Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Feb 18.
Published in final edited form as: Am J Drug Alcohol Abuse. 2019 Jul 31;45(6):563–579. doi: 10.1080/00952990.2019.1634086

Cannabis effects on brain structure, function, and cognition: considerations for medical uses of cannabis and its derivatives

Alison C Burggren a, Anaheed Shirazi b, Nathaniel Ginder b, Edythe D London b,c
PMCID: PMC7027431  NIHMSID: NIHMS1557646  PMID: 31365275

Abstract

Background

Cannabis is the most widely used illicit substance worldwide, and legalization for recreational and medical purposes has substantially increased its availability and use in the United States.

Objectives

Decades of research have suggested that recreational cannabis use confers risk for cognitive impairment across various domains, and structural and functional differences in the brain have been linked to early and heavy cannabis use.

Methods

With substantial evidence for the role of the endocannabinoid system in neural development and understanding that brain development continues into early adulthood, the rising use of cannabis in adolescents and young adults raises major concerns. Yet some formulations of cannabinoid compounds are FDA-approved for medical uses, including applications in children.

Results

Potential effects on the trajectory of brain morphology and cognition, therefore, should be considered. The goal of this review is to update and consolidate relevant findings in order to inform attitudes and public policy regarding the recreational and medical use of cannabis and cannabinoid compounds.

Conclusions

The findings point to considerations for age limits and guidelines for use.

Keywords: Cannabis, brain, morphology, endocannabinoid, tetrahydrocannabinol, cannabidiol, medical marijuana, cognition

Introduction

Cannabis (Cannabis sativa) has been used for centuries as a source of fibers, food, oil, and medicine, as well as for recreational and religious purposes (1). It contains over 500 identified natural compounds, including cannabinoids, terpenoids, flavonoids, and alkaloids (2,3). Among these, Δ9-tetrahydrocannabinol (THC), the primary psychoactive ingredient, has promoted widespread recreational use and misuse of the plant. Cannabis is the most widely used illicit substance worldwide, with an estimated 183 million past-year users in 2017 (4). In the United States, 18% of persons 12 or more years of age reported previous month use, and 1.5% in this age category met diagnostic criteria for Cannabis Use Disorder (4). The Drug Enforcement Agency has placed cannabis in Schedule I of the Controlled Substances Act, but 10 states have legalized its recreational use, and 32 have legalized its use for medicinal purposes.

Because of the increased and widespread availability and use of cannabis, and FDA-approved medical uses of cannabinoid compounds, information regarding potential untoward effects and safety limits is needed to guide public policy. Of primary concern are potential effects on the brain and cognition, which are reviewed here.

Cannabis and the endocannabinoid system

The endocannabinoid system is phylogenetically old, having been identified in the most primitive animals with a neuronal network. In animals, N-arachidonoylethanolamine (anandamide) and 2-arachidonoyl glycerol (2-AG) are the major endocannabinoids. Many of their effects and those of phytocannabinoids are mediated by CB1 and CB2 receptors, which primarily couple to G proteins of the Gi and G0 classes, although some cannabinoids engage other receptors (i.e., transient receptor potential channels and peroxisome proliferator-activated receptors) (5,6). CB1 receptors, which mediate many of the psychoactive effects of cannabinoids, are found in high densities in several brain regions and the eye, and in lower densities throughout the body (6,7). Among the most abundant G protein-coupled receptors in the brain (5), they are localized primarily to neurons but also are expressed in glia (8). CB2 receptors are found in immune cells and in some neurons. The endocannabinoid system comprises these receptors, the endocannabinoids anandamide and 2-AG, and the enzymes that regulate their production and degradation (9,10).

Within the brain, CB1 receptors are expressed in cortical areas involved in higher cognitive functions, midbrain regions associated with motor control, and hindbrain regions that participate in control of motor and sensory functions of the autonomic nervous system (11). The endocannabinoid system plays a role in homeostasis and neuroplasticity, including neurogenesis and refinement of neuronal connections (1214). Increased endocannabinoid signaling is associated with reduced stress response, improved emotion regulation, and increased reward signaling (12,15). Endocannabinoids modulate the function of diverse neurotransmitter systems, some of which may have opposing roles. To the extent that exogenously administered cannabinoids affect the same targets, the effects can resemble or diverge from those of endogenous cannabinoids depending on the respective actions at relevant receptors. Notably, the generally high potency of synthetic cannabinoids relative to the natural psychoactive Δ9-tetrahydrocannabinol (THC) in cannabis has led to substantial problems with toxicity (16).

THC acts as a partial agonist with high affinities for both CB1 and CB2 receptors (Ki = 10 nM and 24 nM, respectively). Autoradiographic and positron emission tomographic studies of the rhesus monkey and human brain have shown high densities of CB1 receptors in the cerebral cortex (cingulate gyrus, middle frontal gyrus, entorhinal cortex, and Wernicke’s area), hippocampus, caudate/putamen, globus pallidus, substantia nigra, and cerebellum (17,18). This receptor distribution is consistent with the psychoactive actions of THC and thereby with potential effects of cannabis on memory, stress-responsivity, reward, and motivated behavior, as well as self-monitoring.

The localizations of cannabinoid receptors also are consistent with important roles in reward, reinforcement, and addiction. CB2 receptors are expressed in dopamine neurons of the midbrain ventral tegmental area (19), where effects on THC receptors may modulate addiction-related behaviors, such as drug reinforcement (19). CB1 and mu receptors are co-localized in striatal output projection neurons of the nucleus accumbens and dorsal striatum, which modulate reward and habit formation, respectively (20).

CB1 and CB2 receptors are expressed from early embryonic stages, and there is evidence that endocannabinoid tone is dynamically regulated during neurogenesis, and that CB1 receptors have a regulatory role in the development of the embryologic neural system (21,22). Although findings regarding effects on infant behavior and cognition are inconsistent, evidence suggests that prenatal cannabis exposure can influence neuronal maturation and cognitive function later in life (23). Because brain development continues to proceed through adolescence and early adulthood (24,25), the rising use of cannabis in these age periods raises concerns.

Unlike THC, which is psychoactive and is self-administered by rats (e.g. (26),), CBD is considered non-psychotropic and inhibits drug-seeking and self-administration in animal models (26,27). CBD does not bind to the orthosteric binding sites of CB1 and CB2 receptors with high affinity (2729), but acts as an allosteric inhibitor of both cannabinoid receptors subtypes (27,30,31). Recent evidence indicates that CBD is a negative allosteric modulator at CB1 receptors and a partial agonist at CB2 receptors (32). CBD also is an allosteric modulator of mu and delta opioid receptors (33). Through these mechanisms, CBD may modulate opioid actions and addiction vulnerability.

Effects on brain structure

Most information on the impact of cannabis use in humans that has come from studies of individuals with chronic, heavy recreational use, and relevant reviews from 1976 to 2002 have presented some inconsistencies regarding effects on brain structure (3437). Structural neuroimaging studies provide evidence of morphological abnormalities in chronic adolescent as well as adult users (38,39); these effects may be related to the amount of cannabis exposure. One might predict that the brain regions with the highest densities of CB1 receptors would show changes due to heavy cannabis use. CB1 receptors are expressed at high levels in the temporal lobe (olfactory system, the hippocampal formation, and amygdala), the cerebellum and neocortex, and are expressed widely at lower levels in other brain regions (6,40,41).

Within the temporal lobe, which has a high density of CB1 receptors, studies have focused on the hippocampus. Despite some inconsistencies across study designs and in findings (42), structural neuroimaging has indicated abnormalities in hippocampal volume (4347) and gray matter density (48,49) of cannabis users relative to controls. Hippocampal atrophy was noted even after more than 6 months of supervised abstinence in 14 young adults with a history of heavy cannabis use (5.8 joints/day) (43). While one study reported no significant differences in hippocampal volume (50) between 22 long term, heavy cannabis users and 26 non-users, others reported finding smaller hippocampal and amygdala volumes associated with long-term cannabis use (46,47). Additionally, a longitudinal study of 20 heavy cannabis users, who reported smoking cannabis more than 5 days a week, did not find hippocampal volume alterations in cannabis users compared to controls at either baseline or at an average of 39-months post-baseline (51).

Other work showed that abnormalities in hippocampal volume and shape may be seen in individuals who have cannabis dependence, and not necessarily in those who engage in regular cannabis use without exhibiting dependence (52). Nonetheless, several review articles in the past decade (5355) have concluded that chronic cannabis use has a significant effect on hippocampal structure in adolescents and suggested that such effects reflect interactions with cannabinoid CB1 receptors, which are densely expressed in the hippocampus.

There are indications that frequent cannabis use may be particularly harmful to the adolescent brain (56). It is plausible that those who begin cannabis use early in adolescence would be more likely to become heavily dependent. Early, heavy use may then interfere with educational and vocational training, leading to long-term consequences in adulthood. From a more biological perspective, use of cannabis during critical developmental periods may cause persistent, long-term alterations in brain structure and brain function. Some studies suggest that the effects of cannabis use during adolescence could be more serious than during adulthood (57) because it may alter the trajectory of brain development (24).

Recent findings indicate that changes to hippocampal structure due to heavy cannabis use, starting in adolescence, persist well into adulthood even following abstinence for several decades (45). These changes are regionally specific to hippocampal subregions with high densities of CB1 receptors, and are not seen in parietal cortex, where the density of CB1 receptors is relatively low. Although obtained in a retrospective assessment, these results argue for more long-term prospective studies to examine the effects of adolescent use in late life, to help clarify how changes resulting from heavy cannabis use interact with brain aging.

Assessments of brain regions other than the hippocampus have revealed disparate results. Several reports found no group differences in amygdala volume (43,5862), but according to one report, the amygdala was 7.1% smaller in users than in controls (47). A large study of 22–35-year-old participants (483 in all, 282 reporting having ever used cannabis) found smaller left amygdala and right ventral striatum volumes; but the differences were within the range of normal variation, and the apparent effect on amygdala was largely attributed to shared genetic factors (63).

A study of young adults found that 20 recreational cannabis users had greater gray matter density than in 20 controls in the left nucleus accumbens extending to subcallosal cortex, hypothalamus, sublenticular-extended amygdala, and left amygdala, with shape differences in the left nucleus accumbens and right amygdala (59). Yet a recent, large study of two population-based samples 622 young Australian adults [66% female] and 474 middle-aged US males found no differences in cannabis users compared with controls in subcortical volumes (putamen, caudate, pallidum, hippocampus, amygdala, nucleus accumbens, and thalamus) (64).

Other subcortical assessments found that cannabis users had similar (60,62) or larger cerebellar volumes (44,58) than nonusers. Finally, there is some evidence for damage to white matter in specific brain regions of cannabis users as compared with controls, possibly reflecting demyelination or axonal damage resulting in altered brain connectivity and functional impairment (6568).

A multi-site MRI study in long-term cannabis users found no association between cannabis use and cortical morphology (52). Three studies found smaller orbitofrontal cortical volume in cannabis users compared to controls (44,69,70), whereas three other studies did not (58,60,71). In a recent study, gray- and white-matter volumes, cortical thickness, and gray matter density were measured in a relatively large sample of adolescent to emergent adult cannabis users: n = 147 (109 occasional users [<1–2 times/week] and 38 frequent [>3 times/week]) as compared with 634 non-users (72). No significant group differences in global or regional brain volumes were noted; the authors suggested that discrepancies with prior positive findings reflected differences in dose metrics in young cannabis users.

Although the majority of the relevant structural neuroimaging studies investigated the results of heavy, chronic cannabis use, a recent investigation showed that 46 adolescents (14 years old) who used cannabis only once or twice showed greater gray matter volume in bilateral medial temporal lobes, posterior cingulate, lingual gyri, and cerebellum (73). The authors offered that, although cannabis use has typically been associated with below control brain volumes, most previous neuroimaging research on cannabis effects involved participants who had heavy substance use histories.

Increasing usage rates by people in every age range in the United States (74,75) highlight the need to address unanswered questions about the effects of cannabis on the brain (see Table 1). Legalization has enhanced public awareness of questions about the effects of cannabis, and may also facilitate the recruitment of participants for observational studies to answer these questions. Rapidly advancing data collection supported by funding for large-scale longitudinal studies, such as the Adolescent Brain Cognitive Development Longitudinal Study, will be addressing many of these questions (Collaborative Research on Addiction at NIH).

