Skip to main content
Bentham Open Access logoLink to Bentham Open Access
. 2014 Apr;20(13):2126–2137. doi: 10.2174/13816128113199990428

Effects of Cannabis on Impulsivity: A Systematic Review of Neuroimaging Findings

Johannes Wrege 1, André Schmidt 1, Anna Walter 1, Renata Smieskova 1, Kerstin Bendfeldt 2, Ernst-Wilhelm Radue 2, Undine E Lang 1, Stefan Borgwardt 1,2,*
PMCID: PMC4052819  PMID: 23829358

Abstract

We conducted a systematic review to assess the evidence for specific effects of cannabis on impulsivity, disinhibition and motor control. The review had a specific focus on neuroimaging findings associated with acute and chronic use of the drug and covers literature published up until May 2012. Seventeen studies were identified, of which 13 met the inclusion criteria; three studies investigated acute effects of cannabis (1 fMRI, 2 PET), while six studies investigated non-acute functional effects (4 fMRI, 2 PET), and four studies investigated structural alterations. Functional imaging studies of impulsivity studies suggest that prefrontal blood flow is lower in chronic cannabis users than in controls. Studies of acute administration of THC or marijuana report increased brain metabolism in several brain regions during impulsivity tasks. Structural imaging studies of cannabis users found differences in reduced prefrontal volumes and white matter integrity that might mediate the abnormal impulsivity and mood observed in marijuana users. To address the question whether impulsivity as a trait precedes cannabis consumption or whether cannabis aggravates impulsivity and discontinuation of usage more longitudinal study designs are warranted.

Keywords: Cannabis, marijuana, cannabinoids, impulsivity, inhibition, neuroimaging, magnetic resonance imaging, MRI, fMRI.

INTRODUCTION

Cannabis sativa is a widely used drug comprising a broad spectrum of usage ranging from recreational users to chronic addicts [1, 2]. Over the last years, accumulating evidence revealed that cannabis use leads to structural and functional brain abnormalities in cannabis users [3, 4]. For example, neuroimaging data has shown that cannabis use reduces grey and white matter volumes in cannabinoid-receptor rich areas, as well as changes functional activity in the prefrontal and anterior cingulate cortex [5]. Furthermore, pharmacological neuroimaging studies of healthy volunteers has shown brain functional mechanisms underlying the effects of cannabis [6-14]. Although research on the neurobiological effects of cannabis is increasing, how it affects the neuronal correlates underlying cognitive control remain still poorly understood, in particular the interlacement of cannabis use and impulsivity. In this review, we provide an overview about the effects of cannabis has on impulsivity, a key function in cognitive self-control and goal-directed behavior. Abnormal impulsivity as a compromised ability to exert control over drug urges or to inhibit compulsive drug-driven behavior has repeatedly been reported because of drug abuse. Thus, this systematic review incorporates studies addressing the effects of cannabis on the modulation of impulsive behavior by revisiting behavioral, as well as functional and structural neuroimaging findings.

Impulsive Behavior

Although impulsive behavior is a pre-existing personality trait that may promote the usage of drugs, consuming cannabis may result in behavioral changes including alterations of impulsivity [15]. According to Durana and Barnes [16], impulsivity is defined as “actions which are poorly conceived, prematurely expressed, unduly risky or inappropriate to the situation and that often result in undesirable consequences”. As such, it is part of normal behavior, but the multifaceted construct as well encompasses behavioral characteristics contributing to different psychopathological symptoms in a broad spectrum of neuropsychiatric disorders [17], and is a core deficit in substance abuse disorders [18]. Furthermore, impulsivity has been regarded as a lack of “executive control”, including deficits in higher-order cognitive functions such as inhibition, shifting and updating of information and behavior [19]. There is robust evidence that impulsive behavior reflects neurodevelopmental processes mediated in distinct brain networks (i.e. frontal cortex) contributing to its cognitive, clinical and behavioral aspects [20]. Behaviorally, impulsivity can be operationalized as a `predisposition toward rapid, unplanned reactions to internal and external stimuli without regard for the negative consequences of these reactions to themselves or others` [21]. Two domains are of relevance: the choice of a smaller, immediate reward over a larger, delayed reward [22] or the inability to inhibit behavior by changing the course of action or to stop a response once initiated [23].

Role of the Prefrontal Cortex in Neurocognitive Networks of Impulsivity and the Transition to Addiction

A key role has been ascribed to the prefrontal cortex (PFC) in its regulation of reward and its involvement in higher-order executive functions such as cognitive self-control (goal-directed behavior, response inhibition etc.), salience attribution and awareness. Goldstein and Volkow [24] reviewed neuroimaging data of distinct PFC regions and their roles in the neuropsychological mechanisms that underlie the relapsing cycle of addiction. Based on that data, with a focus on inhibitory control and emotion regulation, the iRISA-model of Goldstein and Volkow [24] distinguishes between dorsal PFC regions including the dorsolateral PFC, dorsal ACC, and the IFG, which are implicated in higher-order cognitive processes (“cold” processes), and ventral PFC regions subsuming the ventral OFC, ventromedial PFC, and rostroventral ACC, which are engaged during emotion-related “hot” processes. In the healthy state, automatic drug-related responses are suppressed by the input from dorsal PFC regions. During the state of withdrawal or craving, drug-related functions start to eclipse non-drug related functions, leading to a conflict situation with increasing drug-biased cognition and cue-induced craving. When the drug is reinstated, higher-order non-drug related cognitive functions are overrun by the input from PFC regions involved in drug-related `hot` processes. Thus, attention narrows to drug-related cues, impulsivity increases and basic emotions are unleashed resulting in automatic, stimulus-driven behavior.

Operationalization of Impulsivity

There are several tasks and subjective questionnaires to measure impulsivity. Here, we specifically focused on reviewing seven subdomains of impulsivity, namely response inhibition, motor control, interference/incongruity, attention, decision making/ gambling, cognitive flexibility and delay discounting. Table 1 gives an overview of the tasks used in the reviewed neuroimaging studies.

Table 1.

Impulsivity instruments used in the reviewed neuroimaging studies

Response inhibition:
Commission errors, reaction time, competing responses
Go-/No-Go task
Stop signal reaction time task (SSRTT)
(EFT)
Motor control accuracy, errors, and reaction time Critical tracking task
Self-paced finger-tapping task
Interference: incongruent/ congruent STROOP task
The multi-source interference task
Attention:
-divided
-sustained
Divided attention task,
Stop signal task
The multi-source interference task
SAT, WCST, MicroCogTM)
Self-paced counting task
Decision making and risk taking: Iowa gambling task
Cognitive flexibility:
-shifting
-perseveration
Wisconsin Cart sorting test
Delay discounting Barratt Impulsivity Scale - BIS-11

In addition, other studies’ designs often use psychometric impulsivity measures as a trait using self-reporting scales like the Barratt Impulsivity Scale (BIS-11)[25], the UPPS-P Impulsive Behavior Scale (IBS) [26], and the Kirby test of delayed discounting [27]. Psychometric measurements of impulsivity often fail to show clear correlations with objective methods and it has been suggested that self-report scales may reflect subjective commentaries on impulsive-like behavioral output [21].

METHODS

Selection Procedures and Search Strategy

We performed a PUBMED database search including all entries until the end of May 2012 on the following blocks of search terms: “Cannabis”, “Cannabinoid”, “THC”, “Marihuana” OR “Marijuana” AND “Impulsivity”, “Motor Control”, “Motor Inhibition” OR “Disinhibition”. We aimed to provide an overview on research of impulsivity and cannabis use, we arranged all included studies in tables. Thus, we differentiated neuroimaging and epidemiological studies in humans from behavioral studies in animals and humans, and genetic studies, again in animals and humans. In this review, we want to focus on disinhibition and task-related brain alterations of functional and structural neuroimaging data in cannabis users. We reviewed studies in English, German and Spanish. Two researchers were responsible for study selections: Johannes Wrege (JW) and Stefan Borgwardt (SB). Initially, selection was independent, then screening and full-text assessment were carried out via group discussion.

Selection Criteria and Recorded Variables

We hand-searched all publications in order to find studies investigating the effects of cannabis use on impulsivity or studies trying to elucidate the influence of the endocannabinoid system on inhibition and motor control. We also searched the references of all included manuscripts for further relevant publications. In order to be included, studies had to have a parallel, crossover or case-control design with an appropriate control group of healthy controls, placebo or baseline comparisons and an original publication in a peer-reviewed journal. To address the principal aim of this review, studies had to include an impulsivity measure or task in at least one of the above-mentioned domains. The outcome measures had to be valid neuropsychological tests (see operationalization of impulsivity section). Studies with subjects of co-use had to have a “pure” group of “cannabis users” only. Furthermore, studies had to address other potential substance use, potential history of neurological or psychiatric problems, or had to report length of abstinence from Cannabis before testing. Exclusion criteria were any psychiatric or neurological disorder with the exception of substance use disorders when providing a Cannabis group only.

