannabis in Australia, 2016: the Cannabis as Medicine Survey (CAMS-16). Med J Aust. 2018;209(5):211–6.

    Article  PubMed  Google Scholar 

  27. Lisboa SF, Gomes FV, Terzian AL, Aguiar DC, Moreira FA, Resstel LB, et al. The endocannabinoid system and anxiety. Vitam Horm. 2017;103:193–279.

    Article  CAS  PubMed  Google Scholar 

  28. Ruehle S, Rey AA, Remmers F, Lutz B. The endocannabinoid system in anxiety, fear memory and habituation. J Psychopharmacol (Oxford, UK). 2012;26(1):23–39.

    Article  CAS  Google Scholar 

  29. Russo EB, Burnett A, Hall B, Parker KK. Agonistic properties of cannabidiol at 5-HT1a receptors. Neurochem Res. 2005;30(8):1037–43.

    Article  CAS  PubMed  Google Scholar 

  30. Howlett AC, Blume LC, Dalton GD. CB(1) cannabinoid receptors and their associated proteins. Curr Med Chem. 2010;17(14):1382–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153(2):199–215.

    Article  CAS  PubMed  Google Scholar 

  32. Bhattacharyya S, Morrison PD, Fusar-Poli P, Martin-Santos R, Borgwardt S, Winton-Brown T, et al. Opposite effects of delta-9-tetrahydrocannabinol and cannabidiol on human brain function and psychopathology. Neuropsychopharmacology. 2010;35(3):764–74.

    Article  CAS  PubMed  Google Scholar 

  33. Dalton WS, Martz R, Lemberger L, Rodda BE, Forney RB. Influence of cannabidiol on delta-9-tetrahydrocannabinol effects. Clin Pharmacol Ther. 1976;19(3):300–9.

    Article  CAS  PubMed  Google Scholar 

  34. Englund A, Morrison PD, Nottage J, Hague D, Kane F, Bonaccorso S, et al. Cannabidiol inhibits THC-elicited paranoid symptoms and hippocampal-dependent memory impairment. J Psychopharmacol. 2013;27(1):19–27.

    Article  CAS  PubMed  Google Scholar 

  35. Karniol IG, Shirakawa I, Kasinski N, Pfeferman A, Carlini EA. Cannabidiol interferes with the effects of delta 9 – tetrahydrocannabinol in man. Eur J Pharmacol. 1974;28(1):172–7.

    Article  CAS  PubMed  Google Scholar 

  36. Kedzior KK, Laeber LT. A positive association between anxiety disorders and cannabis use or cannabis use disorders in the general population--a meta-analysis of 31 studies. BMC Psychiatry. 2014;14:136.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Feingold D, Rehm J, Factor H, Redler A, Lev-Ran S. Clinical and functional outcomes of cannabis use among individuals with anxiety disorders: a 3-year population-based longitudinal study. Depress Anxiety. 2018;35(6):490–501.

    Article  PubMed  Google Scholar 

  38. Bergamaschi MM, Queiroz RH, Chagas MH, de Oliveira DC, De Martinis BS, Kapczinski F, et al. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naive social phobia patients. Neuropsychopharmacology. 2011;36(6):1219–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Crippa JA, Derenusson GN, Ferrari TB, Wichert-Ana L, Duran FL, Martin-Santos R, et al. Neural basis of anxiolytic effects of cannabidiol (CBD) in generalized social anxiety disorder: a preliminary report. J Psychopharmacol (Oxford, UK). 2011;25(1):121–30.

    Article  CAS  Google Scholar 

  40. Buckner JD, Heimberg RG, Ecker AH, Vinci C. A biopsychosocial model of social anxiety and substance use. Depress Anxiety. 2013;30(3):276–84.

    Article  PubMed  Google Scholar 

  41. Blessing EM, Steenkamp MM, Manzanares J, Marmar CR. Cannabidiol as a potential treatment for anxiety disorders. Neurotherapeutics. 2015;12(4):825–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hitchcock L. 2018. Available from https://clinicaltrials.gov/ct2/show/NCT03491384?cond=Anxiety&intr=Cannabis&rank=2.

  43. ACTRN12617000825358. The cannabidiol youth anxiety pilot study (CAPS): a 12-week open- label pilot study of the safety, tolerability and efficacy of cannabidiol for anxiety disorders.