Table 1.

Research Questions to be Addressed in Well-designed Studies of How Cannabis use Affects Brain Structure and Function.

Research Question Design Timeframe for Results
1 Does adolescent cannabis use have persistent long-term effects, including enhanced risk for schizophrenia? 1. Retrospective assessment of young adults who report early life use; assess brain metrics and typically confounding factors: education, intelligence, socioeconomic status. A few years to gather and analyze neuroimaging and cognitive assessments.
2. Prospective longitudinal design, accounting for confounding variables. 40–50 years; possible alternative is to obtain cognitive or other metrics relevant to neurological integrity before onset of use.
2 Does age of onset of cannabis use determine severity of outcomes; is earlier adolescent use linked to more serious, longer lasting brain changes? Retrospective and prospective designs in studies of young adults. A few years to a few decades. Studies investigating this hypothesis are already yielding initial results from retrospective designs.
3 What are the thresholds of frequency of use and dosage beyond which changes to brain structure or cognition become measurable? Retrospective assessment is challenging, as details regarding use may be unreliable. Legalization should facilitate prospective assessment and objective study design easier. Short-term outcomes are published. Prospective data are becoming increasingly available from studies that track dosages and frequency of usage; recreational users may feel freeer to discuss usage openly with increasing legalization.
4 Does recreational cannabis use later in life produce long-term effects on brain morphology or cognitive performance? Older, regular cannabis users can be compared to non-users after a period of abstinence (e.g., 30 days) before cognitive testing. Older adults represent one of the fastest growing usage groups since legalization, making it easier to address this question. A few years to recruit a population of older, current cannabis users. Usage rates are increasing in this population, including first-time users.
5 How do the medicinal uses of cannabis (and its constituent phytocannabinoids) compare with potential negative effects seen in recreational users? Observational studies of patient populations who have already been prescribed marijuana. Focus on: 1) older persons, who have higher rates of cancer, pain management problems, nausea, and sleep disturbance to name a few; 2) patents with pediatric epilepsy, who are non-responsive to standard forms of treatment. A few years to recruit patient populations who taking prescribed marijuana. Dissociating clinical and recreational use will be easier now that full legalization of cannabis use for any purpose negates the need to obfuscate driving factor behind usage
Randomized, blinded clinical trials of cannabis or its derivatives in conditions where standard medical treatment is ineffective. Experimental design using cannabis or cannabis-derived compounds is becoming more feasible with the lifting of federal restrictions previously placed on research involving cannabis or cannabis-derived compounds. Medical research should begin to answer many questions regarding efficacy and outcome in the coming years.
6 Does THC actively inhibit the enzyme acetylcholinesterase and prevents aggregation of the amyloid beta peptide? Studies of participants at greater risk than the general public for subsequent development of Alzheimer’s disease, half of whom report current, regular cannabis use. At least 5 years to recruit a sample of prodromal participants, who use cannabis for a longitudinal study to assess levels of amyloid-p deposition in the brain.

Investigators conducting these longitudinal studies or using translational animal models of developmental cannabinoid drug exposure are encouraged to enable assessment of causality in associations between cannabis use and alterations in brain structure and behavior. Animal models of cannabis inhalation (vs. injection) in preclinical studies may facilitate translation of results to the human population.

Cannabis effects on brain function

There is accumulating evidence that regular cannabis use can alter brain function, especially in networks that support working memory, attention, and cognitive control processing (76). Several prior reviews have addressed the functional impact of chronic cannabis use in both adults and adolescents (38,71,77,78). Functional MRI (fMRI) paired with cognitive testing typically has demonstrated abnormalities in brain activity, although the results have varied with study parameters (77), inter-subject variation (79), and amount of cannabis use (80). In comparisons of adult chronic cannabis users with healthy controls, neural activation was measured in paradigms including tests of attention (81), cognitive control (78,80), memory (8284), decision-making (8588), motor performance (89) and affective processing (90). Most of these studies have revealed changes in brain function, often without notable performance deficits, suggesting that performance may be maintained through recruitment of brain regions not typically engaged in a particular cognitive function. In contrast, schizophrenic patients with cannabis abuse had better emotional memory than schizophrenic patients who did not use cannabis, possibly by reducing negative symptoms (91).

In task-based fMRI, a response-inhibition task showed greater connectivity between a right frontal control network and substantia nigra-subthalamic nucleus network in cannabis-dependent users compared to nondependent users (69). Another study used fMRI data form 158 20-year-old men, whose cannabis use had been tracked during adolescence. Brain activity was measured while they performed a card-guessing game that assessed responses to anticipation and receipt of monetary reward (92). Functional connectivity of the nucleus accumbens to the medial prefrontal cortex was influenced by the trajectory of cannabis use during adolescence. Among the participants, those that were identified as having an escalating trajectory showed a pattern of negative functional connectivity between the nucleus accumbens and medial prefrontal cortex activity was linked to higher levels of depressive symptoms, anhedonia, and lower educational achievement. The authors suggested that cannabis use in adolescence may have consequences for mood symptoms and educational achievement in early adulthood via alterations in neural reward circuitry.

Several investigations have compared differences between cannabis users and nonusers in functional brain networks, both during task performance and in the resting state, when fMRI was used in the absence of a task. Adult cannabis users generally differed from controls in resting-state functional connectivity (for review see (93)). Cheng et al. (94) used a two-level multi-voxel pattern analysis of resting state fMRI data to classify cannabis users from control participants with an accuracy rate of 84–88% in predicting whether a single participant was a cannabis user. In another study, adult cannabis users showed stronger functional connectivity compared to controls within the default mode network, and this difference persisted after 1 month of abstinence (95). Another study found differences in resting state connectivity of the middle frontal gyrus, precentral gyrus, superior frontal gyrus, posterior cingulate cortex, cerebellum and some other regions of male heavy cannabis users compared with controls (94). Also, psychophysiological interaction analysis indicated that functional connectivity (but not regional activation) in the reward network differentiated dependent from non-dependent cannabis users in a cannabis cue paradigm (76). Finally, functional connectivity of the ventral striatum and midbrain, key brain areas for reward circuitry, as well as the brainstem and lateral thalamus was stronger in cannabis users than in controls (38,71,96).

In adolescents, heavy cannabis use was most commonly linked with abnormal frontoparietal network activity, but these findings may reflect a compensatory mechanism, particularly in prefrontal cortex (97103), to maintain behavioral performance. Studies of memory (84,104,105), attention (106), decision-making (107), and inhibitory control (108) in adolescents all demonstrate abnormal functional activation patterns. Similar to studies in adults, many task-based fMRI studies also found intact behavioral performance (88,89,106,109,110) in adolescent cannabis users compared to controls. Thus, the adolescent brain apparently achieves some level of reorganization, engaging regions not typically involved in performing a particular task (71,105). Whether such a compensation extends into adulthood after prolonged usage is questionable, and a mechanistic clarification of how long-term usage and prolonged functional brain alterations transform behavioral or cognitive output will require further investigation.

The application of fMRI is only beginning to address the neural mechanisms associated with the cognitive consequences observed in cannabis users, and more work is needed to elucidate the links between cannabis use, brain function and cognitive output. Factors to consider in future research include age of onset, mode of use, frequency and extent of cannabis use, recovery of function with abstinence, composition of the cannabis product. The implications of functional brain changes resulting from cannabis use are yet to be determined, but changes in brain activity may be an early indicator of long-term consequences before cognitive deficits are measurable (42).

Cannabis use and cognition

Research to date has suggested that acute and chronic use of cannabis leads to cognitive impairments (111,112). Various factors, including sex differences and genetic variations may influence these effects (113,114).

Acute effects

There is substantial evidence that acute administration of cannabis or THC adversely affects executive function. On tasks of planning, reasoning, interference control, and problem solving, impaired performance was observed in some (115120), but not all studies (121123) of occasional, moderate and heavy users. In test of inhibitory control, such as go/no-go or stop-signal tasks, THC administration increased reaction time in occasional and heavier cannabis users (116,118,122,124), but other findings in chronic users were mixed (125129).

Of concern are the effects of cannabis use on decision-making, especially when it involves risk-taking. Self-report questionnaires and laboratory risk-taking tasks have demonstrated differences between cannabis users and non-users, possibly related to the severity of cannabis use. The Iowa Gambling Task, delayed discounting tasks, and risk-taking decision-making tasks have been used. Acute administration of THC altered sensitivity to reward and punishment and increased risk-taking behavior in infrequent (130) and regular users (120,131,132), but not all relevant studies found impaired decision-making.

Chronic effects

Mounting evidence points to cognitive impairment after chronic, heavy cannabis use (133135), enduring beyond the acute effects, although there is also a large body of evidence with negative findings in cannabis users (136138). In prospective studies of adolescents, findings regarding general intelligence are contradictory (56,139,140), but negative effects have been observed across a wide range of cognitive domains, including, but not limited to, various aspects of memory, executive function/working memory, and processing speed. Consistency in experimental design remains a challenging aspect of studying the long-term effects of chronic cannabis use on cognition (141).

Memory has been the cognitive domain most consistently impaired, with verbal learning and memory tasks particularly sensitive to the acute (142144) and chronic (134) effects of cannabis. Several individual aspects of memory appear to be affected (46,134,145,146), with the most robust effects on verbal learning, including decrements in measures of encoding, recall, and recognition (see (134) for review). Associations between poorer performance in regular cannabis users and frequency, quantity, duration, and age of onset of cannabis use have also been reported (97,98,114,147,148). In long-term users, lasting impairments in memory and attention worsened with increasing years of regular cannabis use (135,140,149,150).

Contrary to these findings, recent studies have shown that THC can promote neurogenesis, restore memory and prevent neurodegenerative processes and cognitive decline in animal models of Alzheimer’s disease (151153). CBD also improves cognition in preclinical models of cognitive impairment in schizophrenia (154). To reconcile these seemingly contradictory results, it has been suggested that THC modulates memory and cognition in an age- and dose-dependent manner (155).

A systematic review cited evidence of deficits in attention or concentration and in memory function, with a trend toward impairments in inhibition, impulsivity, and decision-making in cannabis users (146). More cannabis use was linked with poorer episodic memory and decision-making but not inhibitory control, and sex-specific dissociations were apparent – amount of cannabis use more consistently associated with poorer episodic memory in females than males, but with poorer decision-making performance by males only (114). Notably, reviews of numerous studies of young cannabis users concluded that regular use during the adolescent and emerging adult periods may produce lasting negative effects on cognitive functioning and IQ (140,156,157). However, several recent studies have found no evidence that adolescent cannabis use or dependence was associated with IQ decline or neurocognitive performance. A study by Meier et al. of co-twins discordant for cannabis use, found little evidence that cannabis use was associated with impaired executive function between, and suggested that family background may explain the lower neurocognitive performance often reported in cannabis users (158). Another group investigated associations between adolescent cannabis use and IQ and educational attainment and found no association (159). However, without longer follow-up of adolescent cannabis users, whether impairment will emerge later in life is not clear. Additionally, impairment may depend on many factors, such as dose and route of administration, prior exposure, and blood cannabinoid concentrations before and after dosing, which have varied across studies (122).

Maladaptive decision-making has been demonstrated in cannabis users (99101,128,160,161). In one case, performance below control levels on a reward-based decision-making task was seen in participants who had been abstinent for 25 days (102). Yet, in other studies of impulsive behavior tasks (i.e., Iowa Gambling Task, Go-Stop Task, Monetary Choice Questionnaire, and Balloon Analogue Risk Task (127)) and a monetary risk-taking task (103) current users did not differ from controls. In addition, delay discounting performance did not differ between current and abstinent users and control subjects (103). Thus, whereas acute intoxication by THC appears to increase risky decision-making and sensitivity to reward, the extent to which these effects persist in chronic or abstinent users remains unclear.

The degree of recovery of function with abstinence is a topic of great interest. In a study of adolescents and emergent adults (16–25 years of age), whose abstinence was monitored over 1 month following regular use, improvements were seen in verbal memory, primarily due to improved verbal learning in the first week of abstinence (162). However, cross-sectional studies indicate that effects on attention, verbal and working memory, and psychomotor speed, but not on other cognitive domains, persist in adolescents abstinent for 28 (163) and 35 days (164). Poorer cognitive performance was associated with lifetime cumulative cannabis exposure (163) or an earlier age of onset in adolescents who were abstinent for 30 days (165), and predicted relapse to cannabis use during a 1-year follow-up.