The final selection of systematically reviewed studies consists of neuroimaging studies in humans. We reviewed 13 neuroimaging studies between 2003 and 2012 on the effects of cannabis on impulsivity. Of these studies, only three tested acute effects and 11 non-acute effects, with either structural or functional imaging methods. We chose to present the reviewed studies in informative tables in order to provide a comprehensible interpretation of the main findings and conclusions drawn.

In these tables, we differentiated between chronic and recreational users, abstinent and non-users, acute and non-acute effects, functional and structural studies, the type of comparison, type of administration: delta-9-THC, Cannabis, the route of drug investigated, the study design and more, see Table 2 for further information.

Table 2.

  Author Journal Study aim POPULATION
C / Usertype
SPECIFICATION OF STUDIE DESIGN
Parallel, crossover, placebo / abstinence time
Tests/ measurements of Impulsivity and Motor Control
acute effects Borgwardt et al. 2008 Biological Psychiatry fMRI, acute effects of THC on motor inhibition in healthy volunteers - 15 healthy male volunteers (<15 times use in lifetime) double-blind, pseudo-random., placebo-contr. rep. meas. within-sub. 1 month Go/No-Go-task
Weinstein et al. 2007 Psychopharmacology 18FDG-PET, acute 17mg THC on motor control in regular marijuana users - 12 regular users (1 joint per day at least 5 years) Double-blind, placebo-controlled, cross-over Requested only, the night before Psycho-motor(virtual maze) task; WCST, IGT, estimates of time and distance of a car
O'Leary et al. 2003 NeuroReport H215O-PET, acute effects of THC, perfusion and internal timing in heavy compared to moderate use - 12 heavy(daily 1.8 joints since 5.4y) /
12 moderate (once a week since 3.9y)
Double-blind placebo-controlled counterbalanced design - Self-paced counting task
Self-paced finger-tapping task
non-acute effects functional Gruber et al. 2012 Neuroscience Letters fMRI, inhibiting impulsive behaviors in early (<16y) vs. late onset MJs 16
<15x
23heavy (2500+joint)MJ (n=9 <16y/ n=15 +16y) /
16 non-smoking HC
Cross-sectional 12h MSIT
Hestor et al. 2009 Neuropsychopharmacology fMRI, inhibitory control and error awareness in chronic MJ 16 16 chronic users (500+ joints, 5-7x/week for 2y) Cross-sectional - Go/No-Go-task; EAT
Tapert et al.
 2007
Psychopharmacology fMRI, go/no-go task in adolescent marijuana users after 28 days of abstinence. 17 16 recreational (+60 lifetime) Cross-sectional 28 days Go/No-Go-task
Bolla et al. 2005 NeuroImage H215O-PET, 25-day abstinent MJ users dose-related alterations in the Iowa Gambling (IGT) 11 11 heavy chronic users
(4/w for 2y)
(8-35j/w vs. 53-84j/w)
Cross-sectional 28 days IGT, resting state, sensorimotor Control-task
Gruber et al.
 2005
Cognitive brain research fMRI, DTI, frontal dysfunctions & structural changes in heavy cannabis use in modified Stroop task 9 9 chronic users
(4000+ joints)
Cross-sectional Urine+/- for THC, but not others STROOP-task
Eldreth et al. 2004 NeuroImage PET 15O, modified Stroop task, 25-day abstinent, heavy MJ users, executive cognitive functioning 11 11 chronic users
(4/w for 2y)
Cross-sectional 23d MJ
3d Controls
Urine sample time range
Rest-R (eyes fixated on a target); Active Task-A (Conflict condition STROOP);Control Task-C (sensorimotor No Conflict condition)
structural Gruber et al.
 2011
Experimental and Clinical Psychopharmacollogy DTI, impulsivity measures in chronic heavy MJ smokers, white matter microstructure 15 15 chronic users Cross-sectional - BIS-11
    Siveri et al.
 2011
Psychiatry Research MRSI in MJ-dependence 11 15 chronic users
MJ 1.use=15.7±2.2y for 5.5±2.6y; 5.6±1.7/week
Cross-sectional Non
Urine+/-
BIS-11, +3 affect/mood scales
Churchwell et al.
 2010
Frontiers in Psychology MRI, functional integrity of moPFC, reward perception, substance abuse, and dependence. 18 18 recreational users Cross-sectional Continued use until study visit BIS-11
Hermann et al. 2007 Biological Psychiatry 1H-MRS, Cannabinoids neurotoxic and neuroprotective properties inconsistent alterations neuropsychological deficits, neuropsychological testing 13 14 chronic recreational users Cross-sectional Non
Urine+/-
WCST, TMT, D2
Gruber et al. 2005 Cognitive brain research DTI, heavy cannabis smokers performing a modification of the classic Stroop task. 9 9 chronic users
(4000+ joints)
Cross-sectional Urine+/- for THC, but not others STROOP

BIS-11: Barratt Impulsivity Scale-11; C: Controls; DTI: Diffusion tensor imaging technique; EAT: Error Awareness Task; IGT: Iowa Gambling task; MJ: Marijuana user; MRSI: proton magnetic resonance spectroscopic imaging; MSIT: Multi-Source Interference Task; PET: positron emission tomographic imaging; STROOP: Stroop Colour Word Test; WCST: Wisconsin card sorting test, (Heaton, 1999);

We included three studies of acute administration of cannabis or delta-9-THC on different types of subjects and tasks (Go-/No-Go task/stop-signal task, self-paced counting task/ virtual psycho-motor task, attention task). Two studies assessed cannabis users (occasional vs. heavy users, regular users - 1 joint per day) and only one study assessed healthy non-using volunteers [14]. This is the only study which assessed the different effects of the two main psychotropic ingredients of cannabis, cannabidiol in comparison to tetrahydrocannabinol. Three out of six reviewed non-acute functional studies applied an interference task, two a Go/No-Go task and one study a decision making gambling task. The five reviewed structural study designs used the BIS-11 (three studies) impulsivity questionnaire, and the stroop and Wisconsin Card sorting test as a measure of impulsivity and executive control.

RESULTS

We found 774 entries listed in Pubmed until May 2012 with the above depicted search terms. Neuroimaging studies on impulsivity and cannabis are limited, whereas, historically reasoned, there is a large number of behavioral studies on cannabis use with systematic reviews on cognitive functions, but not specifically on impulsivity or inhibitory control.

Identified Studies

Seventeen neurophysiological and neuroimaging studies published between 2003 and 2012 met the inclusion criteria and were reviewed. One study was excluded due to a missing control group [28], one due to other study aim [29], and two due to different neuroimaging techniques: event related potential EEG [30], and transcranial magnetic stimulation combined with electromyography [31]. Among the remaining 13 neuroimaging studies, three studies investigated acute effects of either healthy volunteers and occasional or heavy cannabis users (1 fMRI, 2 PET). 10 non-acute studies were included with different abstinence periods ranging from 12 hours up to 28 days, while six studies investigated non-acute functional impacts (four fMRI, two PET), and four studies investigated non-acute structural alterations of cannabis use. One study provided both functional and structural data [41] which sums up the structural data to five studies. We sorted the final number of reviewed studies in acute functional, non-acute functional and non-acute structural studies as you can see in Table 2. It lists all included studies and contains an overview for better orientation. The following flow chart (in Fig. 1) illustrates the selection process.

Fig. (1).

Fig. (1)

Behavioral Findings in Neuroimaging Studies

Two out of the three reviewed studies of acute administration of either cannabis or delta-9-THC reported an impact on the behavioral level. Weinstein and colleagues [32] found significant more virtual wall hits in a reality maze after smoking 17mg delta-9-THC cigarettes in regular users, and subjects in the study of O`Leary et al. [33] showed increased rates of self-paced counting after smoking marijuana in both recreational and heavy users.

All included studies of non-acute functional designs found no significant differences in inhibitory task performance. However, despite comparable inhibitory performances between users and non-users, marijuana users tended to have faster reaction times and higher rates on commission errors [34]. This was even more pronounced in early onset than in late onset of cannabis use, albeit not statistically underpinned. Hester et al. [35] found that earlier onset of cannabis use was associated with poorer inhibitory control, but this association only approached significance. Their cannabis-using subjects showed significantly decreased error awareness. One study used the decision-making Iowa Gambling task (IGT) and found abnormal performance in marijuana-smoking subjects who had chosen more risky cards with higher reward opportunities [36]. Heavy unlike moderate users showed significant performance abnormalities and developed no learning behavior over two trials, which indicates a missing adaptive shift in decision-making and the ability to balance reward and punishment. Studies measuring impulsivity with self-rating questionnaires such as BIS-11 found significantly higher BIS-11 scores in marijuana using subjects. Further studies revealed higher total scores [37, 38], higher cognitive and motor impulsivity subscores [38], and decreased future orientation indexed in the non-planning subscale of the BIS-11 [39].