  44. Yarnell S. The use of medicinal marijuana for posttraumatic stress disorder: a review of the current literature. Prim Care Companion CNS Disord. 2015;17(3)

  45. Betthauser K, Pilz J, Vollmer LE. Use and effects of cannabinoids in military veterans with posttraumatic stress disorder. Am J Health-Syst Pharm. 2015;72(15):1279–84.

    Article  PubMed  Google Scholar 

  46. Glass M, Dragunow M, Faull RL. Cannabinoid receptors in the human brain: a detailed anatomical and quantitative autoradiographic study in the fetal, neonatal and adult human brain. Neuroscience. 1997;77(2):299–318.

    Article  CAS  PubMed  Google Scholar 

  47. Quirk GJ, Mueller D. Neural mechanisms of extinction learning and retrieval. Neuropsychopharmacology. 2008;33(1):56–72.

    Article  PubMed  Google Scholar 

  48. Papini S, Sullivan GM, Hien DA, Shvil E, Neria Y. Toward a translational approach to targeting the endocannabinoid system in posttraumatic stress disorder: a critical review of preclinical research. Biol Psychol. 2015;104:8–18.

    Article  PubMed  Google Scholar 

  49. Marsicano G, Wotjak CT, Azad SC, Bisogno T, Rammes G, Cascio MG, et al. The endogenous cannabinoid system controls extinction of aversive memories. Nature. 2002;418(6897):530–4.

    Article  CAS  PubMed  Google Scholar 

  50. Cannich A, Wotjak CT, Kamprath K, Hermann H, Lutz B, Marsicano G. CB1 cannabinoid receptors modulate kinase and phosphatase activity during extinction of conditioned fear in mice. Learn Mem. 2004;11(5):625–32.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Bonn-Miller MO, Babson KA, Vandrey R. Using cannabis to help you sleep: heightened frequency of medical cannabis use among those with PTSD. Drug Alcohol Depend. 2014;136:162–5.

    Article  PubMed  Google Scholar 

  52. Johnson MJ, Pierce JD, Mavandadi S, Klaus J, Defelice D, Ingram E, et al. Mental health symptom severity in cannabis using and non-using Veterans with probable PTSD. J Affect Disord. 2016;190:439–42.

    Article  PubMed  Google Scholar 

  53. Elms L, Shannon S, Hughes S, Lewis N. Cannabidiol in the treatment of post-traumatic stress disorder: a case series. J Altern Complement Med. 2019;25(4):392–7.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Greer GR, Grob CS, Halberstadt AL. PTSD symptom reports of patients evaluated for the New Mexico Medical cannabis Program. J Psychoactive Drugs. 2014;46(1):73–7.

    Article  PubMed  Google Scholar 

  55. NCT02517424. Placebo-Controlled, Triple-Blind, Crossover study of the safety and efficacy of three different potencies of vaporized Cannabis in 42 participants with chronic, Treatment-Resistant Posttraumatic Stress Disorder (PTSD).

  56. NCT02759185. Placebo-Controlled, Triple-Blind, Randomized Crossover Pilot Study of the Safety and Efficacy of Four Different Potencies of Smoked Marijuana in 76 Veterans With Posttraumatic Stress Disorder (PTSD).

  57. Allsop DJ, Copeland J, Lintzeris N, Dunlop AJ, Montebello M, Sadler C, et al. Nabiximols as an agonist replacement therapy during cannabis withdrawal: a randomized clinical trial. JAMA Psychiatry. 2014;71(3):281–91.

    Article  CAS  PubMed  Google Scholar 

  58. Podda G, Constantinescu CS. Nabiximols in the treatment of spasticity, pain and urinary symptoms due to multiple sclerosis. Expert Opin Biol Ther. 2012;12(11):1517–31.

    Article  CAS  PubMed  Google Scholar 

  59. Trigo JM, Soliman A, Quilty LC, Fischer B, Rehm J, Selby P, et al. Nabiximols combined with motivational enhancement/cognitive behavioral therapy for the treatment of cannabis dependence: a pilot randomized clinical trial. PloS One. 2018;13(1)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Portenoy RK, Ganae-Motan ED, Allende S, Yanagihara R, Shaiova L, Weinstein S, et al. Nabiximols for opioid-treated cancer patients with poorly-controlled chronic pain: a randomized, placebo-controlled, graded-dose trial. J Pain. 2012;13(5):438–49.