Given the plasticity of the human brain, recovery of function might be expected, and some data support this (166,167), but evidence for persistent cognitive deficits due to cannabis use continues to emerge. Neither the precise metrics of cannabis use required for the persistence of these deficits nor the neural mechanisms underlying them are known. A recent meta-analysis indicated that in addition to small sample size in cross-sectional studies, overlooking effects of abstinence on recovery may falsely magnify the apparent severity and persistence of cognitive deficits associated with cannabis use (168). Future studies monitoring cognitive performance through prolonged periods of abstinence from chronic cannabis use are required to address these pressing questions.

One contributor to the lack of clarity about the effects of cannabis use on cognition may be considerable heterogeneity in the composition of cannabis (169). One relevant study showed greater memory impairment as well as indices of depression and anxiety associated with using cannabis of higher THC content compared to varieties containing lower THC and higher levels of CBD (149). Future research should attempt to quantify the composition of cannabis in prospective studies in order to address this issue.

Therapeutic use of cannabis and its constituents

Preliminary studies of medical marijuana suggest a variety of benefits, including amelioration of chronic pain, inflammation, spasticity, and other conditions commonly seen in physical therapy practice (170). As of December 21, 2018, 756 trials of cannabis were registered on clinicaltrials.gov for evaluation in a variety of conditions, including the following: neuropathic pain, side effects of cancer chemotherapy, schizophrenia, bipolar disorder, major depressive disorder, Tourette syndrome, retinal degeneration, and tinnitus. Practitioners prescribing cannabis are advised to consider the aforementioned cognitive effects, as well as effects on coordination and balance (145,171173), and on cardiovascular (174,175) and pulmonary function (176,177).

Although evidence suggests that heavy, recreational cannabis use is linked to cognitive deficits and potentially untoward neural changes as outlined above, findings from studies of recreational cannabis use may not be applicable to medical marijuana (178). One study examined whether patients receiving medical marijuana would exhibit improvement in cognitive functioning, perhaps related to primary symptom alleviation (179). The results suggested that these patients experienced improvement in measures of executive functioning, in addition to positive changes in some aspects of quality of life. Few studies beyond this have directly examined the potential impact of medical marijuana on cognitive performance, and further research is needed to clarify the specific neural and cognitive impact of medical marijuana use and how it compares to recreational use.

The ability of cannabinoids to modulate neurotransmission, and to act as anti-inflammatory and antioxidant agents has prompted investigators to evaluate their neuroprotective role in injured brain. Traumatic brain injury and stroke are major causes of death and disability worldwide, with associated long-term cognitive impairment (180,181). The important and complex role of the endocannabinoid system in acute brain injury has led to increasing research in animals and human subjects, suggesting that cannabinoids have an overall positive effect of neuroprotection in acute neuronal injury and that they may enhance neurobehavioral recovery (182185). Human clinical trials still need to validate these findings and to identify the underlying processes occurring in brain injury.

Numerous clinical trials with constituents of cannabis also are ongoing. As of April 14, 2019, 256 trials of THC are registered with clinicaltrials.gov. Aside from studies evaluating effects of THC on brain activity and various functions, such as memory retrieval and emotional processing, potential effects are being evaluated in chronic pain, bipolar disorder, and improvement of sleep. Dronabinol is FDA-approved to treat patients with Acquired Immune Deficiency Syndrome, who are experiencing anorexia and cachexia, as well as cancer patients who are undergoing chemotherapy and suffer from nausea and vomiting that is resistant to conventional antiemetic treatments. It can be used as tablets in the form of Marinol®, or as Syndros™, an oral solution. Marinol® has been assigned to Schedule III of the Controlled Substances Act, but the DEA maintains FDA-approved products of oral solutions containing dronabinol in Schedule II. Another FDA-approved THC formulation for chemotherapy-induced nausea and vomiting is nabilone (Cesamet®).

One hundred eighty-four trials of the non-intoxicating phytocannabinoid CBD are registered with clinicaltrials.gov for various conditions, including bipolar disorder, substance use disorders, anxiety, heart failure, Sturge-Weber syndrome, and Crohn’s disease. Because indications that involve epilepsy and developmental disorders have received substantial attention, we focus on these conditions below. Nabiximols, a 1:1 combination of THC and CBD, is still under study in the U.S. for treatment of chronic cancer pain and muscle spasticity in multiple sclerosis.

Pediatric epilepsy

There is a great need for safe and effective treatment of intractable childhood epilepsy, especially in cases of devastating epileptic encephalopathies, such as infantile spasms, Lennox-Gastaut syndrome (186) and Dravet syndrome (187). In spite of rather limited preclinical data and a lack of well-designed clinical trials, CBD and CBD-enriched whole cannabis plant extracts, have generated enormous interest as potential treatments for epilepsy (188). Following anecdotal reports of potential efficacy from parents who have administered these products to their children (189,190), clinical trials of multiple preparations of CBD were initiated (190192). The results showed strong efficacy for treatment of Lennox-Gastaut syndrome (193), Dravet syndrome (194), and highly-treatment resistant epilepsy in children and young adults (192), and confirmed reports from open-label studies. Among patients with Dravet syndrome, CBD treatment resulted in a greater reduction in convulsive-seizure frequency than placebo, but was associated with higher rates of adverse events (195). However, accumulating evidence led the U.S. Food and Drug Administration to approve Epidiolex®, a purified CBD-based oral solution, for the treatment of Lennox-Gastaut syndrome and Dravet syndrome on June 26, 2018. Epidiolex® has been assigned to Schedule V of the Controlled Substances Act. Longitudinal prospective studies are required to provide further clarification of the effects of this product in the population intended for its use, especially with respect to the developing brain.

Attention deficit hyperactivity disorder (ADHD)

Although there are no clinical recommendations or systematic research supporting the use of cannabis use ADHD, clinical and anecdotal evidence suggest an increasingly popular perception that cannabis is therapeutic for this disorder (196). Children and adolescents are increasingly being added to medical marijuana registries by their parents for treatment of ADHD and other developmental disorders (197). This practice is occurring despite a dearth of scientific evidence supporting a role for cannabis in ADHD treatment (198). One such study investigated the interaction of ADHD diagnosis and cannabis use in young adults and found no impact on behavioral response inhibition on a Go/NoGo task, but did find that cannabis use was associated with increased signal in the hippocampus and cerebellum during the fMRI Go/NoGo task only in cannabis-using control subjects, but not in cannabis-using ADHD participants (199). The authors suggest this may reflect a delayed maturation trajectory in ADHD participants. Another study found that cannabis use did not exacerbate ADHD-related symptoms (200), but suggested the need for longitudinal neuroimaging studies to investigate the neurodevelopmental cascade that culminates in positive and negative outcomes for those diagnosed with ADHD.

Increased risk for substance use, abuse or dependence of many illicit substances has been well-documented in adolescents and adults with a childhood diagnosis of ADHD, further clouding investigations of the effects of cannabis per se on the brain in this patient population (201203). Among illicit substances used by people with ADHD, cannabis is the most commonly used (203,204), providing opportunities for researchers to design cohort studies on the effects of cannabis in patients with ADHD. Although it is crucial to understand how cannabis use interacts with the neurocognitive vulnerabilities related to ADHD, ethical considerations would preclude assigning pediatric patients with ADHD to receive cannabis in the absence of previous use. Substantial gaps remain in examining neurocognitive and psychiatric outcomes in later life after treatment with cannabis among children and adolescents with ADHD.

Autism spectrum disorder (ASD)

There is increasing interest in cannabinoids, especially CBD as add-on treatment for the core symptoms and comorbidities of autism spectrum disorder (ASD). The endocannabinoid system is often affected in ASD patients with comorbidities, such as seizures, anxiety, cognitive impairments and sleep disturbances (205). Recent findings indicate that anandamide-mediated signaling at CB1 receptors, modulates oxytocin-dependent social reward, suggesting that deficits in the signaling mechanism of anandamide may contribute to social impairment in ASD (206,207). Additionally, epilepsy is common in ASD (20–30%) and more prevalent in individuals with autism-like behavior resulting from particular genetic predispositions, such as Angelman syndrome, Rett syndrome, or Dup15q syndrome (208). In a preclinical study that tested the efficacy of CBD in a mouse model for Dravet syndrome, CBD reduced both seizures and ASD behaviors (209). To date, however, no clinical studies have investigated the effects of any cannabinoid on epilepsy reduction specifically in ASD patients. Further preclinical and clinical studies are needed in order to examine the pros and cons of CBD and other cannabinoids in ASD before they are established as treatment for ASD symptoms and co-morbidities. While all of the medical uses of cannabis mentioned above for pediatric patients with epilepsy, ADHD or autism show merit, care is warranted in taking into account the use of cannabis on the developing adolescent brain. There is little-to-no long-term outcome data on cognition or brain structure in older patients with early-life cannabis exposure for medical intervention.

Conclusion

Decades of research have focused on the impact of recreational cannabis use, documenting decrements across various cognitive domains (e.g., memory, executive function, and likely processing speed), as well as structural and functional brain differences, which often underlie poorer cognitive performance or suggest inefficient processing in chronic, heavy users. These changes are most evident among adolescent users or those with early onset of cannabis use, as adolescence represents a critical period of neurodevelopment, making youth more vulnerable to exogenous influences, including cannabis. Accordingly, frequency and magnitude of use, product choice/potency, mode of use, and age of the consumer are all likely to influence the effects of cannabis on the brain. It is important, however, to recognize that cannabis is a complex plant comprising numerous constituents, which exhibit unique effects when studied alone as well as in the presence of other cannabinoids. Despite the range of effects conferred by individual constituents of cannabis, many of which are non-intoxicating and have no diversion potential, cannabis is currently treated as a single entity and classified as a Schedule I substance, the most restrictive drug class, which has hindered research efforts.

Current prospective studies on how cannabis exposure can impact brain structure and cognition are beginning to inform public policy, including considerations for age limits and guidelines for use. Nonetheless, additional research is needed to fully understand the impact of marijuana on the brain, especially for medical marijuana where there may be various confounding biological variables unique to individual medical conditions. Extreme care is warranted when evaluating the impact of cannabis on the still-developing adolescent brain. While recreational use among adolescents and early onset users is relatively well studied, a number of areas remain understudied and urgently need data to inform rapidly changing public policy. For example, additional research is needed to clarify the impact of moderate cannabis use, short- and long-term consequences of using high-potency products and novel delivery methods, effects of cannabis use in older adults, and the efficacy and safety of existing products as well as those in development. Additionally, considerations for preclinical models of cannabis inhalation (instead of injection) may facilitate translation of results in the human population.

Acknowledgments

Funding

Supported in part by the Thomas P. and Katherine K Pike Chair in Addiction studies (EDL), UCLA Training Program in the Translational Neuroscience of Drug Abuse [T32 DA024635-11] (AS), a NIDA K01 DA034728 award (AB), and an endowment from the Marjorie Greene Trust.

Footnotes

Disclosure Statement

The authors have no interests to declare.