Neuroimaging Findings

Table 3 depicts the details of higher or lower brain activation in terms of bold response contrasts, greater or less regional volume or modified regional cerebral blood flow after the administration of cannabis or within the cannabis-user group. In general, significant PFC activation during inhibitory control and cognitive control of impulsivity were found in the ACC, the left and right DLPFC, the inferior and medial PFC and the OFC. In addition, significant activation patterns were observed in parietal, temporal, hippocampal, occipital or cerebellar regions depending on the task used.

Table 3.

graphic file with name CPD-20-2126_T3.jpg

Acute Effects of Cannabis on Functional Brain Activity in Users and Healthy Volunteers

Healthy volunteers administered delta-9-THC acutely showed a decreased bold signal (fMRI) in the ACC and in the inferior frontal gyrus during No-Go conditions [14]. O’Leary et al. [33] found by using 15O-PET that cannabis resulted in increased rCBF in several regions including the anterior cingulate, mesial and orbital frontal lobes, insula, temporal poles, and cerebellum compared to placebo. These subjects of occasional vs. regular users (daily usage) had to attend to a finger tapping and self-paced counting task and revealed significant group differences. After smoking marijuana, occasional user showed decreased rCBF in cerebellar vermis and the thalamus, whereas chronic user showed less frontal lobe activation. This corresponds to the results of Borgwardt et al. [14] with decreased bold signaling under No-Go conditions. Nevertheless, a greater rCBF increase from pre- to post-smoking in the chronic compared to occasional user group has been found in thalamus and cerebellum. Regular cannabis users [40] underwent an 18FDG-PET scan analysis while applying a virtual reality maze task after the administration of 17mg delta-9-THC or placebo. More virtual wall hits under delta-9-THC have been accompanied by an increased brain metabolism during task performance in the middle and medial frontal cortices and anterior cingulate, while a reduced metabolism was found in the occipital lobes.

Non-acute Effects of Regular Cannabis use on Functional Brain Activity

Five (out of six) functional neuroimaging studies applied an inhibition task and revealed different bold responses during the No-Go conditions or during suppression of interfering information in cannabis users. During the task, marijuana users showed significantly higher ACC activity compared to healthy controls [40]. Furthermore, an earlier onset of cannabis use was associated with more commission errors compared to late onset users concomitant with more focal clusters. Gruber et al. [41] further revealed reduced activity during the Stroop task in focal areas of the ACC, but more activation in the middle cingulate and a more diffuse and bilateral pattern in the dorso-lateral PFC during interference condition in the marijuana-using group, while healthy controls showed only right dorso-lateral PFC activation during interference.

Hestor et al. [35] showed significant stop-related activity in right prefrontal, parietal and anterior cingulate regions. In the group comparison, cannabis users showed significantly greater activations in the right inferior parietal lobe, right putamen, and right middle cingulate gyrus. They combined the Stroop and Go/No-Go task to compare the bold signals of aware and unaware inhibition errors and found decreased activity in the ACC and right insula when errors were unaware. Pearson`s correlation analysis revealed that higher levels of cannabis use were associated with a lower bold signal in the right ACC and right insula in the marijuana group.

Tapert et al. [42] revealed that the cannabis-related bold signal increased during No-Go conditions in the right dorso-lateral PFC, bilaterally in middle frontal, inferior and superior parietal lobes and in the right occipital gyrus after 28 days of abstinence, indicating a greater brain effort while having same behavioral performance. Results remained significant after controlling for lifetime usage and alcohol use. Duration of regular marijuana use was negatively related to the activity in the right anterior superior frontal gyrus (BA 10), right superior middle frontal gyrus (BA 6), and left anterior superior frontal gyrus (BA 10) in response to No-Go conditions. Similarly, an early onset of regular marijuana use and more lifetime marijuana use episodes were related to less inhibitory responses in the right anterior superior frontal gyrus (BA 10). The number of marijuana hits per month was also negatively related to brain responses in the right anterior superior frontal gyrus (BA 10), right superior middle frontal gyrus (BA 6), left anterior superior frontal gyrus (BA 10), and left posterior parietal cortex.

Eldreth and colleagues [43] applied a modified stroop task during a PET scan after 25 days abstinence and revealed decreased activation in the left perigenual ACC, left lateral PFC and hippocampus bilaterally.

Bolla et al. [44] found decreased activation in the right lateral OFC, right dorso-lateral PFC and left cerebellum when cannabis users had to make decisions in a gambling task after 25 days of abstinence. A second-step analysis with a comparison between heavy and moderate use revealed decreased activity in the left medial OFC and an increase in left cerebellum in the heavy-user group.

Non-acute Effects of Regular Cannabis use on Brain Structures

Gruber et al. [37] found reduced fractional anisotropy (FA) of left frontal lobe in chronic heavy users and positive correlations with BIS-11 total and motor sub-scores, together with reduced FA and enhanced diffusivity in right genu of corpus callosum. Left frontal and right callosal alterations correlated with age of onset and duration of cannabis use. Without significant structural group difference, there was a correlation between right frontal diffusion tensor imaging (DTI)measures and BIS-11 total and attention sub-score. A reduced volume in medio-orbital PFC has been shown by Churchwell et al. [39], which was correlated with age of first usage. These occasional users scored more on non-planning subscale of the BIS-11. Two studies did not find regional specific structural alterations, but a different slope of metabolism of white matter fiber tracts and a trend towards an increase of non-parallel hence therefore non-directional diffusivity which is a sign of reduced integrity in frontal brain areas in dependent heavy marijuana using subjects [38, 41]. Two structural studies found positive correlations between age of onset of Marijuana use and frontal brain areas: [37] [39].

DISCUSSION

The aim of this systematic review was to assort the recent literature addressing the effect of cannabis use on impulsivity, disinhibition, and motor control. To the best of our knowledge, this is the first systematic review specifically disentangling the effect of cannabis on impulsivity or disinhibition.

Because impulsivity is a multifaceted construct (cf. [45]), comparisons between different study designs have to address the different operationalization used in that construct. Two facets of impulsivity often predominate research on impulsivity in the context of drug abuse [46], namely impulsive choice and impaired inhibition (see also [47]). In the current review, we included studies on cannabis and impulsivity measuring the following domains: response inhibition, motor control, interference/suppression of information, and executive functions of divided/sustained attention and decision making/risk taking. Neurophysiological definitions of impulsivity share all these aspects (see introduction). The focus of this review was to assess structural and functional imaging studies examining the effect of cannabis on impulsivity. We analyzed a broad cross-section of studies which each applied at least one impulsivity measure. These included acute and non-acute designs, different abstention periods and different consumption intensities.

On the behavioral level, the included studies did not find performance differences on inhibition in cannabis users, but trends towards faster reaction times and higher rates on commission errors were observed [34]. In contrast, behavioral studies of inhibitory and motor control found deficits of inhibitory control in cannabis users under acute exposure [48] [49, 50] [32, 51, 52] 53, and in regular users after abstention of use [36, 54] 55. Duration, time of onset and total lifetime amount of cannabis use, as well as hits per month have a detrimental impact on executive functions [56] [57] [58]. Earlier onset, longer duration, and heavier usage have been associated with altered structural frontal integrity and decreased activations in users compared to non-users in the superior and middle frontal gyrus [34, 37]. Behavioral studies showed impulsive decision-making in cannabis users under acute [32, 50, 59] and non-acute conditions [54] [60], while others did not find significant differences [61] [62] [53]. In recreational users a positive correlation between reduced volume in the medial prefrontal cortex and age of first marijuana use was found [39] while chronic users reported higher scores on the “non-planning” but lesser scores on the “motor impulsivity” subscale of the BIS-11 [38]. Put together, mere behavioral studies show detrimental effects of cannabis consumption on different domains of impulsivity even after prolonged abstention. While the behavioral data of neuroimaging studies show only trends, there are correlations between severity of consumption and structural and functional alterations in these studies.

Beyond behavioral findings, we also reviewed neuroimaging data with a focus on alterations in dorsal and ventral PFC regions including the dorsolateral (dl-), ventrolateral (vl-), ventromedial (vm-)PFC, subgenial, dorsal, or rostral ACC, inferior frontal gyrus (IFG), and orbital frontal cortex (OFC), because these brain regions have been proposed to be critically implicated in drug addiction [24].