    Article  CAS  PubMed  Google Scholar 

  61. Lev-Ran S, Roerecke M, Le Foll B, George TP, McKenzie K, Rehm J. The association between cannabis use and depression: a systematic review and meta-analysis of longitudinal studies. Psychol Med. 2014;44(4):797–810.

    Article  CAS  PubMed  Google Scholar 

  62. Sexton M, Cuttler C, Finnell JS, Mischley LK. A cross-sectional survey of medical cannabis users: patterns of use and perceived efficacy. (2378–8763 (Print)).

  63. Russo EB, Guy GW, Robson PJ. Cannabis, pain, and sleep: lessons from therapeutic clinical trials of Sativex, a cannabis-based medicine. Chem Biodivers. 2007;4(8):1729–43.

    Article  CAS  PubMed  Google Scholar 

  64. Gruber SA, Sagar KA, Dahlgren MK, Racine MT, Smith RT, Lukas SE. Splendor in the grass? A pilot study assessing the impact of medical marijuana on executive function. Front Pharmacol. 2016;7

  65. Webb CW, Webb SM. Therapeutic benefits of cannabis: a patient survey. Hawai’i J Med Public Health. 2014;73(4):109–11.

    Google Scholar 

  66. Ostadhadi S, Rahmatollahi M, Dehpour AR, Rahimian R. Therapeutic potential of cannabinoids in counteracting chemotherapy-induced adverse effects: an exploratory review. Phytother Res PTR. 2015;29(3):332–8.

    Article  CAS  PubMed  Google Scholar 

  67. Cameron C, Watson D, Robinson J. Use of a synthetic cannabinoid in a correctional population for posttraumatic stress disorder–related insomnia and nightmares, chronic pain, harm reduction, and other indications: a retrospective evaluation. J Clin Psychopharmacol. 2014;34(5):559–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Reinarman C, Nunberg H, Lanthier F, Heddleston T. Who are medical marijuana patients? Population characteristics from nine California assessment clinics. J Psychoactive Drugs. 2011;43(2):128–35.

    Article  PubMed  Google Scholar 

  69. Shannon S, Opila-Lehman J. Effectiveness of cannabidiol oil for pediatric anxiety and insomnia as part of posttraumatic stress disorder: a case report. Perm J. 2016;20(4):108–11.

    Google Scholar 

  70. Shannon S, Lewis N, Lee H, Hughes S. Cannabidiol in anxiety and sleep: a large case series. Perm J. 2019;23:18–041.

    PubMed  PubMed Central  Google Scholar 

  71. Johnson JR, Lossignol D, Burnell-Nugent M, Fallon MT. An open-label extension study to investigate the long-term safety and tolerability of THC/CBD oromucosal spray and oromucosal THC spray in patients with terminal cancer-related pain refractory to strong opioid analgesics. J Pain Symptom Manag. 2013;46(2):207–18.

    Article  CAS  Google Scholar 

  72. ACTRN12618000078257. A study to evaluate the efficacy of sublingual cannabinoid based medicine extract compared with placebo for the treatment of Insomnia.

  73. D’Souza DC, Sewell RA, Ranganathan M. Cannabis and psychosis/schizophrenia: human studies. Eur Arch Psychiatry Clin Neurosci. 2009;259(7):413–31.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Manseau MW, Goff DC. Cannabinoids and schizophrenia: risks and therapeutic potential. Neurotherapeutics. 2015;12(4):816–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Moore THM, Zammit S, Lingford-Hughes A, Barnes TRE, Jones PB, Burke M, et al. Cannabis use and risk of psychotic or affective mental health outcomes: a systematic review. Lancet. 2007;370(9584):319–28.

    Article  PubMed  Google Scholar 

  76. Carney R, Cotter J, Firth J, Bradshaw T, Yung AR. Cannabis use and symptom severity in individuals at ultra high risk for psychosis: a meta-analysis. Acta Psychiatr Scand. 2017;136(1):5–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Gage SH, Hickman M, Zammit S. Association between cannabis and psychosis: epidemiologic evidence. Biol Psychiatry. 2016;79(7):549–56.