References

  • 1.Piluzza G, Delogu G, Cabras A, Marceddu S, Bullitta S. Differentiation between fiber and drug types of hemp (Cannabis sativa L.) from a collection of wild and domesticated accessions. Genet Resour Crop Evol. 2013;60:2331–42. doi: 10.1007/s10722-013-0001-5. [DOI] [Google Scholar]
  • 2.Andre CM, Hausman J-F, Guerriero G. Cannabis sativa: the Plant of the thousand and one molecules. Front Plant Sci. 2016;7:19. doi: 10.3389/fpls.2016.00019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Pellati F, Brighenti V, Sperlea J, et al. New methods for the comprehensive analysis of bioactive compounds in Cannabis sativa L. (hemp). Mol Basel Switz 2018;23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.U.S. Department of Health and Human Services. National survey on drug use and health [Internet]. Rockville, MD: Substance Abuse and Mental Health Services Administration, Center for Behavioral Health Statistics and Quality; 2018. Available from: https://datafiles.samhsa.gov/. [Google Scholar]
  • 5.Djeungoue-Petga M-A, Hebert-Chatelain E. Linking mitochondria and synaptic transmission: the CB1 receptor. BioEssays News Rev Mol Cell Dev Biol. 2017;39. [DOI] [PubMed] [Google Scholar]
  • 6.Mackie K Cannabinoid receptors: where they are and what they do. J Neuroendocrinol. 2008;20:10–14. doi: 10.1111/j.1365-2826.2008.01671.x. [DOI] [PubMed] [Google Scholar]
  • 7.Straiker AJ, Maguire G, Mackie K, et al. Localization of cannabinoid CB1 receptors in the human anterior eye and retina. Invest Ophthalmol Vis Sci. 1999;40:2442–48. [PubMed] [Google Scholar]
  • 8.Bilkei-Gorzo A The endocannabinoid system in normal and pathological brain ageing. Philos Trans R Soc Lond B Biol Sci. 2012;367:3326–41. doi: 10.1098/rstb.2011.0388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Freund TF, Katona I, Piomelli D. Role of endogenous cannabinoids in synaptic signaling. Physiol Rev. 2003;83:1017–66. doi: 10.1152/physrev.00004.2003. [DOI] [PubMed] [Google Scholar]
  • 10.Regehr WG, Carey MR, Best AR. Activity-dependent regulation of synapses by retrograde messengers. Neuron. 2009;63:154–70. doi: 10.1016/j.neuron.2009.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.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;77:299–318. doi: 10.1016/S0306-4522(96)00428-9. [DOI] [PubMed] [Google Scholar]
  • 12.Befort K Interactions of the opioid and cannabinoid systems in reward: insights from knockout studies. Front Pharmacol 2015;6:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Egerton A, Allison C, Brett RR, Pratt JA. Cannabinoids and prefrontal cortical function: insights from preclinical studies. Neurosci Biobehav Rev. 2006;30:680–95. doi: 10.1016/j.neubiorev.2005.12.002. [DOI] [PubMed] [Google Scholar]
  • 14.Katona I, Freund TF. Multiple functions of endocannabinoid signaling in the brain. Annu Rev Neurosci. 2012;35:529–58. doi: 10.1146/annurev-neuro-062111-150420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hill MN, McEwen BS. Involvement of the endocannabinoid system in the neurobehavioural effects of stress and glucocorticoids. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:791–97. doi: 10.1016/j.pnpbp.2009.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Diao X, Huestis MA. New synthetic cannabinoids metabolism and strategies to best identify optimal marker metabolites. Front Chem. 2019;7:109. doi: 10.3389/fchem.2019.00109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bloomfield MAP, Hindocha C, Green SF, Wall MB, Lees R, Petrilli K, Costello H, Ogunbiyi MO, Bossong MG, Freeman TP. The neuropsychopharmacology of cannabis: A review of human imaging studies. Pharmacol Ther. 2019;195:132–61. doi: 10.1016/j.pharmthera.2018.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Burns HD, Van Laere K, Sanabria-Bohórquez S, Hamill TG, Bormans G, Eng W-S, Gibson R, Ryan C, Connolly B, Patel S, et al. [18F]MK-9470, a positron emission tomography (PET) tracer for in vivo human PET brain imaging of the cannabinoid-1 receptor. Proc Natl Acad Sci USA. 2007;104:9800–05. doi: 10.1073/pnas.0703472104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Zhang H-Y, Gao M, Liu Q-R, Bi G-H, Li X, Yang H-J, Gardner EL, Wu J, Xi Z-X. Cannabinoid CB2 receptors modulate midbrain dopamine neuronal activity and dopamine-related behavior in mice. Proc Natl Acad Sci USA. 2014;111:E5007–5015. doi: 10.1073/pnas.1413210111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Rodriguez JJ, Mackie K, Pickel VM. Ultrastructural localization of the CB1 cannabinoid receptor in mu-opioid receptor patches of the rat Caudate putamen nucleus. J Neurosci Off J Soc Neurosci. 2001;21:823–33. doi: 10.1523/JNEUROSCI.21-03-00823.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Díaz-Alonso J, Guzmán M, Galve-Roperh I. Endocannabinoids via CB1 receptors act as neurogenic niche cues during cortical development. Philos Trans R Soc Lond B Biol Sci. 2012;367:3229–41. doi: 10.1098/rstb.2011.0385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Friedrich J, Khatib D, Parsa K, Santopietro A, Gallicano GI. The grass isn’t always greener: the effects of cannabis on embryological development. BMC Pharmacol Toxicol. 2016;17:45. doi: 10.1186/s40360-016-0085-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Huizink AC. Prenatal cannabis exposure and infant outcomes: overview of studies. Prog Neuropsychopharmacol Biol Psychiatry. 2014;52:45–52. doi: 10.1016/j.pnpbp.2013.09.014. [DOI] [PubMed] [Google Scholar]
  • 24.Crews F, He J, Hodge C. Adolescent cortical development: a critical period of vulnerability for addiction. Pharmacol Biochem Behav. 2007;86:189–99. doi: 10.1016/j.pbb.2006.12.020. [DOI] [PubMed] [Google Scholar]
  • 25.Giedd JN. Structural magnetic resonance imaging of the adolescent brain. Ann N Y Acad Sci. 2004;1021:77–85. doi: 10.1196/annals.1308.009. [DOI] [PubMed] [Google Scholar]
  • 26.Wakeford AGP, Wetzell BB, Pomfrey RL, Clasen MM, Taylor WW, Hempel BJ, Riley AL. The effects of cannabidiol (CBD) on Δ9-tetrahydrocannabinol (THC) self-administration in male and female Long-Evans rats. Exp Clin Psychopharmacol. 2017;25:242–48. doi: 10.1037/pha0000135. [DOI] [PubMed] [Google Scholar]
  • 27.Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153:199–215. doi: 10.1038/bjp.2008.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Martínez-Pinilla E, Varani K, Reyes-Resina I, Angelats E, Vincenzi F, Ferreiro-Vera C, Oyarzabal J, Canela EI, Lanciego JL, Nadal X, et al. Binding and signaling studies disclose a potential allosteric site for cannabidiol in cannabinoid CB2 receptors. Front Pharmacol 2017;8:744. doi: 10.3389/fphar.2017.00744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Morales P, Reggio PH, Jagerovic N. An overview on medicinal chemistry of synthetic and natural derivatives of cannabidiol. Front Pharmacol. 2017;8:422. doi: 10.3389/fphar.2017.00422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Navarro G, Reyes-Resina I, Rivas-Santisteban R, Sánchez de Medina V, Morales P, Casano S, Ferreiro-Vera C, Lillo A, Aguinaga D, Jagerovic N, et al. Cannabidiol skews biased agonism at cannabinoid CB1 and CB2 receptors with smaller effect in CB1-CB2 heteroreceptor complexes. Biochem Pharmacol. 2018;157:148–58. doi: 10.1016/j.bcp.2018.08.046. [DOI] [PubMed] [Google Scholar]
  • 31.Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG. Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol. 2007;150:613–23. doi: 10.1038/sj.bjp.0707133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tham M, Yilmaz O, Alaverdashvili M, Kelly MEM, Denovan-Wright EM, Laprairie RB. Allosteric and orthosteric pharmacology of cannabidiol and cannabidiol-dimethylheptyl at the type 1 and type 2 cannabinoid receptors. Br J Pharmacol. 2019;176:1455–69. doi: 10.1111/bph.14440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kathmann M, Flau K, Redmer A, Tränkle C, Schlicker E. Cannabidiol is an allosteric modulator at mu- and delta-opioid receptors. Naunyn. Schmiedebergs Arch. Pharmacol. 2006;372:354–61. doi: 10.1007/s00210-006-0033-x. [DOI] [PubMed] [Google Scholar]
  • 34.Dornbush RL, Freedman AM. Chronic cannabis use: introduction. Ann N Y Acad Sci. 1976;282:vii–viii. doi: 10.1111/nyas.1976.282.issue-1. [DOI] [PubMed] [Google Scholar]
  • 35.Hollister LE. Health aspects of cannabis. Pharmacol Rev. 1986;38:1–20. [PubMed] [Google Scholar]
  • 36.Hollister LE. Health aspects of cannabis: revisited. Int J Neuropsychopharmacol. 1998;1:71–80. doi: 10.1017/S1461145798001060. [DOI] [PubMed] [Google Scholar]
  • 37.Iversen L Cannabis and the brain. Brain J Neurol. 2003;126:1252–70. doi: 10.1093/brain/awg143. [DOI] [PubMed] [Google Scholar]
  • 38.Batalla A, Bhattacharyya S, Yücel M, Fusar-Poli P, Crippa JA, Nogué S, Torrens M, Pujol J, Farré M, Martin-Santos R, et al. Structural and functional imaging studies in chronic cannabis users: a systematic review of adolescent and adult findings. PLoS One. 2013;8:e55821. doi: 10.1371/journal.pone.0055821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lorenzetti V, Chye Y, Silva P, Solowij N, Roberts CA. Does regular cannabis use affect neuroanatomy? An updated systematic review and meta-analysis of structural neuroimaging studies. Eur Arch Psychiatry Clin Neurosci. 2019;269:59–71. doi: 10.1007/s00406-019-00979-1. [DOI] [PubMed] [Google Scholar]
  • 40.Egertová M, Elphick MR. Localisation of cannabinoid receptors in the rat brain using antibodies to the intracellular C-terminal tail of CB. J Comp Neurol. 2000;422:159–71. doi:. [DOI] [PubMed] [Google Scholar]
  • 41.Katona I, Rancz EA, Acsady L, Ledent C, Mackie K, Hájos N, Freund TF. Distribution of CB1 cannabinoid receptors in the amygdala and their role in the control of GABAergic transmission. J Neurosci Off J Soc Neurosci. 2001;21:9506–18. doi: 10.1523/JNEUROSCI.21-23-09506.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Nader DA, Sanchez ZM. Effects of regular cannabis use on neurocognition, brain structure, and function: a systematic review of findings in adults. Am J Drug Alcohol Abuse. 2018;44:4–18. doi: 10.1080/00952990.2017.1306746. [DOI] [PubMed] [Google Scholar]
  • 43.Ashtari M, Avants B, Cyckowski L, Cervellione KL, Roofeh D, Cook P, Gee J, Sevy S, Kumra S. Medial temporal structures and memory functions in adolescents with heavy cannabis use. J Psychiatr Res. 2011;45:1055–66. doi: 10.1016/j.jpsychires.2011.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Battistella G, Fornari E, Annoni J-M, Chtioui H, Dao K, Fabritius M, Favrat B, Mall J-F, Maeder P, Giroud C. Long-term effects of cannabis on brain structure. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2014;39:2041–48. doi: 10.1038/npp.2014.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Burggren AC, Siddarth P, Mahmood Z, London ED, Harrison TM, Merrill DA, Small GW, Bookheimer SY. Subregional hippocampal thickness abnormalities in older adults with a history of heavy cannabis use. Cannabis Cannabinoid Res. 2018;3:242–51. doi: 10.1089/can.2018.0035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yücel M, Lorenzetti V, Suo C, Zalesky A, Fornito A, Takagi MJ, Lubman DI, Solowij N. Hippocampal harms, protection and recovery following regular cannabis use. Transl Psychiatry. 2016;6:e710. doi: 10.1038/tp.2015.201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Yücel M, Solowij N, Respondek C, et al. Regional brain abnormalities associated with long-term heavy cannabis use. Arch Gen Psychiatry. 2008;65:694–701. [DOI] [PubMed] [Google Scholar]