PET studies of acute administration of cannabis or delta9-THC found pre- and post-treatment increases in rCBF and brain metabolism in the PFC (mesial, orbital, middle and medial frontal cortices) and ACC during task performance (O’Leary et al.,2003, Weinstein et. al., 2007), indicating an increased brain effort to perform equally (see also [63]). The only fMRI study we reviewed here found significant bold signal decreases after delta-9-THC and CBD administration among healthy non-smoking volunteers in the ACC and in the inferior frontal gyrus during response inhibition [14]. These functional brain imaging data suggest that delta-9-THC acutely attenuates the engagement of brain regions that mediate response inhibition. Independent from pre- to post-treatment increases in rCBF, between-group comparison showed significant less frontal activations in chronic versus occasional users [33]. These results support the iRISA model proposed by Goldstein and Volkow [24] models by showing that acute cannabis exposure reduced frontal activations in chronic users and that delta9-THC has a direct diminishing impact on brain areas relevant for executive control.

With respect to non-acute studies of cannabis administration in two fMRI studies, chronic and recreational cannabis users showed greater activation and a more diffuse and bilateral activation pattern of the dlPFC and middle frontal gyrus during inhibition compared to non-users [41, 42]. More diffuse and bilateral activations in users compared to more focal and lateralized cluster in non-using controls may reflect a compensatory brain effort mechanism [42] to achieve similar inhibitory control.

Attentional functions involved in error detection, interference and stop signaling are mediated mainly by the ACC. Reduced ACC activity has been linked with altered metabolisms in cingulate cortex (e.g. [79, 80]). There is evidence for reduced focal ACC activity during task performance in chronic cannabis users [35, 41, 43]. Chronic users also show a more diffuse activation pattern in the cingulum among different research groups with greater activations in the middle compared to the anterior cingulate cortex, and together with activations in parietal regions [34, 35, 41, 42]. This favors the hypothesis of additional recruitment of parietal cortices to maintain attentional competence in order to equally perform during task. Garavan et al. [79] proposed two interacting cortical systems of response inhibition based on fMRI and EEG data depending on high- or low-absent-mindedness: recruitment of right prefrontal and parietal regions on the one hand, and activation of cingulate regions on the other hand. Hence, different cingulum activations and additional parietal activation in cannabis users would represent difficulties or inefficacies to sustain sufficient attention when inhibiting responses.

Abnormal monitoring abilities may furthermore affect decision-making [34, 35, 41, 42] and this corresponds with reduced activations in right dlPFC and right lateral OFC [44]. Cannabis users also show diminished choice optimization and decreased functional activation of orbital prefrontal regions during performance of the IGT [81]. Regions of interest in fMRI research of the IGT are the OFC and dlPFC [82, 83]. The OFC is involved in reward attribution and altered in addiction [84, 85]. Poor decision-making is a hallmark of addiction, and the IGT can discriminate controls from patients also with ventral medial lesions [86, 87], substance-dependence, or pathological gamblers [44, 88, 89].

Frontal correlates of response inhibition are linked to the pre-supplementary motor cortex to simply suppress unwanted movements, while the dlPFC needs to be recruited during tasks involving higher working memory load [64, 65]. Inhibition-related functions have been found to be significantly heritable [66-68] [69]. It has been shown, by using a variety of imaging modalities, that the prefrontal cortex (most prominently the inferior prefrontal cortex and the supplementary motor area (SMA), as well as subcortical striato-thalamic projection targets), represents a neural correlate of response inhibition [70-78]. The neuroimaging findings reviewed here indicate both reduced task-specific and predefined region-specific (region of interests - ROI) activations in PFC, OFC and ACC in acute as well as non-acute studies, and greater activations in a more diffuse, bilateral pattern together with supplementary parietal cortex recruitments. On the behavioral level, neuroimaging data showed no significant differences, but tendencies in impairments towards task complexities as well as correlations with earlier onset and heavier consumption. Thus, cannabis under acute and non-acute condition attenuates the focal engagement of inhibitory networks, namely the right dorsolateral and inferior as well as the orbital prefrontal cortex and ACC. To perform equally as a compensatory mechanism, cannabis leads to bilateral and broader, more diffuse activations depending on the specific task. To maintain attentional competence, greater activation occurs in the middle, while a broader, more diffuse pattern of activity is found within the anterior cingulum. Supplementary parietal activations have also been found. When cognitive load in the task is higher, chronic users decompensate and have less PFC activation (hence prefrontal control) than occasional users. Impulsive decision-making is associated with reductions in dlPFC and OFC.

Out of the five structural imaging studies one found a reduction in the medial prefrontal cortex in recreational users [39]. Recently, findings from structural neuroimaging studies have found positive, negative or even inverse results of persisting alterations on brain morphology after long-term marijuana use that underlies deficits in attention, learning, memory, executive functions and emotional processes [5, 90, 91], for review see [92]. Abnormalities in neuropsychological testing and fMRI activation are consistent with imaging studies demonstrating morphological changes in cannabis users [5]. Some reports have described reduced grey matter in limbic areas [92, 93] and abnormal gyrification [94]. Recent reviews of structural neuroimaging studies revealed no significant differences on global measures of brain volume following cannabis use [5]. Region-specific brain volume alterations have been found in different areas, most consistently in hippocampal and parahippocampal regions, but were not consistent across studies [92, 95]. Inconsistencies might arrive from different imaging modalities, as well as from different analysis approaches such as region-of-interest (ROI) or whole brain voxel-based morphometry.

DTI of white matter microstructures has identified reduced fractional anisotropy (FA) in cannabis users [37, 41]. In accordance, increases of axonal diffusivity have also been found in fronto-temporal brain connections [96]. Furthermore Arnone et al. [97, 98] found a marijuana-related damage in the corpus callosum. Chronic marijuana use may result in reduced FA and increased diffusivity, which may be associated with increased impulsivity, and ultimately contribute to the initiation of marijuana use or the inability to discontinue use [37].

Morphological changes of neuronal structures that are rich in cannabinoid receptors have been reported most consistently in the hippocampus [99] [100, 101] and the cerebellum [102, 103]). However, Zalesky et al. [104] found no evidence for brain-wide cannabis-induced shortening of axonal fibers except in the right fimbria of the hippocampus (fornix), splenium of the corpus callosum and commissural fibers. Another study used (1H-MRS-)markers to investigate neurogenesis, synaptogenesis (NAA), and synaptic plasticity (Cho) in recreational cannabis users and found reduced neuronal and axonal integrity in the dlPFC and ACC. Thus, there is growing evidence of structural disintegration after prolonged cannabis exposure not only in whole-brain analyses, but also specifically in brain regions that are crucial for impulse control [29]. Whether structural alterations remain persistent after prolonged discontinuation of cannabis use is still in debate. There are hazardous white matter connectivity impacts when the developing brain is exposed to long-term cannabis usage, but these alterations could be reversible after abstinence or functional adaptation [104].

In general, reviews show either the effects of cannabis on brain structures in psychiatric patients, individuals at a high risk of developing psychosis, or the interplay between psychosis and co-morbidities. These data may be distinct from non-psychiatric samples [5,91,95,105]. Nevertheless, cannabis use in clinical subjects is associated with decreases in some brain regions, particularly those rich in CB1–receptors such as the cingulum, the dlPFC, the cerebellum, and striatum. According to the iRISA model, the cingulum, PFC and striatum are crucial regions of the inhibition network required to monitor, suppress and countermand hot drives and to instill cold, volitional control. Chang and Chronicle [106] reviewed the chronic effects in cannabis users and found equivocal evidence that it might result in structural brain changes, blood-oxygenation-level-dependent-fMRI studies consistently show alterations, or neuroadaptation, in the activation of brain networks responsible for higher cognitive functions, but whether changes are reversible with abstinence was unclear. Likewise Chang and Chronicle [106], Batalla and colleagues [107] reviewed neuroimaging data in chronic adolescent and adult cannabis users with no other psychiatric disorder. Probands had to have consumed cannabis at least several times per week for a minimum of two years. Morphological alterations were found in medial, temporal and frontal cortices, as well as in the cerebellum. They also found different patterns of resting in global and brain activity during different task performances, which may also have indicated compensatory effects in chronic exposure. In contrast to that review, we focused on the impact of cannabis use on impulsivity measures and included acute and non-acute studies of recreational drug users and drug-naïve individuals. Comparisons of studies are often limited because of the differing definitions of cannabis exposure. Only a few studies provide absolute numbers of consumption in order to use cannabis abuse as a continuous variable. This is important when taking into account the possibility of a threshold effect of cannabis on brain morphology. Furthermore, we reviewed the literature with respect to impulsivity, using the iRISA model as a framework, to provide a conclusive overview of neuroimaging data regarding the impact of cannabis on brain structures and the functionality of habitual control networks in humans.