    Article  PubMed  Google Scholar 

  78. van Winkel R, Kuepper R. Epidemiological, neurobiological, and genetic clues to the mechanisms linking cannabis use to risk for nonaffective psychosis. Annu Rev Clin Psychol. 2014;10:767–91.

    Article  PubMed  Google Scholar 

  79. Zammit S, Moore TH, Lingford-Hughes A, Barnes TR, Jones PB, Burke M, et al. Effects of cannabis use on outcomes of psychotic disorders: systematic review. Br J Psychiatry. 2008;193(5):357–63.

    Article  PubMed  Google Scholar 

  80. Kraan T, Velthorst E, Koenders L, Zwaart K, Ising HK, van den Berg D, et al. Cannabis use and transition to psychosis in individuals at ultra-high risk: review and meta-analysis. Psychol Med. 2016;46(4):673–81.

    Article  CAS  PubMed  Google Scholar 

  81. Di Forti M, Quattrone D, Freeman TP, Tripoli G, Gayer-Anderson C, Quigley H, et al. The contribution of cannabis use to variation in the incidence of psychotic disorder across Europe (EU-GEI): a multicentre case-control study. Lancet Psychiatry. 2019;6(5):427–36.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Gage SHJTLP. Cannabis and psychosis: triangulating the evidence. 2019;6(5):364–5.

  83. Sommer IE, van den Brink WIM. High-potency cannabis and incident psychosis: correcting the causal assumption. 2019;6(6):464–5.

  84. Linnman CJTLP. High-potency cannabis and incident psychosis: correcting the causal assumption. 2019;6(6):465–6.

  85. Clark CSJTLP. High-potency cannabis and incident psychosis: correcting the causal assumption. 2019;6(6):e14.

  86. Gillespie N, Pasman J, Treur J, Derks EM, Verweij K, Vink JJLP. High-potency cannabis and incident psychosis: correcting the causal assumption (vol 6, pg 464, 2019). 2019;6(8):E19-E.

    Article  PubMed  Google Scholar 

  87. Di Forti M, Morgan C, Selten JP, Lynskey M, Murray RM. High-potency cannabis and incident psychosis: correcting the causal assumption – Authors’ reply. Lancet Psychiatry. 2019;6(6):466–7.

    Article  PubMed  Google Scholar 

  88. Weiser M, Noy S. Interpreting the association between cannabis use and increased risk for schizophrenia. Dialogues Clin Neurosci. 2005;7(1):81–5.

    PubMed  PubMed Central  Google Scholar 

  89. Colizzi M, Iyegbe C, Powell J, Ursini G, Porcelli A, Bonvino A, et al. Interaction between functional genetic variation of DRD2 and cannabis use on risk of psychosis. Schizophr Bull. 2015;41(5):1171–82.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Di Forti M, Morgan C, Dazzan P, Pariante C, Mondelli V, Marques TR, et al. High-potency cannabis and the risk of psychosis. Br J Psychiatry. 2009;195(6):488–91.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Henquet C, Di Forti M, Morrison P, Kuepper R, Murray RM. Gene-environment interplay between cannabis and psychosis. Schizophr Bull. 2008;34(6):1111–21.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Marconi A, Di Forti M, Lewis CM, Murray RM, Vassos E. Meta-analysis of the association between the level of cannabis use and risk of psychosis. Schizophr Bull. 2016;42(5):1262–9.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Pistis M, Ferraro L, Pira L, Flore G, Tanganelli S, Gessa GL, et al. Δ9-Tetrahydrocannabinol decreases extracellular GABA and increases extracellular glutamate and dopamine levels in the rat prefrontal cortex: an in vivo microdialysis study. Brain Res. 2002;948(1):155–8.

    Article  CAS  PubMed  Google Scholar 

  94. Colizzi M, Weltens N, McGuire P, Lythgoe D, Williams S, Van Oudenhove L, et al. Delta-9-tetrahydrocannabinol increases striatal glutamate levels in healthy individuals: implications for psychosis. Mol Psychiatry. 2019;

  95. D’Souza DC, Perry E, MacDougall L, Ammerman Y, Cooper T, Wu YT, et al. The psychotomimetic effects of intravenous delta-9-tetrahydrocannabinol in healthy individuals: implications for psychosis. Neuropsychopharmacology. 2004;29(8):1558–72.