  • 48.Demirakca T, Sartorius A, Ende G, et al. Diminished gray matter in the hippocampus of cannabis users: possible protective effects of cannabidiol. Drug Alcohol Depend. 2011;114:242–45. [DOI] [PubMed] [Google Scholar]
  • 49.Matochik JA, Eldreth DA, Cadet J-L, Bolla KI. Altered brain tissue composition in heavy marijuana users. Drug Alcohol Depend. 2005;77:23–30. doi: 10.1016/j.drugalcdep.2004.06.011. [DOI] [PubMed] [Google Scholar]
  • 50.Tzilos GK, Cintron CB, Wood JBR, Simpson NS, Young AD, Pope HG, Yurgelun-Todd DA. Lack of hippocampal volume change in long-term heavy cannabis users. Am J Addict. 2005;14:64–72. doi: 10.1080/10550490590899862. [DOI] [PubMed] [Google Scholar]
  • 51.Koenders L, Lorenzetti V, de Haan L, Suo C, Vingerhoets W, van Den Brink W, Wiers RW, Meijer CJ, Machielsen M, Goudriaan AE, et al. Longitudinal study of hippocampal volumes in heavy cannabis users. J Psychopharmacol Oxf Engl. 2017;31:1027–34. doi: 10.1177/0269881117718380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Chye Y, Lorenzetti V, Suo C, Batalla A, Cousijn J, Goudriaan AE, Jenkinson M, Martin-Santos R, Whittle S, Yücel M, et al. Alteration to hippocampal volume and shape confined to cannabis dependence: a multi-site study. Addict Biol 2019;24:822–34. doi: 10.1111/adb.2019.24.issue-4. [DOI] [PubMed] [Google Scholar]
  • 53.Lopez-Quintero C, Pérez de Los Cobos J, Hasin DS, Okuda M, Wang S, Grant BF, Blanco C. Probability and predictors of transition from first use to dependence on nicotine, alcohol, cannabis, and cocaine: results of the National Epidemiologic Survey on Alcohol and Related Conditions (NESARC). Drug Alcohol Depend. 2011;115:120–30. doi: 10.1016/j.drugalcdep.2010.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lorenzetti V, Lubman DI, Whittle S, Solowij N, Yücel M. Structural MRI findings in long-term cannabis users: what do we know?. Subst Use Misuse. 2010;45:1787–808. doi: 10.3109/10826084.2010.482443. [DOI] [PubMed] [Google Scholar]
  • 55.Volkow ND, Compton WM, Weiss SRB. Adverse health effects of marijuana use. N Engl J Med. 2014;371:879. doi: 10.1056/NEJMoa1410490. [DOI] [PubMed] [Google Scholar]
  • 56.Mokrysz C, Freeman TP, Korkki S, Griffiths K, Curran HV. Are adolescents more vulnerable to the harmful effects of cannabis than adults? A placebo-controlled study in human males. Transl Psychiatry. 2016;6:e961. doi: 10.1038/tp.2016.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jager G, Ramsey NF. Long-term consequences of adolescent cannabis exposure on the development of cognition, brain structure and function: an overview of animal and human research. Curr Drug Abuse Rev. 2008;1:114–23. doi: 10.2174/1874473710801020114. [DOI] [PubMed] [Google Scholar]
  • 58.Cousijn J, Wiers RW, Ridderinkhof KR, van Den Brink W, Veltman DJ, Goudriaan AE. Grey matter alterations associated with cannabis use: results of a VBM study in heavy cannabis users and healthy controls. NeuroImage. 2012;59:3845–51. doi: 10.1016/j.neuroimage.2011.09.046. [DOI] [PubMed] [Google Scholar]
  • 59.Gilman JM, Kuster JK, Lee S, Lee MJ, Kim BW, Makris N, van der Kouwe A, Blood AJ, Breiter HC. Cannabis use is quantitatively associated with nucleus accumbens and amygdala abnormalities in young adult recreational users. J Neurosci Off J Soc Neurosci. 2014;34:5529–38. doi: 10.1523/JNEUROSCI.4745-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Koenders L, Cousijn J, Vingerhoets WAM, van Den Brink W, Wiers RW, Meijer CJ, Machielsen MWJ, Veltman DJ, Goudriaan AE, de Haan L, et al. grey matter changes associated with heavy cannabis use: a longitudinal sMRI study. PLoS One. 2016;11: e0152482. doi: 10.1371/journal.pone.0152482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Moreno-Alcázar A, Gonzalvo B, Canales-Rodríguez EJ, Blanco L, Bachiller D, Romaguera A, Monté-Rubio GC, Roncero C, McKenna PJ, Pomarol-Clotet E. Larger Gray matter volume in the basal ganglia of heavy Cannabis users detected by voxel-based morphometry and subcortical volumetric analysis. Front Psychiatry. 2018;9:175. doi: 10.3389/fpsyt.2018.00175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Weiland BJ, Thayer RE, Depue BE, Sabbineni A, Bryan AD, Hutchison KE. Daily marijuana use is not associated with brain morphometric measures in adolescents or adults. J Neurosci Off J Soc Neurosci. 2015;35:1505–12. doi: 10.1523/JNEUROSCI.2946-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Pagliaccio D, Barch DM, Bogdan R, Wood PK, Lynskey MT, Heath AC, Agrawal A. Shared predisposition in the association between cannabis use and subcortical brain structure. JAMA Psychiatry. 2015;72:994–1001. doi: 10.1001/jamapsychiatry.2015.1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Gillespie NA, Neale MC, Bates TC, Eyler LT, Fennema-Notestine C, Vassileva J, Lyons MJ, Prom-Wormley EC, McMahon KL, Thompson PM, et al. Testing associations between cannabis use and subcortical volumes in two large population-based samples. Addict Abingdon Engl 2018. doi: 10.1111/add.v113.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Arnone D, Barrick TR, Chengappa S, Mackay CE, Clark CA, Abou-Saleh MT. Corpus callosum damage in heavy marijuana use: preliminary evidence from diffusion tensor tractography and tract-based spatial statistics. NeuroImage 2008;41:1067–74. doi: 10.1016/j.neuroimage.2008.02.064. [DOI] [PubMed] [Google Scholar]
  • 66.Becker MP, Collins PF, Lim KO, Muetzel RL, Luciana M. Longitudinal changes in white matter microstructure after heavy cannabis use. Dev Cogn Neurosci. 2015;16:23–35. doi: 10.1016/j.dcn.2015.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Gruber SA, Silveri MM, Dahlgren MK, Yurgelun-Todd D. Why so impulsive? White matter alterations are associated with impulsivity in chronic marijuana smokers. Exp Clin Psychopharmacol. 2011;19:231–42. doi: 10.1037/a0023034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zalesky A, Solowij N, Yücel M, Lubman DI, Takagi M, Harding IH, Lorenzetti V, Wang R, Searle K, Pantelis C, et al. Effect of long-term cannabis use on axonal fibre connectivity. Brain J Neurol. 2012;135:2245–55. doi: 10.1093/brain/aws136. [DOI] [PubMed] [Google Scholar]
  • 69.Filbey F, Yezhuvath U. Functional connectivity in inhibitory control networks and severity of cannabis use disorder. Am J Drug Alcohol Abuse. 2013;39:382–91. doi: 10.3109/00952990.2013.841710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Price JS, McQueeny T, Shollenbarger S, Browning EL, Wieser J, Lisdahl KM. Effects of marijuana use on prefrontal and parietal volumes and cognition in emerging adults. Psychopharmacology (Berl.). 2015;232:2939–50. doi: 10.1007/s00213-015-3931-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Lorenzetti V, Alonso-Lana S, Youssef GJ, et al. Adolescent Cannabis use: what is the evidence for functional brain alteration?. Curr Pharm Des 2016;22:6353–65. [DOI] [PubMed] [Google Scholar]
  • 72.Scott JC, Rosen AFG, Moore TM, Roalf DR, Satterthwaite TD, Calkins ME, Ruparel K, Gur RE, Gur RC. Cannabis use in youth is associated with limited alterations in brain structure. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2019;44:1362–69. doi: 10.1038/s41386-019-0347-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Orr C, Spechler P, Cao Z, Albaugh M, Chaarani B, Mackey S, D’Souza D, Allgaier N, Banaschewski T, Bokde ALW, et al. Grey matter volume differences associated with extremely low levels of cannabis use in adolescence. J Neurosci Off J Soc Neurosci. 2019;39:1817–27. doi: 10.1523/JNEUROSCI.3375-17.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hasin DS, Saha TD, Kerridge BT, Goldstein RB, Chou SP, Zhang H, Jung J, Pickering RP, Ruan WJ, Smith SM, et al. Prevalence of Marijuana use disorders in the United States between 2001–2002 and 2012–2013. JAMA Psychiatry. 2015;72:1235–42. doi: 10.1001/jamapsychiatry.2015.1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Mauro PM, Carliner H, Brown QL, Hasin DS, Shmulewitz D, Rahim-Juwel R, Sarvet AL, Wall MM, Martins SS. Age differences in daily and nondaily Cannabis use in the United States, 2002–2014. J Stud Alcohol Drugs. 2018;79:423–31. doi: 10.15288/jsad.2018.79.423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kim D-J, Schnakenberg Martin AM, Shin Y-W, Jo HJ, Cheng H, Newman SD, Sporns O, Hetrick WP, Calkins E, O’Donnell BF. Aberrant structural-functional coupling in adult cannabis users. Hum Brain Mapp. 2019;40:252–61. doi: 10.1002/hbm.v40.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Sagar KA, Gruber SA. Interactions between recreational cannabis use and cognitive function: lessons from functional magnetic resonance imaging. Ann. N Y Acad Sci. 2018. doi: 10.1111/nyas.13990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Yanes JA, Riedel MC, Ray KL, Kirkland AE, Bird RT, Boeving ER, Reid MA, Gonzalez R, Robinson JL, Laird AR, et al. Neuroimaging meta-analysis of cannabis use studies reveals convergent functional alterations in brain regions supporting cognitive control and reward processing. J Psychopharmacol Oxf Engl. 2018;32:283–95. doi: 10.1177/0269881117744995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Atakan Z, Bhattacharyya S, Allen P, Martín-Santos R, Crippa JA, Borgwardt SJ, Fusar-Poli P, Seal M, Sallis H, Stahl D, et al. Cannabis affects people differently: inter-subject variation in the psychotogenic effects of Δ9-tetrahydrocannabinol: a functional magnetic resonance imaging study with healthy volunteers. Psychol Med. 2013;43:1255–67. doi: 10.1017/S0033291712001924. [DOI] [PubMed] [Google Scholar]
  • 80.Harding IH, Solowij N, Harrison BJ, Takagi M, Lorenzetti V, Lubman DI, Seal ML, Pantelis C, Yücel M. Functional connectivity in brain networks underlying cognitive control in chronic cannabis users. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2012;37:1923–33. doi: 10.1038/npp.2012.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Chang L, Yakupov R, Cloak C, Ernst T. Marijuana use is associated with a reorganized visual-attention network and cerebellar hypoactivation. Brain J Neurol. 2006;129:1096–112. doi: 10.1093/brain/awl064. [DOI] [PubMed] [Google Scholar]
  • 82.Jager G, Kahn RS, Van Den Brink W, Van Ree JM, Ramsey NF. Long-term effects of frequent cannabis use on working memory and attention: an fMRI study. Psychopharmacology (Berl.). 2006;185:358–68. doi: 10.1007/s00213-005-0298-7. [DOI] [PubMed] [Google Scholar]
  • 83.Jager G, Van Hell HH, De Win MML, Kahn RS, Van Den Brink W, Van Ree JM, Ramsey NF. Effects of frequent cannabis use on hippocampal activity during an associative memory task. Eur Neuropsychopharmacol J Eur Coll Neuropsychopharmacol. 2007;17:289–97. doi: 10.1016/j.euroneuro.2006.10.003. [DOI] [PubMed] [Google Scholar]
  • 84.Kanayama G, Rogowska J, Pope HG, Gruber SA, Yurgelun-Todd DA. Spatial working memory in heavy cannabis users: a functional magnetic resonance imaging study. Psychopharmacology (Berl.). 2004;176:239–47. doi: 10.1007/s00213-004-1885-8. [DOI] [PubMed] [Google Scholar]