Impulsivity is a proposed endophenotype for substance dependence as well as a possible consequence of prolonged drug use [108]. Drugs of addiction and subconscious craving are likely to increase impulsiveness due to the loss of frontal cortical inhibition of impulses and increased limbic drive (see [24] [109]). Neural substrates of impulsivity encompass circuitry involving both cortical (top-down volitional control) and subcortical mechanisms, particularly within the basal ganglia, and have mainly been studied on response inhibition in humans and experimental animals. Although there are insights into the interplay of compulsive stimulant taking and impulsivity, this remains unclear in the case of cannabis. However, there are behavioral and epidemiological findings, which emphasize such a relationship, as well as the fact that cannabis use may lead to a loss of attention, reflection and inhibitory control. Associated cerebral activations consist of “right lateralized fronto-parietal” attentional or response selection mechanisms, as well as “midline performance monitoring processes” (ACC). To stop already pre-processed activation inferior frontal region are involved to countermand responses (see [80]). Thus, giving the broad epidemiological “burden of society” cannabis leads to dose-related impairments of psychomotor performance with implications for car driving, and personal health risk [110].

Limitations of this review include the analysis of different user types (heavy vs. recreational), small study groups, and a failing to control for co-use between studies. Whole brain search strategies often fail to show significant results, but localized differences may be found in connectivity analyses in future structural and functional neuroimaging studies.

ACKNOWLEDGEMENTS

We would like to thank Doris Blaser for her help preparing this manuscript.

SUMMARY AND CONCLUSION

This systematic review provides evidence that acute administration of cannabis increases regional CBF and brain metabolism, which might indicate an increase in brain effort during task performance.

Neuroimaging data provide evidence of detrimental effects of cannabis on inhibitory control. There is a threshold effect with alterations being found preferably in heavier and more regular users. When subjects showed an early onset of use, especially during brain maturation (before age of 16), structural brain alterations seem to be stronger. Research on the acute impacts of cannabis is limited and should be further engaged. As yet, there is no longitudinal study addressing the question whether impaired inhibition is due to a preceding trait of impulsivity and therefore leads to cannabis use, or whether it represents a result of use, and is therefore intertwined with difficulties to discontinue usage (see [111]). There is evidence of region-specific reduced volume and white matter integrity of the PFC, which leads to increased diffusivity and different metabolism in CB1-receptor-rich brain areas that might mediate the abnormal impulsivity and mood observed in marijuana users.

CONFLICT OF INTEREST

The authors confirm that this article content has no conflicts of interest.

ABBREVIATIONS

ACC

 =  Anterior cingulate cortex

BIS-11

 =  Barratt Impulsivity Scale-11

C

 =  Controls

CBD

 =  cannabidiol

DTI

 =  Diffusion tensor imaging technique

EAT

 =  Error Awareness Task

IGT

 =  Iowa Gambling task

FA

 =  fractional anisotropy e.g.