    Article  PubMed  CAS  Google Scholar 

  96. D’Souza DC, Abi-Saab Wm Fau – Madonick S, Madonick S Fau – Forselius-Bielen K, Forselius-Bielen K Fau – Doersch A, Doersch A Fau – Braley G, Braley G Fau – Gueorguieva R, et al. Delta-9-tetrahydrocannabinol effects in schizophrenia: implications for cognition, psychosis, and addiction. (0006–3223 (Print)).

  97. Rohleder C, Müller JK, Lange B, Leweke FM. Cannabidiol as a potential new type of an antipsychotic. A critical review of the evidence. Front Pharmacol. 2016;7

  98. Leweke FM, Mueller JK, Lange B, Rohleder C. Therapeutic potential of cannabinoids in psychosis. Biol Psychiatry. 2016;79(7):604–12.

    Article  CAS  PubMed  Google Scholar 

  99. Leweke FM, Piomelli D, Pahlisch F, Muhl D, Gerth CW, Hoyer C, et al. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl Psychiatry. 2012;2(3):e94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Zuardi AW, Morais SL, Guimarães FS, Mechoulam R. Antipsychotic effect of cannabidiol. J Clin Psychiatry. 1995;56(10):485–6.

    CAS  PubMed  Google Scholar 

  101. McGuire P, Robson P, Cubala WJ, Vasile D, Morrison PD, Barron R, et al. Cannabidiol (CBD) as an adjunctive therapy in schizophrenia: a multicenter randomized controlled trial. Am J Psychiatry. 2018;175(3):225–31.

    Article  PubMed  Google Scholar 

  102. Boggs DL, Surti T, Gupta A, Gupta S, Niciu M, Pittman B, et al. The effects of cannabidiol (CBD) on cognition and symptoms in outpatients with chronic schizophrenia a randomized placebo controlled trial. Psychopharmacology. 2018;

  103. Bhattacharyya S, Wilson R, Appiah-Kusi E, O’Neill A, Brammer M, Perez J, et al. Effect of cannabidiol on medial temporal, midbrain, and striatal dysfunction in people at clinical high risk of psychosis: a randomized clinical trial. JAMA Psychiatry. 2018;

  104. ISRCTN10334895. Cannabidiol for the treatment of patients at a high-risk of psychosis.

  105. Zuardi A, Crippa J, Dursun S, Morais S, Vilela J, Sanches R, et al. Cannabidiol was ineffective for manic episode of bipolar affective disorder. J Psychopharmacol. 2010;24:135–7.

    Article  CAS  PubMed  Google Scholar 

  106. Mitchell JT, Sweitzer MM, Tunno AM, Kollins SH, McClernon FJ. “I use weed for my ADHD”: a qualitative analysis of online forum discussions on cannabis use and ADHD. PloS one. 2016;11(5):e0156614.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Marijuana and Medicine: The Need for a Science-Based Approach: Hearing before the Subcommittee on Criminal Justice, Drug Policy and Human Resources, U.S. House of Representatives, Second Sess. (April 1, 2004).

  108. Cooper RE, Williams E, Seegobin S, Tye C, Kuntsi J, Asherson P. Cannabinoids in attention-deficit/hyperactivity disorder: a randomised-controlled trial. Eur Neuropsychopharmacol. 2017;27(8):795–808.

    Article  CAS  PubMed  Google Scholar 

  109. Beaulieu P, Boulanger A, Desroches J, Clark AJ. Medical cannabis: considerations for the anesthesiologist and pain physician. Canadian journal of anaesthesia =. J Can d’anesthesie. 2016;63(5):608–24.

    Article  Google Scholar 

  110. HelloMD. Medical marijuana: patient survey results. 2016.

    Google Scholar 

  111. Millar S, Stone N, Bellman Z, Yates A, England T, O’Sullivan S. A systematic review of cannabidiol dosing in clinical populations. Br J Clin Pharmacol. 2019;

  112. MacCallum CA, Russo EB. Practical considerations in medical cannabis administration and dosing. Eur J Intern Med. 2018;49:12–9.