  • 85.Bechara A, Tranel D, Damasio H. Characterization of the decision-making deficit of patients with ventromedial prefrontal cortex lesions. Brain J Neurol. 2000;123:2189–202. doi: 10.1093/brain/123.11.2189. [DOI] [PubMed] [Google Scholar]
  • 86.Nestor L, Hester R, Garavan H. Increased ventral striatal BOLD activity during non-drug reward anticipation in cannabis users. NeuroImage. 2010;49:1133–43. doi: 10.1016/j.neuroimage.2009.07.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Vaidya JG, Block RI, O’Leary DS, Ponto LB, Ghoneim MM, Bechara A. Effects of chronic marijuana use on brain activity during monetary decision-making. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2012;37:618–29. doi: 10.1038/npp.2011.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wesley MJ, Hanlon CA, Porrino LJ. Poor decision-making by chronic marijuana users is associated with decreased functional responsiveness to negative consequences. Psychiatry Res. 2011;191:51–59. doi: 10.1016/j.pscychresns.2010.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.King GR, Ernst T, Deng W, Stenger A, Gonzales RMK, Nakama H, Chang L. Altered brain activation during visuomotor integration in chronic active cannabis users: relationship to cortisol levels. J Neurosci Off J Soc Neurosci. 2011;31:17923–31. doi: 10.1523/JNEUROSCI.4148-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gruber SA, Rogowska J, Yurgelun-Todd DA. Altered affective response in marijuana smokers: an FMRI study. Drug Alcohol Depend. 2009;105:139–53. doi: 10.1016/j.drugalcdep.2009.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Bourque J, Mendrek A, Durand M, Lakis N, Lipp O, Stip E, Lalonde P, Grignon S, Potvin S. Cannabis abuse is associated with better emotional memory in schizophrenia: a functional magnetic resonance imaging study. Psychiatry Res. 2013;214:24–32. doi: 10.1016/j.pscychresns.2013.05.012. [DOI] [PubMed] [Google Scholar]
  • 92.Lichenstein SD, Musselman S, Shaw DS, Sitnick S, Forbes EE. Nucleus accumbens functional connectivity at age 20 is associated with trajectory of adolescent cannabis use and predicts psychosocial functioning in young adulthood. Addict Abingdon Engl. 2017;112:1961–70. doi: 10.1111/add.v112.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Chang L, Chronicle EP. Functional imaging studies in cannabis users. Neurosci Rev J Bringing Neurobiol Neurol Psychiatry. 2007;13:422–32. [DOI] [PubMed] [Google Scholar]
  • 94.Cheng H, Skosnik PD, Pruce BJ, Brumbaugh MS, Vollmer JM, Fridberg DJ, O’Donnell BF, Hetrick WP, Newman SD. Resting state functional magnetic resonance imaging reveals distinct brain activity in heavy cannabis users - a multi-voxel pattern analysis. J Psychopharmacol Oxf Engl. 2014;28:1030–40. doi: 10.1177/0269881114550354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Pujol J, Blanco-Hinojo L, Batalla A, López-Solà M, Harrison BJ, Soriano-Mas C, Crippa JA, Fagundo AB, Deus J, de la Torre R, et al. Functional connectivity alterations in brain networks relevant to self-awareness in chronic cannabis users. J Psychiatr Res. 2014;51:68–78. doi: 10.1016/j.jpsychires.2013.12.008. [DOI] [PubMed] [Google Scholar]
  • 96.Manza P, Tomasi D, Volkow ND. Subcortical local functional hyperconnectivity in cannabis dependence. Biol Psychiatry Cogn Neurosci Neuroimaging. 2018;3:285–93. doi: 10.1016/j.bpsc.2017.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Cuttler C, McLaughlin RJ, Graf P. Mechanisms underlying the link between cannabis use and prospective memory. PLoS One. 2012;7:e36820. doi: 10.1371/journal.pone.0036820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Solowij N, Jones KA, Rozman ME, Davis SM, Ciarrochi J, Heaven PCL, Lubman DI, Yücel M. Verbal learning and memory in adolescent cannabis users, alcohol users and non-users. Psychopharmacology (Berl.). 2011;216:131–44. doi: 10.1007/s00213-011-2203-x. [DOI] [PubMed] [Google Scholar]
  • 99.Moreno M, Estevez AF, Zaldivar F, Montes JMG, Gutiérrez-Ferre VE, Esteban L, Sánchez-Santed F, Flores P. Impulsivity differences in recreational cannabis users and binge drinkers in a university population. Drug Alcohol Depend. 2012;124:355–62. doi: 10.1016/j.drugalcdep.2012.02.011. [DOI] [PubMed] [Google Scholar]
  • 100.Solowij N, Jones KA, Rozman ME, Davis SM, Ciarrochi J, Heaven PCL, Pesa N, Lubman DI, Yücel M. Reflection impulsivity in adolescent cannabis users: a comparison with alcohol-using and non-substance-using adolescents. Psychopharmacology (Berl.). 2012;219:575–86. doi: 10.1007/s00213-011-2486-y. [DOI] [PubMed] [Google Scholar]
  • 101.Whitlow CT, Liguori A, Livengood LB, Hart SL, Mussat-Whitlow BJ, Lamborn CM, Laurienti PJ, Porrino LJ. Long-term heavy marijuana users make costly decisions on a gambling task. Drug Alcohol Depend. 2004;76:107–11. doi: 10.1016/j.drugalcdep.2004.04.009. [DOI] [PubMed] [Google Scholar]
  • 102.Verdejo-Garcia A, Benbrook A, Funderburk F, David P, Cadet J-L, Bolla KI. The differential relationship between cocaine use and marijuana use on decision-making performance over repeat testing with the Iowa Gambling Task. Drug Alcohol Depend. 2007;90:2–11. doi: 10.1016/j.drugalcdep.2007.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Gilman JM, Calderon V, Curran MT, Hopfer CJ, Mikulich-Gilbertson SK. Young adult cannabis users report greater propensity for risk-taking only in non-monetary domains. Drug Alcohol Depend. 2015;147:26–31. doi: 10.1016/j.drugalcdep.2014.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Becker B, Wagner D, Gouzoulis-Mayfrank E, Spuentrup E, Daumann J. The impact of early-onset cannabis use on functional brain correlates of working memory. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:837–45. doi: 10.1016/j.pnpbp.2010.03.032. [DOI] [PubMed] [Google Scholar]
  • 105.Jager G, Block RI, Luijten M, et al. Cannabis use and memory brain function in adolescent boys: a cross-sectional multicenter functional magnetic resonance imaging study. J Am Acad Child Adolesc Psychiatry. 2010;49:561–572, 572.e1–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Abdullaev Y, Posner MI, Nunnally R, Dishion TJ. Functional MRI evidence for inefficient attentional control in adolescent chronic cannabis abuse. Behav Brain Res. 2010;215:45–57. doi: 10.1016/j.bbr.2010.06.023. [DOI] [PubMed] [Google Scholar]
  • 107.Cousijn J, Wiers RW, Ridderinkhof KR, van Den Brink W, Veltman DJ, Porrino LJ, Goudriaan AE. Individual differences in decision making and reward processing predict changes in cannabis use: a prospective functional magnetic resonance imaging study. Addict Biol. 2013;18:1013–23. doi: 10.1111/adb.2013.18.issue-6. [DOI] [PubMed] [Google Scholar]
  • 108.Behan B, Connolly CG, Datwani S, Doucet M, Ivanovic J, Morioka R, Stone A, Watts R, Smyth B, Garavan H. Response inhibition and elevated parietal-cerebellar correlations in chronic adolescent cannabis users. Neuropharmacology. 2014;84:131–37. doi: 10.1016/j.neuropharm.2013.05.027. [DOI] [PubMed] [Google Scholar]
  • 109.Block RI, O’Leary DS, Hichwa RD, Augustinack JC, Boles Ponto LL, Ghoneim MM, Arndt S, Hurtig RR, Watkins GL, Hall JA, et al. Effects of frequent marijuana use on memory-related regional cerebral blood flow. Pharmacol Biochem Behav. 2002;72:237–50. doi: 10.1016/S0091-3057(01)00771–7. [DOI] [PubMed] [Google Scholar]
  • 110.Bolla KI, Eldreth DA, Matochik JA, CADET J. Neural substrates of faulty decision-making in abstinent marijuana users. NeuroImage. 2005;26:480–92. doi: 10.1016/j.neuroimage.2005.02.012. [DOI] [PubMed] [Google Scholar]
  • 111.Bogaty SER, Lee RSC, Hickie IB, Hermens DF. Meta-analysis of neurocognition in young psychosis patients with current cannabis use. J Psychiatr Res. 2018;99:22–32. doi: 10.1016/j.jpsychires.2018.01.010. [DOI] [PubMed] [Google Scholar]
  • 112.Gonzalez R, Pacheco-Colón I, Duperrouzel JC, Hawes SW. Does Cannabis use cause declines in neuropsychological functioning? A Review of longitudinal studies. J Int Neuropsychol Soc JINS. 2017;23:893–902. doi: 10.1017/S1355617717000789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Cosker E, Schwitzer T, Ramoz N, Ligier F, Lalanne L, Gorwood P, Schwan R, Laprévote V. The effect of interactions between genetics and cannabis use on neurocognition. Review Prog Neuropsychopharmacol Biol Psychiatry. 2018;82:95–106. doi: 10.1016/j.pnpbp.2017.11.024. [DOI] [PubMed] [Google Scholar]
  • 114.Crane NA, Schuster RM, Gonzalez R. Preliminary evidence for a sex-specific relationship between amount of cannabis use and neurocognitive performance in young adult cannabis users. J Int Neuropsychol Soc JINS. 2013;19:1009–15. doi: 10.1017/S135561771300088X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Liem-Moolenaar M, Te Beek ET, de Kam ML, Franson KL, Kahn RS, Hijman R, Touw D, van Gerven JM. Central nervous system effects of haloperidol on THC in healthy male volunteers. J Psychopharmacol Oxf Engl. 2010;24:1697–708. doi: 10.1177/0269881109358200. [DOI] [PubMed] [Google Scholar]
  • 116.Metrik J, Kahler CW, Reynolds B, McGeary JE, Monti PM, Haney M, de Wit H, Rohsenow DJ. Balanced placebo design with marijuana: pharmacological and expectancy effects on impulsivity and risk taking. Psychopharmacology (Berl.). 2012;223:489–99. doi: 10.1007/s00213-012-2740-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Morrison PD, Zois V, McKeown DA, Jennaway M, Basudewa IDG, Taylor R. The acute effects of synthetic intravenous Delta9-tetrahydrocannabinol on psychosis, mood and cognitive functioning. Psychol Med. 2009;39:1607–16. doi: 10.1017/S0033291708004893. [DOI] [PubMed] [Google Scholar]
  • 118.Ramaekers JG, Kauert G, van Ruitenbeek P, Theunissen EL, Schneider E, Moeller MR. High-potency marijuana impairs executive function and inhibitory motor control. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2006;31:2296–303. doi: 10.1038/sj.npp.1301068. [DOI] [PubMed] [Google Scholar]
  • 119.Ramaekers JG, Moeller MR, van Ruitenbeek P, Theunissen EL, Schneider E, Kauert G. Cognition and motor control as a function of Delta9-THC concentration in serum and oral fluid: limits of impairment. Drug Alcohol Depend. 2006;85:114–22. doi: 10.1016/j.drugalcdep.2006.03.015. [DOI] [PubMed] [Google Scholar]
  • 120.Weinstein A, Brickner O, Lerman H, Greemland M, Bloch M, Lester H, Chisin R, Sarne Y, Mechoulam R, Bar-Hamburger R, et al. A study investigating the acute dose-response effects of 13 mg and 17 mg Delta 9-tetrahydrocannabinol on cognitive-motor skills, subjective and autonomic measures in regular users of marijuana. J Psychopharmacol Oxf Engl. 2008;22:441–51. doi: 10.1177/0269881108088194. [DOI] [PubMed] [Google Scholar]
  • 121.Anderson BM, Rizzo M, Block RI, Pearlson GD, O’Leary DS. Sex, drugs, and cognition: effects of marijuana. J Psychoact Drugs. 2010;42:413–24. doi: 10.1080/02791072.2010.10400704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Ramaekers JG, Kauert G, Theunissen EL, Toennes SW, Moeller MR. Neurocognitive performance during acute THC intoxication in heavy and occasional cannabis users. J Psychopharmacol Oxf Engl. 2009;23:266–77. doi: 10.1177/0269881108092393. [DOI] [PubMed] [Google Scholar]