IFG

 =  inferior frontal cortex

DLPFC

 =  Dorso-lateral prefrontal cortex

MJ

 =  Marijuana user

L

 =  left

MRSI

 =  Proton magnetic resonance spectroscopic imaging

MSIT

 =  Multi-Source Interference Task

OFC

 =  Orbital frontal cortex

PET

 =  Positron emission

PFC

 =  Prefrontal cortex; tomographic imaging

rCBF

 =  Regional cerebral blood flow

Stroop

 =  Stroop Colour Word Test

R

 =  right

THC

 =  delta-9-tetrahydrocannabinol

WCST

 =  Wisconsin card sorting test

REFERENCES

  • 1.Carmichael ST, Price JL. Sensory and premotor connections of the orbital and medial prefrontal cortex of macaque monkeys. J Comp Neurol. 1995;363(4):642–64. doi: 10.1002/cne.903630409. [DOI] [PubMed] [Google Scholar]
  • 2.Zuurman L, Passier PC, de Kam M, Kleijn HJ, Cohen AF, van Gerven JM. Pharmacodynamic and pharmacokinetic effects of the intravenously administered CB1 receptor agonist Org 28611 in healthy male volunteers. J Psychopharmacol. 2009;23(6):633–44. doi: 10.1177/0269881108091551. [DOI] [PubMed] [Google Scholar]
  • 3.Indlekofer F, Piechatzek M, Daamen M , et al. Reduced memory and attention performance in a population-based sample of young adults with a moderate lifetime use of cannabis. ecstasy and alcohol. J Psychopharmacol. 2009;23(5):495–509. doi: 10.1177/0269881108091076. [DOI] [PubMed] [Google Scholar]
  • 4.Solowij N, Michie PT. Cannabis and cognitive dysfunction: parallels with endophenotypes of schizophrenia?. J Psychiatry Neurosci. 2007;32(1):30–52. [PMC free article] [PubMed] [Google Scholar]
  • 5.Martin-Santos R, Fagundo AB, Crippa JA , et al. Neuroimaging in cannabis use: a systematic review of the literature. Psychol Med . 2010 Mar;40(3):383–98. doi: 10.1017/S0033291709990729. [DOI] [PubMed] [Google Scholar]
  • 6.Atakan Z, Bhattacharyya S, Allen P , et al. Cannabis affects people differently: inter-subject variation in the psychotogenic effects of Delta9-tetrahydrocannabinol: a functional magnetic resonance imaging study with healthy volunteers. Psychol Med. 2012:1–13. doi: 10.1017/S0033291712001924. [DOI] [PubMed] [Google Scholar]
  • 7.Martin-Santos R, Crippa JA, Batalla A , et al. Acute effects of a single. oral dose of d9-tetrahydrocannabinol (THC) and cannabidiol (CBD) administration in healthy volunteers. Curr Pharm Des. 2012;18(32):4966–79. doi: 10.2174/138161212802884780. [DOI] [PubMed] [Google Scholar]
  • 8.Bhattacharyya S, Crippa JA, Allen P , et al. Induction of psychosis by Delta9-tetrahydrocannabinol reflects modulation of prefrontal and striatal function during attentional salience processing. Arch Gen Psychiatry. 2012;69(1):27–36. doi: 10.1001/archgenpsychiatry.2011.161. [DOI] [PubMed] [Google Scholar]
  • 9.Winton-Brown TT, Allen P, Bhattacharyya S , et al. Modulation of auditory and visual processing by delta-9-tetrahydrocannabinol and cannabidiol: an FMRI study. Neuropsychopharmacology. 2011;36(7):1340–8. doi: 10.1038/npp.2011.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bhattacharyya S, Morrison PD, Fusar-Poli P , et al. Opposite effects of delta-9-tetrahydrocannabinol and cannabidiol on human brain function and psychopathology. Neuropsychopharmacology. 2010;35(3):764–74. doi: 10.1038/npp.2009.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Fusar-Poli P, Allen P, Bhattacharyya S , et al. Modulation of effective connectivity during emotional processing by Delta 9-tetrahydrocannabinol and cannabidiol. Int J Neuropsychopharmacol. 2010;13(4):421–32. doi: 10.1017/S1461145709990617. [DOI] [PubMed] [Google Scholar]
  • 12.Bhattacharyya S, Fusar-Poli P, Borgwardt S , et al. Modulation of mediotemporal and ventrostriatal function in humans by Delta9-tetrahydrocannabinol: a neural basis for the effects of Cannabis sativa on learning and psychosis. Arch Gen Psychiatry. 2009;66(4):442–51. doi: 10.1001/archgenpsychiatry.2009.17. [DOI] [PubMed] [Google Scholar]
  • 13.Fusar-Poli P, Crippa JA, Bhattacharyya S , et al. Distinct effects of {delta}9-tetrahydrocannabinol and cannabidiol on neural activation during emotional processing. Arch Gen Psychiatry. 2009;66(1):95–105. doi: 10.1001/archgenpsychiatry.2008.519. [DOI] [PubMed] [Google Scholar]
  • 14.Borgwardt SJ, Allen P, Bhattacharyya S , et al. Neural basis of Delta-9-tetrahydrocannabinol and cannabidiol: effects during response inhibition. Biol Psychiatry. 2008;64(11):966–73. doi: 10.1016/j.biopsych.2008.05.011. [DOI] [PubMed] [Google Scholar]
  • 15.Dalley JW, Everitt BJ, Robbins TW. Impulsivity. compulsiity.and top-down cognitive control. Neuron . 2011;69(4):680–94. doi: 10.1016/j.neuron.2011.01.020. [DOI] [PubMed] [Google Scholar]
  • 16.Durana J, Barnes P, McCown W, Johnson J, Shure M, editors. Washington: American Psychologcal Association; 1993. A neurodevelopmental view of impulsivity and its relationship to the superfactors of personality.. In: editors. The impulsive client: thory.research and treatment. [Google Scholar]
  • 17.Robbins T, Curran H, de Wit H. Special issue on impulsivity and compulsivity. Psychopharmacology (Berl) 2012;219(2):251–2. doi: 10.1007/s00213-011-2584-x. [DOI] [PubMed] [Google Scholar]
  • 18.Allen TJ, Moeller FG, Rhoades HM, Cherek DR. Impulsivity and history of drug dependence. Drug Alcohol Depend. 1998;50(2):137–45. doi: 10.1016/s0376-8716(98)00023-4. [DOI] [PubMed] [Google Scholar]
  • 19.Miyake A, Friedman NP, Emerson MJ, Witzki AH, Howerter A, Wager TD. The unity and diversity of executive functions and their contributions to complex "Frontal Lobe" tasks: a latent variable analysis. Cogn Psychol. 2000;41(1):49–100. doi: 10.1006/cogp.1999.0734. [DOI] [PubMed] [Google Scholar]
  • 20.Whelan R, Conrod PJ, Poline JB , et al. Adolescent impulsivity phenotypes characterized by distinct brain networks. Nat Neurosci. 2012;15(6):920–5. doi: 10.1038/nn.3092. [DOI] [PubMed] [Google Scholar]
  • 21.Moeller FG, Barratt ES, Dougherty DM, Schmitz JM, Swann AC. Psychiatric aspects of impulsivity. Am J Psychiatry. 2001;158(11):1783–93. doi: 10.1176/appi.ajp.158.11.1783. [DOI] [PubMed] [Google Scholar]
  • 22.Rachlin H, Green L. Commitment choice and self-control. J Exp Anal Behav. 1972;17(1):15–22. doi: 10.1901/jeab.1972.17-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Logan GD, Schachar RJ, Tannock R. Impulsivity and inhibitory control. Psychological Sci. 1997;8:60–4. [Google Scholar]
  • 24.Goldstein RZ, Volkow ND. Dysfunction of the prefrontal cortex in addiction neuroimaging findings and clinical implications. Nat Rev Neurosci. 2011;12(11):652–69. doi: 10.1038/nrn3119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Patton JH, Stanford MS, Barratt ES. Factor structure of the Barratt impulsiveness scale. J Clin Psychol. 1995;51(6):768–74. doi: 10.1002/1097-4679(199511)51:6<768::aid-jclp2270510607>3.0.co;2-1. [DOI] [PubMed] [Google Scholar]
  • 26.Whiteside SP, Lynam DR. Understanding the role of impulsivity and externalizing psychopathology in alcohol abuse application of the UPPS impulsive behavior scale. Exp Clin Psychopharmacol. 2003;11(3):210–7. doi: 10.1037/1064-1297.11.3.210. [DOI] [PubMed] [Google Scholar]
  • 27.Kirby KN, Petry NM. Heroin and cocaine abusers have higher discount rates for delayed rewards than alcoholics or non-drug-using controls. Addiction. 2004;99(4):461–71. doi: 10.1111/j.1360-0443.2003.00669.x. [DOI] [PubMed] [Google Scholar]
  • 28.Smith AM, Fried PA, Hogan MJ, Cameron I. Effects of prenatal marijuana on response inhibition an fMRI study of young adults. Neurotoxicol Teratol. 2004;26(4):533–42. doi: 10.1016/j.ntt.2004.04.004. [DOI] [PubMed] [Google Scholar]
  • 29.Hermann D, Sartorius A, Welzel Hetal. Dorsolateral prefrontal cortex N-acetylaspartate/total creatine (NAA/tCr) loss in male recreational cannabis users. Biol Psychiatry. 2007;61(11):1281–9. doi: 10.1016/j.biopsych.2006.08.027. [DOI] [PubMed] [Google Scholar]
  • 30.Theunissen EL, Kauert GF, Toennes SW , et al. Neurophysiological functioning of occasional and heavy cannabis users during THC intoxication. Psychopharmacology (Berl). 2012;220(2):341–50. doi: 10.1007/s00213-011-2479-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fitzgerald PB, Williams S, Daskalakis ZJ. A transcranial magnetic stimulation study of the effects of cannabis use on motor cortical inhibition and excitability. Neuropsychopharmacology. 2009;34(11):2368–75. doi: 10.1038/npp.2009.71. [DOI] [PubMed] [Google Scholar]
  • 32.Weinstein A, Brickner O, Lerman H , 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. 2008;22(4):441–51. doi: 10.1177/0269881108088194. [DOI] [PubMed] [Google Scholar]
  • 33.O'Leary DS, Block RI, Turner BM , et al. Marijuana alters the human cerebellar clock. Neuroreport. . 2003;14(8):1145–51. doi: 10.1097/00001756-200306110-00009. [DOI] [PubMed] [Google Scholar]
  • 34.Gruber SA, Dahlgren MK, Sagar KA, Gonenc A, Killgore WD. Age of onset of marijuana use impacts inhibitory processing. Neurosci Lett. 2012;511(2):89–94. doi: 10.1016/j.neulet.2012.01.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hester R, Nestor L, Garavan H. Impaired error awareness and anterior cingulate cortex hypoactivity in chronic cannabis users. Neuropsychopharmacology. 2009;34(11):2450–8. doi: 10.1038/npp.2009.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bolla KI, Brown K, Eldreth D, Tate K, Cadet JL. Dose-related neurocognitive effects of marijuana use. Neurology. 2002;59(9):1337–43. doi: 10.1212/01.wnl.0000031422.66442.49. [DOI] [PubMed] [Google Scholar]
  • 37.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(3):231–42. doi: 10.1037/a0023034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Silveri MM, Jensen JE, Rosso IM, Sneider JT, Yurgelun-Todd DA. Preliminary evidence for white matter metabolite differences in marijuana-dependent young men using 2D J-resolved magnetic resonance spectroscopic imaging at 4 Tesla. Psychiatry Res. 2011;191(3):201–11. doi: 10.1016/j.pscychresns.2010.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Churchwell JC, Lopez-Larson M, Yurgelun-Todd DA. Altered frontal cortical volume and decision making in adolescent cannabis users. Front Psychol. 2010;1:225. doi: 10.3389/fpsyg.2010.00225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Weinstein A, Brickner O, Lerman H , et al. Brain imaging study of the acute effects of Delta9-tetrahydrocannabinol (THC) on attention and motor coordination in regular users of marijuana. Psychopharmacology (Berl) 2008;196(1):119–31. doi: 10.1007/s00213-007-0940-7. [DOI] [PubMed] [Google Scholar]
  • 41.Gruber SA, Yurgelun-Todd DA. Neuroimaging of marijuana smokers during inhibitory processing: a pilot investigation. Brain Res Cogn Brain Res. 2005;23(1):107–18. doi: 10.1016/j.cogbrainres.2005.02.016. [DOI] [PubMed] [Google Scholar]
  • 42.Tapert SF, Schweinsburg AD, Drummond SP , et al. Functional MRI of inhibitory processing in abstinent adolescent marijuana users. Psychopharmacology (Berl) 2007;194(2):173–83. doi: 10.1007/s00213-007-0823-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Eldreth DA, Matochik JA, Cadet JL, Bolla KI. Abnormal brain activity in prefrontal brain regions in abstinent marijuana users. Neuroimage. 2004;23(3):914–20. doi: 10.1016/j.neuroimage.2004.07.032. [DOI] [PubMed] [Google Scholar]
  • 44.Bolla KI, Eldreth DA, Matochik JA, Cadet JL. Neural substrates of faulty decision-making in abstinent marijuana users. Neuroimage. 2005;26(2):480–92. doi: 10.1016/j.neuroimage.2005.02.012. [DOI] [PubMed] [Google Scholar]
  • 45.Evenden JL. Varieties of impulsivity. Psychopharmacology (Berl) 1999;146(4):348–61. doi: 10.1007/pl00005481. [DOI] [PubMed] [Google Scholar]
  • 46.Olmstead MC. Animal models of drug addiction: Where do we go from here?. Q J Exp Psychol (Hove) 2006;59(4):625–53. doi: 10.1080/17470210500356308. [DOI] [PubMed] [Google Scholar]
  • 47.Perry JL, Carroll ME. The role of impulsive behavior in drug abuse. Psychopharmacology (Berl) 2008;200(1):1–26. doi: 10.1007/s00213-008-1173-0. [DOI] [PubMed] [Google Scholar]
  • 48.Metrik J, Kahler CW, Reynolds B , et al. Balanced placebo design with marijuana: pharmacological and expectancy effects on impulsivity and risk taking. Psychopharmacology (Berl) 2012;223(4):489–99. doi: 10.1007/s00213-012-2740-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ramaekers JG, Kauert G, Theunissen EL, Toennes SW, Moeller MR. Neurocognitive performance during acute THC intoxication in heavy and occasional cannabis users. J Psychopharmacol. 2009;23(3):266–77. doi: 10.1177/0269881108092393. [DOI] [PubMed] [Google Scholar]
  • 50.Ramaekers JG, Kauert G. van Ruitenbeek P, Theunissen EL. Schneider E, Moeller MR. High-potency marijuana impairs executive function and inhibitory motor control. Neuropsychopharmacology. 2006;31(10):2296–303. doi: 10.1038/sj.npp.1301068. [DOI] [PubMed] [Google Scholar]
  • 51.Hunault CC, Mensinga TT, Bocker KB , et al. Cognitive and psychomotor effects in males after smoking a combination of tobacco and cannabis containing up to 69 mg delta-9-tetrahydrocannabinol (THC). Psychopharmacology (Berl) 2009;204(1):85–94. doi: 10.1007/s00213-008-1440-0. [DOI] [PubMed] [Google Scholar]
  • 52.McDonald J, Schleifer L, Richards JB, de Wit H. Effects of THC on behavioral measures of impulsivity in humans. Neuropsychopharmacology. 2003;28(7):1356–65. doi: 10.1038/sj.npp.1300176. [DOI] [PubMed] [Google Scholar]
  • 53.Hart CL, van Gorp W, Haney M, Foltin RW, Fischman MW. Effects of acute smoked marijuana on complex cognitive performance. Neuropsychopharmacology. 2001;25(5):757–65. doi: 10.1016/S0893-133X(01)00273-1. [DOI] [PubMed] [Google Scholar]
  • 54.Moreno M, Estevez AF, Zaldivar F , et al. Impulsivity differences in recreational cannabis users and binge drinkers in a university population. Drug Alcohol Depend. 2012;124(3):355–62. doi: 10.1016/j.drugalcdep.2012.02.011. [DOI] [PubMed] [Google Scholar]
  • 55.Croft RJ, Mackay AJ, Mills AT, Gruzelier JG. The relative contributions of ecstasy and cannabis to cognitive impairment. Psychopharmacology (Berl) 2001;153(3):373–9. doi: 10.1007/s002130000591. [DOI] [PubMed] [Google Scholar]
  • 56.Solowij N, Pesa N, editors. Cambridge University Press: In: D. C. R. M. DC DS. Marijuana and madness. 2nd edition ed. Cambridge:; 2012. Cannabis and cognition: short and long-term effects. pp. 91–102. [Google Scholar]
  • 57.Crean RD, Crane NA, Mason BJ. An evidence based review of acute and long-term effects of cannabis use on executive cognitive functions. J Addict Med. 2011;5(1):1–8. doi: 10.1097/ADM.0b013e31820c23fa. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hall W, Solowij N. Adverse effects of cannabis. Lancet. 1998;352(9140):1611–6. doi: 10.1016/S0140-6736(98)05021-1. [DOI] [PubMed] [Google Scholar]
  • 59.Lane SD, Cherek DR, Tcheremissine OV, Lieving LM, Pietras CJ. Acute marijuana effects on human risk taking. Neuropsychopharmacology. 2005;30(4):800–9. doi: 10.1038/sj.npp.1300620. [DOI] [PubMed] [Google Scholar]
  • 60.Whitlow CT, Liguori A, Livengood LB , et al. Long-term heavy marijuana users make costly decisions on a gambling task. Drug Alcohol Depend. 2004;76(1):107–11. doi: 10.1016/j.drugalcdep.2004.04.009. [DOI] [PubMed] [Google Scholar]
  • 61.Griffith-Lendering MF, Huijbregts SC, Vollebergh WA, Swaab H. Motivational and cognitive inhibitory control in recreational cannabis users. J Clin Exp Neuropsychol. 2012;34(7):688–97. doi: 10.1080/13803395.2012.668874. [DOI] [PubMed] [Google Scholar]
  • 62.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(4):357–64. doi: 10.1080/13803390600693615. [DOI] [PubMed] [Google Scholar]
  • 63.Roberts GM, Garavan H. Evidence of increased activation underlying cognitive control in ecstasy and cannabis users. Neuroimage. 2010;52(2):429–35. doi: 10.1016/j.neuroimage.2010.04.192. [DOI] [PubMed] [Google Scholar]
  • 64.Mostofsky SH, Schafer JG, Abrams MT , et al. fMRI evidence that the neural basis of response inhibition is task-dependent. Brain Res Cogn Brain Res. 2003;17(2):419–30. doi: 10.1016/s0926-6410(03)00144-7. [DOI] [PubMed] [Google Scholar]
  • 65.Rubia K, Russell T, Overmeyer S , et al. Mapping motor inhibition: conjunctive brain activations across different versions of go/no-go and stop tasks. Neuroimage. 2001;13(2):250–61. doi: 10.1006/nimg.2000.0685. [DOI] [PubMed] [Google Scholar]
  • 66.Friedman NP, Miyake A, Young SE, Defries JC, Corley RP, Hewitt JK. Individual differences in executive functions are almost entirely genetic in origin. J Exp Psychol Gen. 2008;137(2):201–25. doi: 10.1037/0096-3445.137.2.201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Groot AS, de Sonneville LM, Stins JF, Boomsma DI. Familial influences on sustained attention and inhibition in preschoolers. J Child Psychol Psychiatry. 2004;45(2):306–14. doi: 10.1111/j.1469-7610.2004.00222.x. [DOI] [PubMed] [Google Scholar]
  • 68.Heiser P, Heinzel-Gutenbrunner M, Frey J , et al. Twin study on heritability of activity. attenion.and impulsivity as assessed by objective measures. J Atten Disord . 2006; 9(4):575–81. doi: 10.1177/1087054705284298. [DOI] [PubMed] [Google Scholar]
  • 69.Schachar RJ, Forget-Dubois N, Dionne G, Boivin M, Robaey P. Heritability of response inhibition in children. J Int Neuropsychol Soc. 2011;17(2):238–47. doi: 10.1017/S1355617710001463. [DOI] [PubMed] [Google Scholar]
  • 70.Aron AR, Fletcher PC, Bullmore ET, Sahakian BJ, Robbins TW. Stop-signal inhibition disrupted by damage to right inferior frontal gyrus in humans. Nat Neurosci. 2003;6(2):115–6. doi: 10.1038/nn1003. [DOI] [PubMed] [Google Scholar]
  • 71.Aron AR, Robbins TW, Poldrack RA. Inhibition and the right inferior frontal cortex. Trends Cogn Sci. 2004;8(4):170–7. doi: 10.1016/j.tics.2004.02.010. [DOI] [PubMed] [Google Scholar]
  • 72.Aron AR, Durston S, Eagle DM, Logan GD, Stinear CM, Stuphorn V. Converging evidence for a fronto-basal-ganglia network for inhibitory control of action and cognition. J Neurosci. 2007;27(44):11860–4. doi: 10.1523/JNEUROSCI.3644-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Eagle DM, Baunez C, Hutcheson DM, Lehmann O, Shah AP, Robbins TW. Stop-signal reaction-time task performance: role of prefrontal cortex and subthalamic nucleus. Cereb Cortex. 2008;18(1):178–88. doi: 10.1093/cercor/bhm044. [DOI] [PubMed] [Google Scholar]
  • 74.Picton TW, Stuss DT, Alexander MP, Shallice T, Binns MA, Gillingham S. Effects of focal frontal lesions on response inhibition. Cereb Cortex. 2007;17(4):826–38. doi: 10.1093/cercor/bhk031. [DOI] [PubMed] [Google Scholar]
  • 75.Rieger M, Gauggel S, Burmeister K. Inhibition of ongoing responses following frontal. nonfrotal.and basal ganglia lesions. Neuropsychology. 2003; 17(2):272–82. doi: 10.1037/0894-4105.17.2.272. [DOI] [PubMed] [Google Scholar]
  • 76.Rubia K, Smith AB, Taylor E, Brammer M. Linear age-correlated functional development of right inferior fronto-striato-cerebellar networks during response inhibition and anterior cingulate during error-related processes. Hum Brain Mapp. 2007;28(11):1163–77. doi: 10.1002/hbm.20347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Rubia K, Smith AB, Brammer MJ, Taylor E. Right inferior prefrontal cortex mediates response inhibition while mesial prefrontal cortex is responsible for error detection. Neuroimage. 2003;20(1):351–8. doi: 10.1016/s1053-8119(03)00275-1. [DOI] [PubMed] [Google Scholar]
  • 78.Wostmann NM, Aichert DS, Costa A, Rubia K, Moller HJ, Ettinger U. Reliability and plasticity of response inhibition and interference control. Brain Cogn. 2013;81(1):82–94. doi: 10.1016/j.bandc.2012.09.010. [DOI] [PubMed] [Google Scholar]
  • 79.Garavan H, Ross TJ, Murphy K, Roche RA, Stein EA. Dissociable executive functions in the dynamic control of behavior: inhibition. error detecion.and correction. Neuroimage. 2002; 17(4):1820–9. doi: 10.1006/nimg.2002.1326. [DOI] [PubMed] [Google Scholar]
  • 80.Garavan H, Hester R, Murphy K, Fassbender C, Kelly C. Individual differences in the functional neuroanatomy of inhibitory control. Brain Res. 2006;1105(1):130–42. doi: 10.1016/j.brainres.2006.03.029. [DOI] [PubMed] [Google Scholar]
  • 81.Verdejo-Garcia A, Benbrook A, Funderburk F, David P, Cadet JL, 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(1):2–11. doi: 10.1016/j.drugalcdep.2007.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Tanabe J, Thompson L, Claus E, Dalwani M, Hutchison K, Banich MT. Prefrontal cortex activity is reduced in gambling and nongambling substance users during decision-making. Hum Brain Mapp. 2007;28(12):1276–86. doi: 10.1002/hbm.20344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ernst M, Bolla K, Mouratidis M , et al. Decision-making in a risk-taking task: a PET study. Neuropsychopharmacology. 2002 May ;26(5):682–91. doi: 10.1016/S0893-133X(01)00414-6. [DOI] [PubMed] [Google Scholar]
  • 84.Bartzokis G, Lu PH, Beckson M , et al. Abstinence from cocaine reduces high-risk responses on a gambling task. Neuropsychopharmacology. 2000;22(1):102–3. doi: 10.1016/S0893-133X(99)00077-9. [DOI] [PubMed] [Google Scholar]
  • 85.Bonson KR, Grant SJ, Contoreggi CS , et al. Neural systems and cue-induced cocaine craving. Neuropsychopharmacology. 2002;26(3):376–86. doi: 10.1016/S0893-133X(01)00371-2. [DOI] [PubMed] [Google Scholar]
  • 86.Bechara A, Damasio AR, Damasio H, Anderson SW. Insensitivity to future consequences following damage to human prefrontal cortex. Cognition. 1994;50(1-3):7–15. doi: 10.1016/0010-0277(94)90018-3. [DOI] [PubMed] [Google Scholar]
  • 87.Bechara A, Damasio H, Damasio AR, Lee GP. Different contributions of the human amygdala and ventromedial prefrontal cortex to decision-making. J Neurosci. 1999;19(13):5473–81. doi: 10.1523/JNEUROSCI.19-13-05473.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Bolla KI, Eldreth DA, London ED , et al. Orbitofrontal cortex dysfunction in abstinent cocaine abusers performing a decision-making task. Neuroimage. 2003;19(3):1085–94. doi: 10.1016/s1053-8119(03)00113-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Bechara A, Tranel D, Damasio H. Characterization of the decision-making deficit of patients with ventromedial prefrontal cortex lesions. Brain. 2000;123 (Pt 11):2189–202. doi: 10.1093/brain/123.11.2189. [DOI] [PubMed] [Google Scholar]
  • 90.DeLisi LE. The effect of cannabis on the brain: can it cause brain anomalies that lead to increased risk for schizophrenia?. Curr Opin Psychiatry. 2008;21(2):140–50. doi: 10.1097/YCO.0b013e3282f51266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Solowij N, Yucel M, Lorenzetti V, editors. Endophenotyes., and Genes. Dordrecht: The Handbook of Neuropsychiatric Biomarkers.Springer; 2009. Structural brain alterations in cannabis users: association with cognitive deficits and psychiatric symptoms.In: Ritsne MS. pp. 215–25. [Google Scholar]
  • 92.Lorenzetti V, Lubman DI, Whittle S, Solowij N, Yucel M. Structural MRI findings in long-term cannabis users: what do we know?. Subst Use Misuse. 2010;45(11):1787–808. doi: 10.3109/10826084.2010.482443. [DOI] [PubMed] [Google Scholar]
  • 93.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(4):3845–51. doi: 10.1016/j.neuroimage.2011.09.046. [DOI] [PubMed] [Google Scholar]
  • 94.Mata I, Perez-Iglesias R, Roiz-Santianez R, Tordesillas-Gutierrez D, Pazos A, Gutierrez A , et al. Gyrification brain abnormalities associated with adolescence and early-adulthood cannabis use. Brain Res. 2010;1317:297–304. doi: 10.1016/j.brainres.2009.12.069. [DOI] [PubMed] [Google Scholar]
  • 95.Rapp C, Bugra H, Riecher-Rossler A, Tamagni C, Borgwardt S. Effects of cannabis use on human brain structure in psychosis: a systematic review combining in vivo structural neuroimaging and post mortem studies. Curr Pharm Des. 2012;18(32):5070–80. doi: 10.2174/138161212802884861. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ashtari M, Cervellione K, Cottone J, Ardekani BA, Sevy S, Kumra S. Diffusion abnormalities in adolescents and young adults with a history of heavy cannabis use. J Psychiatr Res. 2009;43(3):189–204. doi: 10.1016/j.jpsychires.2008.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Arnone D, Abou-Saleh MT, Barrick TR. Diffusion tensor imaging of the corpus callosum in addiction. Neuropsychobiology. 2006;54(2):107–13. doi: 10.1159/000096992. [DOI] [PubMed] [Google Scholar]
  • 98.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(3):1067–74. doi: 10.1016/j.neuroimage.2008.02.064. [DOI] [PubMed] [Google Scholar]
  • 99.Matochik JA, Eldreth DA, Cadet JL, Bolla KI. Altered brain tissue composition in heavy marijuana users. Drug Alcohol Depend. 2005;77(1):23–30. doi: 10.1016/j.drugalcdep.2004.06.011. [DOI] [PubMed] [Google Scholar]
  • 100.Medina KL, Schweinsburg AD, Cohen-Zion M, Nagel BJ, Tapert SF. Effects of alcohol and combined marijuana and alcohol use during adolescence on hippocampal volume and asymmetry. Neurotoxicol Teratol. 2007;29(1):141–52. doi: 10.1016/j.ntt.2006.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Yucel M, Solowij N, Respondek C , et al. Regional brain abnormalities associated with long-term heavy cannabis use. Arch Gen Psychiatry. 2008;65(6):694–701. doi: 10.1001/archpsyc.65.6.694. [DOI] [PubMed] [Google Scholar]
  • 102.Solowij N, Yucel M, Respondek C , et al. Cerebellar white-matter changes in cannabis users with and without schizophrenia. Psychol Med. 2011;41(11):2349–59. doi: 10.1017/S003329171100050X. [DOI] [PubMed] [Google Scholar]
  • 103.Cohen M, Rasser PE, Peck G , et al. Cerebellar grey-matter deficits. cannabis use and first-episode schizophrenia in adolescents and young adults. Int J Neuropsychopharmacol. 2011:1–11. doi: 10.1017/S146114571100068X. [DOI] [PubMed] [Google Scholar]
  • 104.Zalesky A, Solowij N, Yucel M , et al. Effect of long-term cannabis use on axonal fibre connectivity. Brain. 2012;135(Pt 7):2245–55. doi: 10.1093/brain/aws136. [DOI] [PubMed] [Google Scholar]
  • 105.Denier N, Walter M, Bendfeldt K, Lang U, Borgwardt S. Resting State Abnormalities in Psychosis Compared to Acute Cannabinoids and Opioids Challenges: A Systematic Review of Functional Imaging Studies. Curr Pharm Des. 2012;18(32):5081–5092. doi: 10.2174/138161212802884717. [DOI] [PubMed] [Google Scholar]
  • 106.Chang L, Chronicle EP. Functional Imaging Studies in Cannabis Users. Neuroscientist. 2007;13(5):422–32. doi: 10.1177/1073858406296601. [DOI] [PubMed] [Google Scholar]
  • 107.Batalla A, Bhattacharyya S, Yücel M , et al. Structural and Functional Imaging Studies in Chronic Cannabis Users: A Systematic Review of Adolescent and Adult Findings. PLOS One. 2013;8(2):1–18. doi: 10.1371/journal.pone.0055821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Ersche KD, Turton AJ, Pradhan S, Bullmore ET, Robbins TW. Drug addiction endophenotypes: impulsive versus sensation-seeking personality traits. Biol Psychiatry. 2010;68(8):770–3. doi: 10.1016/j.biopsych.2010.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Robbins TW, Gillan CM, Smith DG, de Wit S, Ersche KD. Neurocognitive endophenotypes of impulsivity and compulsivity: towards dimensional psychiatry. Trends Cogn Sci. 2012;16(1):81–91. doi: 10.1016/j.tics.2011.11.009. [DOI] [PubMed] [Google Scholar]
  • 110.Ashton CH. Pharmacology and effects of cannabis a brief review. Br J Psychiatry. 2001;178:101–6. doi: 10.1192/bjp.178.2.101. [DOI] [PubMed] [Google Scholar]
  • 111.Cousijn J, Wiers RW, Ridderinkhof KR, et al. Individual differences in decision making and reward processing predict changes in cannabis use a prospective functional magnetic resonance imaging study. Addict Biol. 2012;21 doi: 10.1111/j.1369-1600.2012.00498.x. [DOI] [PubMed] [Google Scholar]

Articles from Current Pharmaceutical Design are provided here courtesy of Bentham Science Publishers

RESOURCES