    Article  CAS  PubMed  Google Scholar 

  113. Kamal BS, Kamal F, Lantela DE. Cannabis and the anxiety of fragmentation-a systems approach for finding an anxiolytic cannabis chemotype. Front Neurosci. 2018;12:730.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Hryhorowicz S, Walczak M, Zakerska-Banaszak O, Slomski R, Skrzypczak-Zielinska M. Pharmacogenetics of cannabinoids. Eur J Drug Metab Pharmacokinet. 2018;43(1):1–12.

    Article  CAS  PubMed  Google Scholar 

  115. Goldsmith RS, Hartenbaum NP, Martin DW. Medical marijuana in the workforce. J Occup Environ Med. 2015;57(11):e139.

    Article  PubMed  Google Scholar 

  116. Bowles NP, Herzig MX, Shea SA. Recent legalization of cannabis use: effects on sleep, health, and workplace safety. Nat Sci Sleep. 2017;9:249–51.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Allen JG, Prichard J, Griggs L. A workplace drug testing act for Australia. Univ Qld Law J. 2013;32(2):219–37.

    Google Scholar 

  118. Safe Work Australia. Drugs and alcohol canberra: safe work Australia. 2017. Available from https://www.safeworkaustralia.gov.au/drugs-alcohol.

  119. WorkCover NSW. Alcohol and other drugs in the workplace: guide to developing a workplace alcohol and other drugs policy. Perth: WorkCover NSW; 2006.

    Google Scholar 

  120. Stringham C, Allard I, Knapp S, Minor M. Medical marijuana in the work place: keeping small business informed. Small Bus Inst J. 2017;13(1):16.

    Google Scholar 

  121. Goldsmith RS, Targino MC, Fanciullo GJ, Martin DW, Hartenbaum NP, White JM, et al. Medical marijuana in the workplace: challenges and management options for occupational physicians. J Occup Environ Med. 2015;57(5):518.

    Article  PubMed  PubMed Central  Google Scholar 

  122. CCOHS. Workplace strategies: risk of impairment from cannabis. In: Safety CCfOHa. Ontario: Candian Centre for Occupational Health and Safety; 2018.

    Google Scholar 

  123. Phillips JA, Holland MG, Baldwin DD, Gifford-Meuleveld L, Mueller KL, Perkison B, et al. Marijuana in the workplace: guidance for occupational health professionals and employers: joint guidance statement of the American Association of Occupational Health Nurses and the American College of Occupational and Environmental Medicine. Workplace Health Saf. 2015;63(4):139–64.

    Article  PubMed  Google Scholar 

  124. Casarett D. The achilles heel of medical cannabis research—inadequate blinding of placebo-controlled trials. JAMA Intern Med. 2018;178(1):9–10.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

JS1 is supported by an NHMRC Clinical Research Fellowship (APP112500), JF is supported by a Blackmores Institute Fellowship.

Funding

No funding was solicited or received for this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

The literature search was performed by JS1 and DK. JS1, JS2, DK, MD, and JF contributed to the review of data and preparation of the manuscript. Each author read and approved the final manuscript prior to submission.

Corresponding author

Correspondence to Jerome Sarris.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

JS1 and JS2 conduct cannabis research as university members of the Australia Medicinal Cannabis Research and Education Collaboration (www.AMCREC.org). JS1 has received consultancy payment from a cannabis manufacturing company as an independent scientific advisor (Australian Natural Therapeutics Group); this was however received many months after the creation and submission of this publication and was unrelated to this manuscript. He has also received either presentation honoraria, travel support, clinical trial grants, book royalties, or independent consultancy payments from: Integria Healthcare & MediHerb, Pfizer, Scius Health, Key Pharmaceuticals, Research Reviews, Taki Mai, Fiji Kava, Bioceuticals & Blackmores, Soho-Flordis, Healthworld, HealthEd, HealthMasters, Kantar Consulting, Elsevier, Chaminade University, International Society for Affective Disorders, Complementary Medicines Australia, SPRIM, Terry White Chemists, ANS, Society for Medicinal Plant and Natural Product Research, Sanofi-Aventis, Omega-3 Centre, the National Health and Medical Research Council, CR Roper Fellowship. JS2 has sat on the Scientific Advisory Board for BioCeuticals.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sarris, J., Sinclair, J., Karamacoska, D. et al. Medicinal cannabis for psychiatric disorders: a clinically-focused systematic review. BMC Psychiatry 20, 24 (2020). https://doi.org/10.1186/s12888-019-2409-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12888-019-2409-8

Keywords