  • 123.Ranganathan M, Carbuto M, Braley G, Elander J, Perry E, Pittman B, Radhakrishnan R, Sewell RA, D’Souza DC. Naltrexone does not attenuate the effects of intravenous Δ9-tetrahydrocannabinol in healthy humans. Int J Neuropsychopharmacol. 2012;15:1251–64. doi: 10.1017/S1461145711001830. [DOI] [PubMed] [Google Scholar]
  • 124.Theunissen EL, Heckman P, de Sousa Fernandes Perna EB, Kuypers KPC, Sambeth A, Blokland A, Prickaerts J, Toennes SW, Ramaekers JG. Rivastigmine but not vardenafil reverses cannabis-induced impairment of verbal memory in healthy humans. Psychopharmacology (Berl). 2015;232:343–53. doi: 10.1007/s00213-014-3667-2. [DOI] [PubMed] [Google Scholar]
  • 125.Crane NA, Schuster RM, Fusar-Poli P, et al. Effects of cannabis on neurocognitive functioning: recent advances, neurodevelopmental influences, and sex differences. Neuropsychol Rev. 2013;23:117–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Dougherty DM, Mathias CW, Dawes MA, Furr RM, Charles NE, Liguori A, Shannon EE, Acheson A. Impulsivity, attention, memory, and decision-making among adolescent marijuana users. Psychopharmacology (Berl.). 2013;226:307–19. doi: 10.1007/s00213-012-2908-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Gonzalez R, Schuster RM, Mermelstein RJ, Vassileva J, Martin EM, Diviak KR. Performance of young adult cannabis users on neurocognitive measures of impulsive behavior and their relationship to symptoms of cannabis use disorders. J Clin Exp Neuropsychol. 2012;34:962–76. doi: 10.1080/13803395.2012.703642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Grant JE, Chamberlain SR, Schreiber L, Odlaug BL. Neuropsychological deficits associated with cannabis use in young adults. Drug Alcohol Depend. 2012;121:159–62. doi: 10.1016/j.drugalcdep.2011.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lisdahl KM, Price JS. Increased marijuana use and gender predict poorer cognitive functioning in adolescents and emerging adults. J Int Neuropsychol Soc JINS. 2012;18:678–88. doi: 10.1017/S1355617712000276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Rogers RD, Wakeley J, Robson PJ, Bhagwagar Z, Makela P. The effects of low doses of delta-9 tetrahydrocannabinol on reinforcement processing in the risky decision-making of young healthy adults. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2007;32:417–28. doi: 10.1038/sj.npp.1301175. [DOI] [PubMed] [Google Scholar]
  • 131.Lane SD, Cherek DR, Tcheremissine OV, Lieving LM, Pietras CJ. Acute marijuana effects on human risk taking. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2005;30:800–09. doi: 10.1038/sj.npp.1300620. [DOI] [PubMed] [Google Scholar]
  • 132.Vadhan NP, Hart CL, van Gorp WG, Gunderson EW, Haney M, Foltin RW. Acute effects of smoked marijuana on decision making, as assessed by a modified gambling task, in experienced marijuana users. J Clin Exp Neuropsychol. 2007;29:357–64. doi: 10.1080/13803390600693615. [DOI] [PubMed] [Google Scholar]
  • 133.Schmetzer AD. Book review: cannabis and cognitive functioning, by Nadia Solowij. Ann Clin Psychiatry. 2000;12:254–57. doi: 10.1023/A:1009051030174. [DOI] [Google Scholar]
  • 134.Solowij N, Battisti R. The chronic effects of cannabis on memory in humans: a review. Curr Drug Abuse Rev. 2008;1:81–98. doi: 10.2174/1874473710801010081. [DOI] [PubMed] [Google Scholar]
  • 135.Solowij N, Stephens R, Roffman RA, et al. Does marijuana use cause long-term cognitive deficits?. JAMA. 2002;287:2653–2654; author reply 2654. [PubMed] [Google Scholar]
  • 136.Fontes MA, Bolla KI, Cunha PJ, Almeida PP, Jungerman F, Laranjeira RR, Bressan RA, Lacerda ALT. Cannabis use before age 15 and subsequent executive functioning. Br J Psychiatry J Ment Sci. 2011;198:442–47. doi: 10.1192/bjp.bp.110.077479. [DOI] [PubMed] [Google Scholar]
  • 137.Hester R, Nestor L, Garavan H. Impaired error awareness and anterior cingulate cortex hypoactivity in chronic cannabis users. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2009;34:2450–58. doi: 10.1038/npp.2009.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Tait RJ, Mackinnon A, Christensen H. Cannabis use and cognitive function: 8-year trajectory in a young adult cohort. Addict Abingdon Engl 2011;106:2195–203. doi: 10.1111/j.1360-0443.2011.03574.x. [DOI] [PubMed] [Google Scholar]
  • 139.Jackson NJ, Isen JD, Khoddam R, Irons D, Tuvblad C, Iacono WG, McGue M, Raine A, Baker LA. Impact of adolescent marijuana use on intelligence: results from two longitudinal twin studies. Proc Natl Acad Sci USA. 2016;113:E500–508. doi: 10.1073/pnas.1516648113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Meier MH, Caspi A, Ambler A, Harrington H, Houts R, Keefe RSE, McDonald K, Ward A, Poulton R, Moffitt TE. Persistent cannabis users show neuropsychological decline from childhood to midlife. Proc Natl Acad Sci USA. 2012;109:E2657–2664. doi: 10.1073/pnas.1206820109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Mokrysz C, Freeman TP, Commentary on Meier, et al. 2018: smoke and mirrors-are adolescent cannabis users vulnerable to cognitive impairment? Addict Abingdon Engl 2018;113:266–67. doi: 10.1111/add.14055. [DOI] [PubMed] [Google Scholar]
  • 142.Fletcher PC, Honey GD. Schizophrenia, ketamine and cannabis: evidence of overlapping memory deficits. Trends Cogn Sci. 2006;10:167–74. doi: 10.1016/j.tics.2006.02.008. [DOI] [PubMed] [Google Scholar]
  • 143.Ranganathan M, D’Souza DC. The acute effects of cannabinoids on memory in humans: a review. Psychopharmacology (Berl.). 2006;188:425–44. doi: 10.1007/s00213-006-0508-y. [DOI] [PubMed] [Google Scholar]
  • 144.Solowij N, Pesa N. [Cognitive abnormalities and cannabis use]. Rev Bras Psiquiatr Sao Paulo Braz. 2010;32: S31–40. [PubMed] [Google Scholar]
  • 145.Broyd SJ, van Hell HH, Beale C, Yücel M, Solowij N. Acute and chronic effects of cannabinoids on human cognition-a systematic review. Biol Psychiatry. 2016;79:557–67. doi: 10.1016/j.biopsych.2015.12.002. [DOI] [PubMed] [Google Scholar]
  • 146.Ganzer F, Broning S, Kraft S, Sack P-M, Thomasius R. Weighing the evidence: a systematic review on long-term neurocognitive effects of Cannabis use in abstinent adolescents and adults. Neuropsychol Rev. 2016;26:186–222. doi: 10.1007/s11065-016-9316-2. [DOI] [PubMed] [Google Scholar]
  • 147.Thames AD, Arbid N, Sayegh P. Cannabis use and neurocognitive functioning in a non-clinical sample of users. Addict Behav 2014;39:994–99. doi: 10.1016/j.addbeh.2014.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Wagner D, Becker B, Gouzoulis-Mayfrank E, Daumann J. Interactions between specific parameters of cannabis use and verbal memory. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:871–76. doi: 10.1016/j.pnpbp.2010.04.004. [DOI] [PubMed] [Google Scholar]
  • 149.Morgan CJA, Gardener C, Schafer G, Swan S, Demarchi C, Freeman TP, Warrington P, Rupasinghe I, Ramoutar A, Tan N, et al. Sub-chronic impact of cannabinoids in street cannabis on cognition, psychotic-like symptoms and psychological well-being. Psychol Med. 2012;42:391–400. doi: 10.1017/S0033291711001322. [DOI] [PubMed] [Google Scholar]
  • 150.Solowij N, Stephens RS, Roffman RA, et al. Cognitive functioning of long-term heavy cannabis users seeking treatment. JAMA. 2002;287:1123–31. [DOI] [PubMed] [Google Scholar]
  • 151.Bilkei-Gorzo A, Albayram O, Draffehn A, Sanjiv K, Myrberg IH, Paulin CBJ, Heshmati Y, Hagenkort A, Kutzner J, Page BDG, et al. A chronic low dose of Δ9-tetrahydrocannabinol (THC) restores cognitive function in old mice. Nat Med. 2017;23:782–87. doi: 10.1038/nm.4265. [DOI] [PubMed] [Google Scholar]
  • 152.Martín-Moreno AM, Brera B, Spuch C, Carro E, García-García L, Delgado M, Pozo MA, Innamorato NG, Cuadrado A, de Ceballos ML. Prolonged oral cannabinoid administration prevents neuroinflammation, lowers β-amyloid levels and improves cognitive performance in Tg APP 2576 mice. J Neuroinflammation. 2012;9:8. doi: 10.1186/1742-2094-9-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Ramírez BG, Blázquez C, Gómez Del Pulgar T, et al. Prevention of Alzheimer’s disease pathology by cannabinoids: neuroprotection mediated by blockade of microglial activation. J Neurosci Off J Soc Neurosci. 2005;25:1904–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Osborne AL, Solowij N, Weston-Green K. A systematic review of the effect of cannabidiol on cognitive function: relevance to schizophrenia. Neurosci Biobehav Rev. 2017;72:310–24. doi: 10.1016/j.neubiorev.2016.11.012. [DOI] [PubMed] [Google Scholar]
  • 155.Calabrese EJ, Rubio-Casillas A. Biphasic effects of THC in memory and cognition. Eur J Clin Invest. 2018;48: e12920. doi: 10.1111/eci.2018.48.issue-5. [DOI] [PubMed] [Google Scholar]
  • 156.Levine A, Clemenza K, Rynn M, Lieberman J. Evidence for the risks and consequences of adolescent Cannabis exposure. J Am Acad Child Adolesc Psychiatry. 2017;56:214–25. doi: 10.1016/j.jaac.2016.12.014. [DOI] [PubMed] [Google Scholar]
  • 157.Lisdahl KM, Wright NE, Kirchner-Medina C, Maple KE, Shollenbarger S. Considering Cannabis: the effects of regular Cannabis use on neurocognition in adolescents and young adults. Curr Addict Rep. 2014;1:144–56. doi: 10.1007/s40429-014-0019-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Meier MH, Caspi A, Danese A, Fisher HL, Houts R, Arseneault L, Moffitt TE. Associations between adolescent cannabis use and neuropsychological decline: a longitudinal co-twin control study. Addict Abingdon Engl. 2018;113:257–65. doi: 10.1111/add.v113.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Mokrysz C, Landy R, Gage SH, Munafò MR, Roiser JP, Curran HV. Are IQ and educational outcomes in teenagers related to their cannabis use? A prospective cohort study. J Psychopharmacol Oxf Engl. 2016;30:159–68. doi: 10.1177/0269881115622241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Becker MP, Collins PF, Luciana M. Neurocognition in college-aged daily marijuana users. J Clin Exp Neuropsychol. 2014;36:379–98. doi: 10.1080/13803395.2014.893996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Fridberg DJ, Queller S, Ahn W-Y, Kim W, Bishara AJ, Busemeyer JR, Porrino L, Stout JC. Cognitive mechanisms underlying risky decision-making in chronic Cannabis users. J Math Psychol. 2010;54:28–38. doi: 10.1016/j.jmp.2009.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Schuster RM, Gilman J, Schoenfeld D, et al. One month of Cannabis abstinence in adolescents and young adults is associated with improved memory. J Clin Psychiatry. 2018;79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Medina KL, Hanson KL, Schweinsburg AD, COHEN-ZION M, NAGEL BJ, TAPERT SF. Neuropsychological functioning in adolescent marijuana users: subtle deficits detectable after a month of abstinence. J Int Neuropsychol Soc JINS. 2007;13:807–20. doi: 10.1017/S1355617707071032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Winward JL, Hanson KL, Tapert SF, Brown SA. Heavy alcohol use, marijuana use, and concomitant use by adolescents are associated with unique and shared cognitive decrements. J Int Neuropsychol Soc JINS. 2014;20:784–95. doi: 10.1017/S1355617714000666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Hooper SR, Woolley D, De Bellis MD. Intellectual, neurocognitive, and academic achievement in abstinent adolescents with cannabis use disorder. Psychopharmacology (Berl.). 2014;231:1467–77. doi: 10.1007/s00213-014-3463-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Pope HG, Gruber AJ, Hudson JI, Huestis MA, Yurgelun-Todd D. Neuropsychological performance in long-term cannabis users. Arch Gen Psychiatry. 2001;58:909–15. doi: 10.1001/archpsyc.58.10.909. [DOI] [PubMed] [Google Scholar]
  • 167.Schreiner AM, Dunn ME. Residual effects of cannabis use on neurocognitive performance after prolonged abstinence: a meta-analysis. Exp Clin Psychopharmacol. 2012;20:420–29. doi: 10.1037/a0029117. [DOI] [PubMed] [Google Scholar]
  • 168.Scott JC, Slomiak ST, Jones JD, Rosen AFG, Moore TM, Gur RC. Association of Cannabis with cognitive functioning in adolescents and young adults: a systematic review and meta-analysis. JAMA Psychiatry. 2018;75:585–95. doi: 10.1001/jamapsychiatry.2018.0335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Colizzi M, Bhattacharyya S. Does Cannabis composition matter? Differential effects of delta-9-tetrahydrocannabinol and Cannabidiol on human cognition. Curr Addict Rep. 2017;4:62–74. doi: 10.1007/s40429-017-0142-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Ciccone CD. Medical Marijuana: just the Beginning of a long, strange trip?. Phys Ther. 2017;97:239–48. [DOI] [PubMed] [Google Scholar]
  • 171.Curran HV, Freeman TP, Mokrysz C, Lewis DA, Morgan CJA, Parsons LH. Keep off the grass? Cannabis, cognition and addiction. Nat Rev Neurosci. 2016;17:293–306. doi: 10.1038/nrn.2016.28. [DOI] [PubMed] [Google Scholar]
  • 172.Karila L, Roux P, Rolland B, Benyamina A, Reynaud M, Aubin H-J, Lancon C. Acute and long-term effects of cannabis use: a review. Curr Pharm Des. 2014;20:4112–18. doi: 10.2174/13816128113199990620. [DOI] [PubMed] [Google Scholar]
  • 173.Cannabis Leung L. and its derivatives: review of medical use. J Am Board Fam Med JABFM. 2011;24:452–62. doi: 10.3122/jabfm.2011.04.100280. [DOI] [PubMed] [Google Scholar]
  • 174.Franz CA, Frishman WH. Marijuana use and Cardiovascular disease. Cardiol Rev. 2016;24:158–62. doi: 10.1097/CRD.0000000000000103. [DOI] [PubMed] [Google Scholar]
  • 175.Sidney S Cardiovascular consequences of marijuana use. J Clin Pharmacol. 2002;42:64S–70S. [DOI] [PubMed] [Google Scholar]
  • 176.Biehl JR, Burnham EL. Cannabis smoking in 2015: a concern for lung health? Chest. 2015;148:596–606. doi: 10.1378/chest.15-0447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Papatheodorou SI, Buettner H, Rice MB, Mittleman MA. Recent marijuana use and associations with exhaled nitric oxide and pulmonary function in adults in the United States. Chest. 2016;149:1428–35. doi: 10.1016/j.chest.2015.12.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Sagar KA, Gruber SA. Marijuana matters: reviewing the impact of marijuana on cognition, brain structure and function, & exploring policy implications and barriers to research. Int Rev Psychiatry Abingdon Engl. 2018;30:251–67. doi: 10.1080/09540261.2018.1460334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Gruber SA, Sagar KA, Dahlgren MK, Yao X, Levine SJ. Splendor in the Grass? A pilot study assessing the impact of medical Marijuana on executive function. Front Pharmacol. 2016;7:355. doi: 10.3389/fphar.2016.00323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Hyder AA, Wunderlich CA, Puvanachandra P, et al. The impact of traumatic brain injuries: a global perspective. NeuroRehabilitation. 2007;22:341–53. [PubMed] [Google Scholar]
  • 181.Sun J-H, Tan L, Yu J-T. Post-stroke cognitive impairment: epidemiology, mechanisms and management. Ann Transl Med. 2014;2:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Grundy RI. The therapeutic potential of the cannabinoids in neuroprotection. Expert Opin Investig Drugs. 2002;11:1365–74. doi: 10.1517/13543784.11.10.1365. [DOI] [PubMed] [Google Scholar]
  • 183.Latorre JGS, Schmidt EB. Cannabis, Cannabinoids, and cerebral metabolism: potential applications in stroke and disorders of the central nervous system. Curr Cardiol Rep. 2015;17:627. doi: 10.1007/s11886-015-0627-3. [DOI] [PubMed] [Google Scholar]
  • 184.Magid L, Heymann S, Elgali M, Millis SR, Scott C, Pearson C. Role of CB2 receptor in the recovery of mice after traumatic brain injury. J Neurotrauma. 2019;36:1836–46. doi: 10.1089/neu.2018.5873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Shohami E, Cohen-Yeshurun A, Magid L, Facciolo F, Rendina EA, Page C, Cazzola M, Orlandi A. Endocannabinoids and traumatic brain injury. Br J Pharmacol. 2011;163:1402–10. doi: 10.1111/j.1476-5381.2011.01339.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Hussain S, Sankar R. Pharmacologic treatment of intractable epilepsy in children: a syndrome-based approach. Semin Pediatr Neurol. 2011;18:171–78. doi: 10.1016/j.spen.2011.06.003. [DOI] [PubMed] [Google Scholar]
  • 187.Wirrell EC. Treatment of Dravet Syndrome. Can J Neurol Sci J Can Sci Neurol. 2016;43:S13–18. doi: 10.1017/cjn.2016.249. [DOI] [PubMed] [Google Scholar]
  • 188.Szaflarski JP, Bebin EM. Cannabis, cannabidiol, and epilepsy–from receptors to clinical response. Epilepsy Behav EB. 2014;41:277–82. doi: 10.1016/j.yebeh.2014.08.135. [DOI] [PubMed] [Google Scholar]
  • 189.Hussain SA, Zhou R, Jacobson C, Weng J, Cheng E, Lay J, Hung P, Lerner JT, Sankar R. Perceived efficacy of cannabidiol-enriched cannabis extracts for treatment of pediatric epilepsy: A potential role for infantile spasms and Lennox-Gastaut syndrome. Epilepsy Behav EB. 2015;47:138–41. doi: 10.1016/j.yebeh.2015.04.009. [DOI] [PubMed] [Google Scholar]
  • 190.Sirven JI. Cannabis, cannabidiol, and epilepsies: the truth is somewhere in the middle. Epilepsy Behav EB. 2014;41:270–71. doi: 10.1016/j.yebeh.2014.09.006. [DOI] [PubMed] [Google Scholar]
  • 191.Cilio MR, Thiele EA, Devinsky O. The case for assessing cannabidiol in epilepsy. Epilepsia. 2014;55:787–90. doi: 10.1111/epi.2014.55.issue-6. [DOI] [PubMed] [Google Scholar]
  • 192.Devinsky O, Marsh E, Friedman D, Thiele E, Laux L, Sullivan J, Miller I, Flamini R, Wilfong A, Filloux F, et al. Cannabidiol in patients with treatment-resistant epilepsy: an open-label interventional trial. Lancet Neurol. 2016;15:270–78. doi: 10.1016/S1474-4422(15)00379-8. [DOI] [PubMed] [Google Scholar]
  • 193.Thiele EA, Marsh ED, French JA, Mazurkiewicz-Beldzinska M, Benbadis SR, Joshi C, Lyons PD, Taylor A, Roberts C, Sommerville K, et al. Cannabidiol in patients with seizures associated with Lennox-Gastaut syndrome (GWPCARE4): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Lond Engl. 2018;391:1085–96. doi: 10.1016/S0140-6736(18)30136-3. [DOI] [PubMed] [Google Scholar]
  • 194.McCoy B, Wang L, Zak M, Al-Mehmadi S, Kabir N, Alhadid K, McDonald K, Zhang G, Sharma R, Whitney R, et al. A prospective open-label trial of a CBD/THC cannabis oil in dravet syndrome. Ann Clin Transl Neurol. 2018;5:1077–88. doi: 10.1002/acn3.2018.5.issue-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Devinsky O, Cross JH, Laux L, Marsh E, Miller I, Nabbout R, Scheffer IE, Thiele EA, Wright S. Trial of Cannabidiol for Drug-Resistant Seizures in the Dravet Syndrome. N Engl J Med. 2017;376:2011–20. doi: 10.1056/NEJMoa1611618. [DOI] [PubMed] [Google Scholar]
  • 196.Mitchell JT, Sweitzer MM, Tunno AM, Kollins SH, McClernon FJ, Lidzba K. “I use weed for my ADHD”: A qualitative analysis of online forum discussions on Cannabis use and ADHD. PLoS One. 2016;11:e0156614. doi: 10.1371/journal.pone.0156614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Ferner M Number of children seeking medical Marijuana soars in Colorado [Internet]. HuffPost. Available from: https://www.huffpost.com/entry/medical-marijuana-children_n_4768219.
  • 198.Hadland SE, Knight JR, Harris SK. Medical marijuana: review of the science and implications for developmental-behavioral pediatric practice. J Dev Behav Pediatr JDBP. 2015;36:115–23. doi: 10.1097/DBP.0000000000000129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Rasmussen J, Casey BJ, van Erp TGM, Tamm L, Epstein JN, Buss C, Bjork JM, Molina BSG, Velanova K, Mathalon DH, et al. ADHD and cannabis use in young adults examined using fMRI of a Go/NoGo task. Brain Imaging Behav. 2016;10:761–71. doi: 10.1007/s11682-015-9438-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Kelly C, Castellanos FX, Tomaselli O, Lisdahl K, Tamm L, Jernigan T, Newman E, Epstein JN, Molina BSG, Greenhill LL, et al. Distinct effects of childhood ADHD and cannabis use on brain functional architecture in young adults. NeuroImage Clin. 2017;13:188–200. doi: 10.1016/j.nicl.2016.09.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Charach A, Yeung E, Climans T, Lillie E. Childhood attention-deficit/hyperactivity disorder and future substance use disorders: comparative meta-analyses. J Am Acad Child Adolesc Psychiatry. 2011;50:9–21. doi: 10.1016/j.jaac.2010.09.019. [DOI] [PubMed] [Google Scholar]
  • 202.Klein RG, Mannuzza S, Olazagasti MAR, Craig M, Hallahan B, Murphy C, Johnston P, Spain D, Gillan N, Brammer M, et al. Clinical and functional outcome of childhood attention-deficit/hyperactivity disorder 33 years later. Arch Gen Psychiatry. 2012;69:1295–303. doi: 10.1001/archgenpsychiatry.2012.513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Lee SS, Humphreys KL, Flory K, Liu R, Glass K. Prospective association of childhood attention-deficit/hyperactivity disorder (ADHD) and substance use and abuse/dependence: a meta-analytic review. Clin Psychol Rev. 2011;31:328–41. doi: 10.1016/j.cpr.2011.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Molina BSG, Hinshaw SP, Eugene Arnold L, Swanson JM, Pelham WE, Hechtman L, Hoza B, Epstein JN, Wigal T, Abikoff HB, et al. Adolescent substance use in the multimodal treatment study of attention-deficit/hyperactivity disorder (ADHD) (MTA) as a function of childhood ADHD, random assignment to childhood treatments, and subsequent medication. J Am Acad Child Adolesc Psychiatry. 2013;52:250–63. doi: 10.1016/j.jaac.2012.12.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Zamberletti E, Gabaglio M, Parolaro D. The endocannabinoid system and autism spectrum disorders: insights from animal models. Int J Mol Sci 2017;18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Karhson DS, Krasinska KM, Dallaire JA, Libove RA, Phillips JM, Chien AS, Garner JP, Hardan AY, Parker KJ. Plasma anandamide concentrations are lower in children with autism spectrum disorder. Mol Autism. 2018;9:18. doi: 10.1186/s13229-018-0203-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Wei D, Lee D, Cox CD, Karsten CA, Peñagarikano O, Geschwind DH, Gall CM, Piomelli D. Endocannabinoid signaling mediates oxytocin-driven social reward. Proc Natl Acad Sci USA. 2015;112:14084–89. doi: 10.1073/pnas.1509795112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Gu B Cannabidiol provides viable treatment opportunity for multiple neurological pathologies of autism spectrum disorder. Glob. Drugs Ther. Internet]. 2017. [cited 2019 Jun 10];2 Available from: http://www.oatext.com/cannabidiol-provides-viable-treatment-opportunity-for-multiple-neurological-pathologies-of-autism-spectrum-disorder.php
  • 209.Kaplan JS, Stella N, Catterall WA, Westenbroek RE. Cannabidiol attenuates seizures and social deficits in a mouse model of Dravet syndrome. Proc Natl Acad Sci USA. 2017;114:11229–34. doi: 10.1073/pnas.1711351114. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES