+ All Categories
Home > Documents > Evaluating The Effects of Cannabis Dependence and ...

Evaluating The Effects of Cannabis Dependence and ...

Date post: 16-Oct-2021
Category:
Upload: others
View: 2 times
Download: 0 times
Share this document with a friend
148
Evaluating The Effects of Cannabis Dependence and Abstinence on Cortical Inhibition and Working Memory in Schizophrenia by Michelle Samantha Goodman A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Sciences University of Toronto © Copyright by Michelle Samantha Goodman 2015
Transcript
Page 1: Evaluating The Effects of Cannabis Dependence and ...

Evaluating The Effects of Cannabis Dependence and Abstinence on Cortical Inhibition and Working Memory

in Schizophrenia

by

Michelle Samantha Goodman

A thesis submitted in conformity with the requirements for the degree of Master of Science

Institute of Medical Sciences University of Toronto

© Copyright by Michelle Samantha Goodman 2015

Page 2: Evaluating The Effects of Cannabis Dependence and ...

II  

The Effects The Effects of Cannabis Abstinence on Cortical

Inhibition and Working Memory in Schizophrenia

Michelle Samantha Goodman Master of Science

Institute of Medical Sciences University of Toronto

2015

Background: Cannabis is the most commonly used illicit substance among schizophrenia

patients. GABA is inhibited by cannabis and dysfunctional GABAergic neurotransmission

underlies cognitive and cortical inhibitory deficits in schizophrenia patients. This study

investigated the relationship between cannabis dependence, schizophrenia, cortical inhibition

and cognition.

Methodology: Using transcranial magnetic stimulation and the N-back task with

electroencephalography, we assessed cortical inhibition and working memory at baseline and

following 28-days of abstinence in 12 cannabis dependent patients versus 14 cannabis

dependent non-psychiatric controls.

Results: Enhanced GABAB activity, through a prolonged cortical silent period, was found in

patients compared to controls at baseline, with no differences following abstinence. No

differences were found on working memory accuracy or gamma activity.

Conclusions: While these findings suggest that cannabis dependence may have more

impairing effects on GABAB mediated inhibition in controls compared to patients, it is clear

that continued research is needed to better understand this common co-morbidity.

Page 3: Evaluating The Effects of Cannabis Dependence and ...

III  

Acknowledgments

I would like to express my sincerest gratitude to Dr. Tony George, my supervisor. Thank you for taking a chance on me two years ago and for allowing me to be a part of your amazing team. Your encouragement and confidence in my academic abilities have motivated me to continue on with a career in research.

This study would not have been possible without the guidance and support of Dr. Mera Barr. Thank you for your mentorship, your friendship, and your constant support. I am so grateful that I have the opportunity to continue to work with you and learn from you.

To Alanna, my partner in crime, these past two years would not have been the same without you. I knew we were going to be lifelong friends the day we met (even though you spent most of the day talking about the leafs). Thank you for always keeping me organized and on track, for helping me navigate the confusing halls of med sci, and for always knowing how to make me laugh.

I’d like to give a special thank you to Rachel Rabin, my cannabis teammate throughout these past two years. To Karolina Kozak and Kristen Mackowick, my office mates, thank you for listening to me vent and always laughing at my bad jokes. To all of the wonderful lab mates that I have worked with throughout my time in the BACDRL lab: Emily Simpkin, Maryam Sharif-Razi, Marya Morozova, Matt Tracey, Anushka Talpur, thank you!

To my family members, both given and chosen, I am indebted to you for your unwavering support, guidance and encouragement. To my parents and sisters, thank you for your unconditional love and for supporting me throughout this journey.

And finally, I would like to thank the amazing participants who were integral to this study. I have learned so much from you all and this work truly would not have been possible without your contributions.

Page 4: Evaluating The Effects of Cannabis Dependence and ...

IV  

Contributions

The thesis was a subset of a larger study entitled “Effects of Cannabis Abstinence on Neurocognition in Schizophrenia”. This study was developed by Dr. Tony George (PI), Dr. Konstantine Zakzanis, Rachel Rabin, Dr. Jeff Daskalakis, and Dr. Mera Barr. The neurophysiologic subset of the study was funded by Dr. Mera Barr’s young investigator NARSAD award and by Dr. Tony George’s CIHR grant.

The MSc. candidate, Michelle S. Goodman, was responsible for recruitment, retention, and conduct of all study sessions. Additionally, Michelle conducted all data analysis, interpretation of research data, and drafting of the thesis, with assistance from Dr. Tony George and Dr. Mera Barr. Dr. Mera Barr trained the candidate on all neurophysiological and neurocognitive measures, including working memory and paired associative stimulation. Rachel Rabin was responsible for conducting the structured clinical interviews and physical examinations performed during the screening session.

The Program Advisory Committee (PAC) members, Dr. Daniel Blumberger and Dr. Martin Zack, assisted with the interpretation of the data and provided feedback on the thesis.

 

Page 5: Evaluating The Effects of Cannabis Dependence and ...

V  

Table of Contents

Pages  

Abstract………………………………………………………..………………………

Acknowledgments……………………………………………..………………………

Contributions…………………………………………………..………………………

Table of Contents………………………………………………...................................

List of Abbreviations…………………………………………………………….……

List of Tables………………………………………………………..…………………

List of Figures……………………………………………………...…………………..

List of Appendices……………………………………………………………………..

II

III

IV

V

IX

XI

XII

XV

Chapter 1: Literature Review…………………………………….…………

1.1 Schizophrenia…………………………………………….…….…………...... 1

1.1.1 Phenomenology, Epidemiology, and Etiology…………………..…….……… 1

1.1.2 Pathophysiology of Schizophrenia…………………………….……….……... 2

1.1.2.1

1.1.2.2

1.1.2.3

Dopamine……………………………………………………………..........

GABA…………………………………………………………...................

Glutamate………………………………………………….……….............

Cannabis.………………….………………………………………….……….

Pharmacokinetics of Cannabis………………………………………....………

Acute Psychoactive Effects of Cannabis…………………….……..…..………

3

3

5

6

7

9

1.2

1.2.1

1.2.2

1.2.3 Somatic and Health Effects of Cannabis…………………….……………....… 10

1.2.4 Cognitive Effects of Cannabis…………………….…………......……..……… 11

1.2.5

1.3

Treatment of Cannabis Dependence…………………..…….………………….

The Endocannabinoid System…..……………………………………………

12

13

1.4 Cannabis Use in Schizophrenia…..…….…………………………….……… 16

Page 6: Evaluating The Effects of Cannabis Dependence and ...

VI  

1.4.1

1.4.2

Effects on Prognosis and Course…………….…………………………………

Effects on Cognition………………………………..………………………….

16

17

1.4.3

1.5

Underlying Neurobiological Connection………………………………………

Cortical Inhibition………………………………………………………...….

18

19

1.5.1 Indexing Cortical Inhibition using Transcranial Magnetic Stimulation……..... 19

1.5.2 Cortical Inhibitory Deficits in Schizophrenia……..…………..………………. 22

1.5.3 Cortical Inhibitory Deficits with Cannabis Use……………..……………..…. 26

1.5.4 The Combined Effects of Cannabis Use and Schizophrenia on Cortical Inhibition.………………………...…………………………………………….

26

1.6 Working Memory……………………………………………………………. 27

1.6.1 The Neurophysiology of Working Memory.......................….…………..…… 27

1.6.2 Indexing Working Memory……….………….………………………...…….. 29

1.6.2.1 Neurocognitive Assessments………………………………………..……. 29

1.6.2.2 Neurophysiological Assessments…………………………………..……... 30

1.6.3 Working Memory Deficits in Schizophrenia………….…………..……......... 31

1.6.4 Cannabis’ Effects on Working Memory……………………………..………. 33

1.6.5 The Combined Effects of Cannabis and Schizophrenia on Working Memory. 36

1.7 Overall Of Current Study Objective and Hypotheses………………….... 37

Chapter 2: Methods…………………………………………………..……..

2.1 Study Design…………………………………………………………..…….. 40

2.2 Participants………………………………………………………….……… 42

2.2.1

2.2.2

Power Analysis……………………………………………………..…….......

Total Sample………………………………………………………..………...

42

42

2.3 Measures…………………………………………………………..………… 44

2.3.1 Clinical Interview Measures………………………………………..………... 44

2.3.2 Premorbid IQ……………………………………………………..…………… 45

Page 7: Evaluating The Effects of Cannabis Dependence and ...

VII  

2.3.3 Extrapyramidal Side Effects.………………………………………………..… 46

2.3.4 Substance Abuse Measures…………………………………………………… 47

2.3.5

2.3.6

Cortical Inhibition………………………………………………………..……

Working Memory……………………………………………………..……….

48

52

2.3.7

2.4

Cannabis Abstinence Paradigm……………………………………………..…

Study Procedure…………………………………………………………….….

55

56

2.5 Data Analysis…………………………………...……………………………. 57

2.5.1 Study Sample…………….….………………………………………………… 57

2.5.2 Cortical Inhibition …………….……...…………………………………...….. 58

2.5.3 Working Memory…………………...……………………………………...…. 58

Chapter 3: Results……………………………….…………………………....

3.1 Baseline Results……………………..…………..……………………………. 59

3.1.1 Demographics……………………………………………...………………….. 59

3.1.2 Baseline TMS Assessments………………...…………………………………. 62

3.1.2.1

3.1.2.2

3.1.3

3.1.3.1

3.1.3.2

Baseline Inhibitory Assessments: SICI, LICI, and CSP……………………

Baseline Excitatory Assessments: RMT and ICF…………………………..

Baseline Working Memory Assessments………………………………………

Baseline Working Memory Performance………………………………….

Baseline Gamma Power…………………………………………………...

62

66

68

68

69

3.2 Exploratory Longitudinal Assessments……...……………………………... 71

3.2.1 Longitudinal TMS Assessments..……………………………...……………… 71

3.2.1.1

3.2.1.2

3.2.2

3.2.2.1

3.2.2.2

Longitudinal Inhibitory Assessments: SICI, LICI, and CSP………….….

Longitudinal Excitatory Assessments: ICF……….....................................

Longitudinal Working Memory Assessments…………...................................

Longitudinal Working Memory Performance…………………………….

Longitudinal Gamma Power……………………………………………...

72

75

77

78

79

Page 8: Evaluating The Effects of Cannabis Dependence and ...

VIII  

Chapter 4: Discussion……………………………………………………...

4.1 General Discussion………………………………………………………… 80

4.1.1 Study Sample……………………………………………………………….. 81

4.1.2 TMS Findings……………………………………………………………….. 82

4.1.3 Working Memory Findings……………….………………………………… 87

4.1.4

4.1.5

Exploratory Findings…………………………………………………………

Potential Mechanisms...…………………………..…………………………..

91

92

4.2 Limitations………………………………………..………………………… 97

4.3 Conclusions………………………………………..………………………… 100

4.4 Future Directions…………………………………..……………………….. 101

Literature Cited…………………………………..………………………… 106

Page 9: Evaluating The Effects of Cannabis Dependence and ...

IX  

List of Abbreviations

1 millivolt (1 mV)

2-Arachidonoylglycerol (2-AG)

Abnormal Involuntary Movement Scale (AIMS)

Analysis of Variance (ANOVA)

Barnes Akathisia Rating Scale (BARS)

Biobehavioural Addictions and Concurrent Disorders Research Laboratory (BACDRL)

Carbon Monoxide (CO)

Cannabinoid receptor gene (CNR1)

Cannabinoid Type 1 Receptor (CB1)

Cannabinoid Type 2 Receptor (CB2)

Centre for Addiction and Mental Health (CAMH)

Chlorpromazine (CPZ)

Cigarettes per day (CPD)

Cortical silent period (CSP)

Diagnostic and Statistical Manual of Mental Disorders, 4th. Edition (DSM-IV)

Dorsolateral prefrontal cortex (DLPFC)

Electroencephalography (EEG)

Endocannabinoid (eCB)

Gamma-aminobutyric acid (GABA)

Grams per day (GPD)

Healthy Controls (HC)

Page 10: Evaluating The Effects of Cannabis Dependence and ...

X  

Hopkins Verbal Learning Test (HVLT)

Intelligence Quotient (IQ)

Intercortical facilitation (ICF)

Megnetoencephaolography (MEG)

Long interval cortical inhibition (LICI)

Positron emission tomography (PET)

Positive and Negative Syndrome Scale (PANSS)

Prefrontal cortex (PFC)

Research Ethics Board (REB)

Resting motor threshold (RMT)

Schizophrenia (SZ)

Short interval cortical inhibition (SICI)

Simpson Angus Rating Scale for Extrapyramidal Symptoms (SARS)

Standard Deviation (SD)

Statistical Package for the Social Sciences (SPSS)

Structured Clinical Interview for the DSM-IV (SCID-IV)

Transcranial Magnetic Stimulation (TMS)

Wechsler Test of Adult Reading (WTAR)

Working Memory (WM)

 

 

Page 11: Evaluating The Effects of Cannabis Dependence and ...

XI  

List of Tables

Pages Table 3.1. Baseline TMS demographics, clinical means, and standard deviations by diagnosis………………………………………………………………..… 62 Table 3.2. Cohen’s D values for TMS Measures…………………………………….. 67

Table 3.3. Baseline N-Back demographics, clinical means, and standard deviations by diagnosis…………………………………………………………….......... 68 Table 3.4 Creatinine-normalized THCCOOH levels in patient and control abstainers and relapsers……………………………………………………………….… 71

Table 3.5. Post TMS demographics, clinical means, and standard deviations of the sample by diagnosis…………………………………………….…………………. 72 Table 3.6. Post WM demographics, clinical means, and standard deviations by diagnosis………………………………………………………………….…………….. 77  

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Page 12: Evaluating The Effects of Cannabis Dependence and ...

XII  

List of Figures

Figure 2.1: Outline of overall study design…………………………………………………

Figure 2.2. Electromyography recordings.…………………………………………………

Figure 2.3. N-Back verbal working memory task …..…………………………………….

Figure 3.1. Correlation between IQ and joint years.………………………………………

Figure 3.2. Consort Diagram for patients with schizophrenia and healthy controls….

Figure 3.3. Baseline short interval cortical inhibition (SICI)……………………...…….

Figure 3.4. Baseline long interval cortical inhibition (LICI) …………………….………  Figure 3.5. Baseline cortical silent period (CSP) ……..…………………………….……. Figure 3.6. Baseline cortical facilitation (ICF)……………………………………………. Figure 3.7. Baseline N-back performance between patients and healthy controls…… Figure 3.8. Mean gamma (30-50 Hz) oscillatory power from target correct (TC) responses elicited during the N-back task ………………………………………………….

Figure 3.9. Longitudinal data for short interval cortical inhibition (SICI)……….……

Figure 3.10. Longitudinal data for long interval cortical inhibition (LICI)……………

Figure 3.11. Longitudinal data for cortical silent period (CSP) ……………..…………

Figure 3.12. Longitudinal data for intracortical faciliatation ICF) ………………..…..

Figure 3.13. Longitudinal N-back performance between patients and healthy controls ………………………………………………………………………….……………….

Figure 3.14. Topographical illustration of mean absolute γ power elicited during 1- and 3-back in patients and healthy control ……………………………………………………..

Pages

41

51

53

60

61

63

64

65

66

69

70

73

74

75

76

 

78

79

Page 13: Evaluating The Effects of Cannabis Dependence and ...

XIII  

 List of Appendices

Pages

 A) Telephone Screen.…….…….…………………………………………............ 123

 B) Study Consent………….……………………………………………............… 125

  C) Case Report Form…………………………………………………………... 134  

   

   

 

   

   

   

   

   

   

   

   

 

 

 

 

 

 

 

 

 

Page 14: Evaluating The Effects of Cannabis Dependence and ...

1  

Chapter 1 Literature Review

1.1 Schizophrenia

1.1.1 Phenomenology and Etiology

Schizophrenia is one of the most severe and debilitating brain disorders. While its

prevalence is relatively low compared to other pervasive illnesses, at approximately 1%,

schizophrenia remains to be one of the world’s leading causes of disability (Lewis &

Lieberman, 2000; Whiteford et al., 2013). The early age of onset, high rates of

unemployment, and frequent hospitalizations, highlight the immense personal and societal

burden of this disorder (Rossler, Salize, van Os, & Riecher-Rossler, 2005).

The symptoms associated with schizophrenia are commonly characterized into three

domains, including positive symptoms (e.g., delusions, hallucinations, thought

disorganization), negative symptoms (e.g., blunted affect, alogia, anhedonia, social

dysfunction, amotivation) and cognitive impairment (APA, 2000). Contrary to initial beliefs,

current research supports the notion that functional outcome is better predicted by cognitive

deficits rather than positive symptoms (Green, 2006). Importantly, these cognitive deficits

appear prior to the onset of psychosis and generally persist following stable remission from

positive symptoms (Bora, Yucel, & Pantelis, 2010). Unfortunately, these impairments along

with negative symptoms are not sufficiently treated by current antipsychotic interventions.

The high degree of heterogeneity known to this disorder complicates both diagnostic

and treatment approaches for those with schizophrenia (Andreasen, Arndt, Alliger, Miller, &

Flaum, 1995; Davidson & McGlashan, 1997). This phenotypic heterogeneity is commonly

observed in the age of onset and constellation of symptoms, response to treatment, as well as

Page 15: Evaluating The Effects of Cannabis Dependence and ...

2  

the overall prognosis. Towards addressing this variability, diagnostic subtyping (paranoid,

catatonic, disorganized, undifferentiated, and residual) was introduced in the DSM-IV TR;

however, the efforts to validate these subtypes were deemed unsuccessful. In response, the

DSM-V targeted clinical symptomatology as markers of the disease (American Psychiatric

Association, 2013).

In addition to the complex presentation of this disorder, the etiology of schizophrenia

is poorly understood. Current theories suggest that environmental and/or psychological

stressors interact with specific genetic vulnerabilities contributing to the illnesses

development. A range of environmental factors implicated in this disorder include, viral

infections (Mednick, Machon, Huttunen, & Bonett, 1988), substance abuse (Moore et al.,

2007) (refer to section 1.4.3) and urbanization (Krabbendam & van Os, 2005). Regarding the

genetic predisposition, high heritability contributes to about 80% of the disorder’s liability

(Gottesman, 1991). However, no one gene appears to be either sufficient or necessary for the

development of schizophrenia (Tandon, Keshavan, & Nasrallah, 2008). Of note,

neurobiological evidence suggests that these genetic alterations may be linked to

abnormalities within neurotransmitter systems including dopamine, γ-aminobutyric acid

(GABA) and glutamate (Harrison & Weinberger, 2005).

1.1.2 Pathophysiology of Schizophrenia

1.1.2.1 Dopamine

Dopamine is one of the most pervasive neurotransmitters in the central nervous

system (CNS). The dopaminergic system has been implicated in numerous behavioural

domains including information processing, memory, pleasure and addiction (Jones,

Page 16: Evaluating The Effects of Cannabis Dependence and ...

3  

Kilpatrick, & Phillipson, 1986). Importantly, aberrant dopaminergic functioning is thought to

underlie the pathophysiology of several psychiatric disorders including schizophrenia. More

specifically, the dopamine hypothesis of schizophrenia proposes that excessive dopaminergic

transmission is involved in the onset of psychosis (Carlsson and Lindqvist 1963). This theory

was initially developed resulting from the observation that dopamine-enhancing drugs,

primarily amphetamines, induced psychosis-like symptoms in otherwise healthy individuals

(Seeman and Lee, 1975). In support of this finding, later research revealed that the psychosis

reducing properties of atypical antipsychotic medications were largely due to their D2

receptor blocking capabilities (Lieberman, Kane, and Alvir, 1987). While the first

formulation of this hypothesis accounted for positive symptoms, it could not properly explain

the negative symptoms and cognitive deficits inherent to this disorder. Subsequent

neuroimaging, post-mortem and animal studies provided the basis for a reformulated

hypothesis. This hypothesis incorporated hypodopaminergic activity in the prefrontal cortex

(PFC), better accounting for such cognitive deficits and negative symptoms, along with the

initial observation of hyperdopamiergic activity in other subcortical regions (reviewed in

Davis, Kahn, Ko, & Davidson, 1991).

1.1.2.2 GABA

Dysfunctional GABAergic neurotransmission has also been implicated in the

pathophysiology of schizophrenia (Tse, Piantadosi, & Floresco, 2014). As the primary

inhibitory neurotransmitter in the brain, GABA supresses cortical activity by hyperpolarizing

the cell and reducing the neuronal firing of several excitatory neurotransmitters including

dopamine, noradrenaline, and serotonin. GABA is primarily synthesized from glutamate, and

is comprised of two general classes of receptors: ionotropic GABAA receptors and

Page 17: Evaluating The Effects of Cannabis Dependence and ...

4  

metabotropic GABAB receptors. GABA acts at inhibitory synapses in the brain, binding to

both presynaptic receptors and postsynaptic receptors. This binding ultimately leads to

hyperpolarization due to the influx of negatively charged chloride ions into the cell or

positively charged potassium ions out of the cell.

Following the pivotal discovery of GABA’s role in inhibitory control, the GABA

hypothesis of schizophrenia was developed (Roberts & Frankel, 1950). In support of this,

post-mortem studies revealed numerous deviations in GABAergic functioning. For example,

cortical reductions in the messenger RNA and protein for glutamic acid decarboxylase-67

(GAD 67), an enzyme necessary for the synthesis of GABA, have been found in the DLPFC

(Akbarian & Huang, 2006) and other regions (Hashimoto et al., 2008) in individuals with

schizophrenia. Furthermore, advances in neurophysiological methodologies have allowed

researchers to investigate potential GABAergic neurotransmitter dysfunctions in vivo.

Studies utilizing non-invasive brain stimulation techniques have provided insight into

specific GABAA and GABAB receptor deficits in individuals with schizophrenia (Daskalakis,

Christensen, Chen, et al., 2002; Farzan et al., 2010a; Takahashi et al., 2013). Similarly,

position emission tomography (PET) studies have revealed that aberrant GABA functioning,

specifically in the PFC and hippocampus, may underlie negative symptoms (Asai et al.,

2008) and cognitive deficits in those with schizophrenia (Lewis, Volk, & Hashimoto, 2004).

Clinical findings and pharmacological studies have provided further support for the

involvement of GABA in schizophrenia. For example, benzodiazepines have been shown to

reduce the likelihood of experiencing a psychotic episode during relapse (Carpenter,

Buchanan, Kirkpatrick, & Breier, 1999; Hashimoto et al., 2008). Given GABAs capacity to

reduce synaptic dopamine levels, GABA was originally targeted as a potential treatment for

Page 18: Evaluating The Effects of Cannabis Dependence and ...

5  

schizophrenia; however this approach proved to be somewhat unsuccessful. Instead of

focusing on the GABA’s involvement in positive symptoms, researchers have directed their

efforts towards better understanding the underlying association between aberrant GABA

neurotransmission and cognitive dysfunction.

1.1.2.3 Glutamate

Over the past several decades, evidence supporting the involvement of glutamatergic

neurotransmission in the pathophysiology of schizophrenia has grown substantially.

Glutamate is not only a key precursor of GABA, it is also the most abundant excitatory

neurotransmitter in the brain. Glutamatergic neurotransmission acts through metabotropic

and ionotropic receptors, subdivided into N-methyl-D-aspartate (NMDA) receptors, α-

amino-3-hydroxy-5-methyl-4-isoazolepropionic acid (AMPA) receptors, and kainite. Given

its prevalence in the CNS, glutamate is thought to play a key role in several cognitive and

behavioural domains including synaptic plasticity, learning and memory, and executive

function (Johnson, 1972).

Early observations noted that phencyclidine (PCP) and ketamine, commonly abused

NMDA antagonists, induced schizophrenia-like symptoms in otherwise healthy individuals

(Javitt, 2007; Lahti, Holcomb, Gao, & Tamminga, 1999; Lahti et al., 1997) and significantly

exacerbated positive symptoms in those with schizophrenia (Lahti, Weiler, Tamara

Michaelidis, Parwani, & Tamminga, 2001). Thus, it was originally hypothesized that

hypofunctional glutamatergic activity contributed to the development of schizophrenia. Post-

mortem studies have provided support for this hypothesis, demonstrating decreased NMDA

receptor densities in individuals with schizophrenia, specifically within the superior frontal

(Sokolov, 1998) and temporal cortices (Humphries, Mortimer, Hirsch, & de Belleroche,

Page 19: Evaluating The Effects of Cannabis Dependence and ...

6  

1996). Furthermore, recent neuroimaging studies have revealed that along with psychotic-

like symptoms, ketamine administration in healthy individuals led to abnormal cortical

functional connectivity and activation (Driesen, McCarthy, Bhagwagar, Bloch, Calhoun,

D'Souza, Gueorguieva, He, Ramachandran, et al., 2013; Driesen, McCarthy, Bhagwagar,

Bloch, Calhoun, D'Souza, Gueorguieva, He, Leung, et al., 2013).

Taken together, this extensive clinical and preclinical research suggests that

widespread neurotransmitter dysfunction underlies the pathophysiology of schizophrenia.

These diverse set of findings highlight the fact that no one neurotransmitter alone can

account for this complex brain disorder; as such, symptoms of schizophrenia likely arise

from the dysfunctions within each of these neurotransmitter systems. It is clear however that

many unknowns still exist regarding the pathophysiology and etiology of this disorder and

continued research is necessary.

1.2 Cannabis

Cannabis, derived from the Cannabis sativa, Cannabis indica, or Cannabis ruderalis

plant, has been used for centuries for its psychoactive and medicinal properties. Cannabis is

most often used in the form of marijuana, a mixture of cut, dried, and ground flowers, leaves,

and stems. Less common preparations include kief, a powder sifted from the leaves and

flowers of the cannabis plant, and hashish, which is a concentrated resin formed from pressed

kief. The primary psychoactive component in cannabis is delta-9-tetrahydrocannabinol

(THC); however herbal cannabis contains over 400 additional chemical components and 60

cannabinoids including cannabidiol (CBD), cannabigerol and cannabinol (Iversen, 2008).

These cannabinoid concentrations vary significantly among the different strains of marijuana.

For example, cannabis sativa has been shown to contain high concentrations of THC, thus is

Page 20: Evaluating The Effects of Cannabis Dependence and ...

7  

known for its psychoactive side effects, whereas cannabis indica is prepared from high CBD

concentrations and thus is used for its sedative effects (Joy, 1999). Furthermore, cannabis

potency (i.e., THC concentrations) varies substantially between its preparations. Cannabis or

marijuana typically contains 5% THC, whereas resin and cannabis oils can contain THC

concentrations as high as 20 and 60% respectively (United Nations Office on Drugs and

Crime, 2009). In contrast, there is currently no consistent or reliable information concerning

the concentrations of other cannabinoids including CBD, among the various cannabis strains.

Next to tobacco and alcohol, cannabis is the third most commonly used recreational

drug (Iversen, 2003) and the single most commonly used illicit drug with approximately 150

million users worldwide (Cohen, Solowij, & Carr, 2008). It has been reported that 3.3 to

4.4% of the world’s population between the ages of 15-64 have used cannabis at least once in

their lifetime (United Nations Office on Drugs and Crime, 2009). Among 20,000 individuals

surveyed in North America, over 4% displayed signs of cannabis abuse and nearly 60% of

these cases met criteria for cannabis dependence (Hall, Solowij, & Lemon, 1994). More

specifically, in Canada, lifetime use has increased significantly from 23.2% in 1989 to 44.5%

in 2004, with approximately 3% of Canadians reporting daily use (Adlaf, 2005).

1.2.1 Pharmacokinetics of Cannabis

The most common and most efficient method of cannabis administration is smoking.

Cannabis is typically smoked as a joint or blunt, which differ in terms of the papers used to

wrap the cannabis. Joints typically contain anywhere from 0.25 to 1 gram of cannabis and

may be supplemented with tobacco, which aids in its burning. Additional methods of

smoking include pipes or bongs as well as vaporizers. Vaporizers have become more popular

Page 21: Evaluating The Effects of Cannabis Dependence and ...

8  

in recent years, as these devices are able to extract THC and other cannabinoids at

temperatures lower than burning, thus eliminating the harmful secondary carcinogenic tars

and carbon monoxide associated with smoking. Finally, cannabis may be consumed orally

through “edibles”, which are typically used medicinally. Given that cannabis is hydrophobic,

THC must be extracted using lipids or alcohol in order to preserve its psychoactive properties

(Wolff & Johnston, 2014).

The pharmacokinetics of cannabis vary with respect to the route of administration.

Regardless of the method of consumption, the high lipid-solubility of cannabis aids in its

ability to quickly cross the blood-brain barrier. Cannabinoids accumulate in fatty tissue,

yielding a long elimination half-life and persisting in the body for extended periods of time.

THC is rapidly absorbed through the lungs following inhalation and is detectable in blood

plasma within seconds. The psychotropic outcomes reach their maximum effect within 10

minutes, and can last up to 2-3 hours (Huestis, Henningfield, & Cone, 1992). The

bioavailability varies in regards to depth of inhibition, breathhold, and puff duration.

Additionally, THC may be lost in the butt of the joint, through sidestream smoke or through

incomplete lung absorption (Grotenhermen, 2003). Furthermore, oral administration

introduces more variability as well as delayed or diminished effects by up to 30%, due to the

slower rate of absorption of THC from the digestive tract (Perez-Reyes et al., 1991).

THC metabolism takes place mainly in the liver, and to a lesser extent in the lungs

and heart (Harvey, 1976). Initial hydroxylation results in a short-lived, potent psychoactive

THC metabolite. Further oxidation in the liver converts 11-OH-THC to several inactive

metabolites. To date, close to 100 metabolites have been discovered for THC (Harvey &

Brown, 1991), including THC-COOH, the most abundant non-psychoactive metabolite in

Page 22: Evaluating The Effects of Cannabis Dependence and ...

9  

plasma and urine (Adams & Martin, 1996; Hawksworth & McArdle, 2004). The exact

elimination half-life of THC measured from plasma has proven to be difficult to calculate,

due to variability in the dose, route of administration, period of observation and experience of

the user. THC plasma elimination half-lives have been reported to range anywhere from 20

hours (Hunt & Jones, 1980) to up to 12 days (Johansson, Halldin, Agurell, Hollister, &

Gillespie, 1989). A plausible explanation for this divergence may lie within the ability to

distinguish THC from its other, longer lasting metabolites (Grotenhermen, 2003). Complete

elimination has been shown to take as long as 12.9 days (ranging from 3-29 days) in light

users to 31.5 days in chronic, heavy users (Ellis, Mann, Judson, Schramm, & Tashchian,

1985). Given the long elimination half-life, repeated cannabis use among chronic users leads

to the accumulation of cannabinoids in the body and brain (Ashton, 1999), and has been

suggested to lead to a form of “reverse tolerance” (Julien, 2001). However, there is no clear

and consistent relationship between the plasma THC levels and behavioural effects (Agurell

et al., 1986).

1.2.2 Acute Psychoactive Effects of Cannabis

The use of cannabis for its psychoactive and medicinal qualities dates back centuries

(Zuardi, 2006). The psychoactive effects of cannabis are subjective and vary based on the

experience of the individual, method of administration, dose and strain of cannabis. Cannabis

use produces a variety of psychological and physiological effects; unlike other psychoactive

substances, cannabis exhibits properties of hallucinogens, stimulants and depressants.

Generally, mild cannabis intoxication acutely leads to euphoria, lethargy, perceptual and time

distortions, and the overall augmentation of ordinary sensory experiences (Tart, 1970). At

higher doses, side effects include short-term memory impairment, intense mood alterations,

Page 23: Evaluating The Effects of Cannabis Dependence and ...

10  

and impaired motor coordination (Jaffe, 1985). While many individuals report anxiolytic

effects following cannabis consumption, at higher doses, increased anxiety and panic

reactions including paranoia and acute toxic psychosis are commonly reported (Ashton,

1999; Iversen, 2008).

1.2.3 Somatic and Health Effects of Cannabis

In addition to the psychoactive effects, cannabis produces a variety of acute somatic

effects. While these side effects may only last for a short period of time, research has begun

to uncover the long-term health outcomes associated with chronic cannabis use. For example,

cannabis has been shown to lead to tachycardia, which has been suggested to accelerate the

development of heart problems in at risk individuals. In support of this, studies have shown

that the risk of myocardial infarction during the first hour after cannabis consumption

increased 4.8-fold as compared to non-users (Hartung, Kauferstein, Ritz-Timme, & Daldrup,

2014; Mittleman, Lewis, Maclure, Sherwood, & Muller, 2001). Furthermore, research

regarding the bronchopulmonary side effects of cannabis use have been reliably

demonstrated. These effects include chronic inflammatory changes in the respiratory tract,

which can manifest as chronic cough, wheezing, and phlegm (Van Hoozen & Cross, 1997).

Additional adverse health effects include cancer, bronchitis and emphysema (Ashton, 1999).

Finally, acute somatic effects of cannabis use include ocular redness, dry mouth and

increased appetite commonly referred to as "the munchies" (Sansone & Sansone, 2014). It

should be noted that the acute toxicity of cannabis is extremely low, as there have been no

reported cases of human deaths directly from cannabis poisoning (Hall et al., 1994).

Page 24: Evaluating The Effects of Cannabis Dependence and ...

11  

1.2.4 Cognitive Effects of Cannabis

There is a growing body of research investigating the impairing effects associated

with cannabis use on cognition. Interestingly, comparisons have been drawn between

impairments in perceptual, emotional, and cognitive domains accompanying cannabis use,

and those seen in patients with schizophrenia (Radhakrishnan, Wilkinson, & D'Souza, 2014;

Solowij & Michie, 2007). For example, studies have found that acute cannabis use impairs

features of decision-making and planning including response times and accuracy (Ramaekers

et al., 2006; Vadhan et al., 2007). Moreover, cannabis intoxication is associated with

increased risk-taking through tasks assessing impulsivity and inhibition (McDonald,

Schleifer, Richards, & de Wit, 2003). However, evidence regarding the impairing effects on

attention and concentration is mixed, whereby the impairing effects seem to be stronger in

less frequent users. In contrast, research suggests that attention in the chronic users may be

disrupted more so by acute abstinence than intoxication (Crean, Crane, & Mason, 2011).

These disparate findings may be due in part to the variability in study methodology,

specifically associated with frequency and recency of cannabis use.

The long-term effects of cannabis use have received significant attention in recent

years. Yet similarly to the findings regarding the acute effects, inconsistencies in the

literature exist. Several studies have shown that cannabis appears to have continued

impairing effects on executive function following a prolonged abstinence period. The most

enduring deficits seem to lie within the domains of decision-making, concept formation, and

planning (Verdejo-Garcia, Rivas-Perez, Lopez-Torrecillas, & Perez-Garcia, 2006). However,

contrasting research has shown that cognitive deficits seen in long-term cannabis users may

be eliminated following 28-day abstinence (Pope, Gruber, Hudson, Huestis, & Yurgelun-

Page 25: Evaluating The Effects of Cannabis Dependence and ...

12  

Todd, 2001). The reversibility of these deficits indicates that chronic cannabis use may not

permanently alter cognitive function in all domains.

1.2.5 Treatment of Cannabis Dependence

Treatment approaches targeting cannabis dependence have been adapted in a large

part from alcohol use disorder interventions. However, compared to these better-established

interventions, evidence-base research regarding the management of cannabis dependence is

still somewhat lacking. In terms of behavioural interventions, motivational enhancement

therapy (MET) and cognitive-behavioural therapy (CBT) have proven to be the most reliable

and successful treatment approaches. MET focuses on resolving the users ambivalence about

engaging in treatment and aims to enhance their motivation to change. In contrast, CBT

addresses the skills necessary to quit or reduce ones substance use and change unhelpful

thinking and behaviours that may impact treatment outcomes. Randomized control trials

validating MET and CBT suggest that multicomponent therapy combining these treatments is

most effective (Dennis et al., 2004; Stephens, Roffman, & Curtin, 2000).

A promising addition to these behavioural interventions is contingency management

(Budney, Moore, Rocha, & Higgins, 2006; Kadden, Litt, Kabela-Cormier, & Petry, 2007).

Current literature has provided support for the efficacy of contingency management in

treating cannabis dependence in otherwise healthy individuals (Budney, Higgins,

Radonovich, & Novy, 2000) and those with severe mental illness (Sigmon, Steingard,

Badger, Anthony, & Higgins, 2000). Contingency management promotes behavioural

changes through the use of positive or negative contingencies in an organized manner. In

regards to substance use interventions, contingency management encourages behaviours

Page 26: Evaluating The Effects of Cannabis Dependence and ...

13  

associated with treatment attendance and retention as well as decreasing drug use behaviours.

Importantly, this method has proven to be effective in populations with low motivation for

changing drug use behaviours (Sinha, Easton, Renee-Aubin, & Carroll, 2003).

Finally, there is a growing body of support for the use of pharmacotherapy targeting

withdrawal symptoms associated with cannabis abstinence. However, this work is still in its

early stages, as no government-approved medications currently exist. Furthermore, few

studies have focused specifically on the physiological, subjective, and reinforcing effects of

these medications. Preclinical evidence suggests that Δ9-THC, clonidine and lithium can

effectively decrease withdrawal behaviours in rodents (Cui et al., 2001). In humans,

nabiximols or Sativex, a cannabis extract with a 1:1 mixture of THC and CBD, was

originally developed as a treatment for multiple sclerosis. However, interestingly, this

medication has been shown to attenuate cannabis withdrawal symptoms. For example, in

a treatment-seeking cohort, nabiximols reduced withdrawal symptoms and improved

treatment retention. However, this medication did not effectively promote the long-term

reductions in cannabis use following medication cessation (Allsop et al., 2014). These

preliminary findings support the potential treatment efficacy of pharmacotherapy and

highlight the need for continued research investigating the combined effects of behavioural

and pharmacological interventions.

1.3 The Endogenous Cannabinoid System

The endocannabinoid (eCB) system is one of the most abundant neuromodulatory

systems in the brain. This system is responsible for maintaining homeostasis within the CNS

through its control over synaptic transmission (Pertwee, 2008). It is made up of two G-

protein-coupled receptors, cannabinoid type 1 (CB1) and cannabinoid type 2 (CB2). The

Page 27: Evaluating The Effects of Cannabis Dependence and ...

14  

discovery of the type 1 receptor led to the identification of natural occurring ligands or

endocannabinoids, anandamide and 2-arachidonoylglycerol (2-AG). These endocannabinoids

act at CB1 receptors and are synthesized in response to increased intracellular calcium levels

(Howlett et al 2002). Anandamide behaves similarly to THC, yet with less potent and

pervasive actions (Iversen, 2008), acting as a partial agonist at both CB1 and CB2 receptors.

In contrast, 2-AG is a complete agonist at both receptor subtypes. Cannabis, prepared in all

of its forms, cannabis agonists, and endogenous ligands, act primarily on CB1 receptors.

While the two eCB system receptor subtypes are closely related, type 2 receptors are most

often found on immune cells, and are involved in the immunological activity of leukocytes

(Galiegue et al., 1995).

The complex interaction between CB1 receptors and neurotransmitter systems are

thought to mediate the principal effects of cannabis (Freund, Katona, & Piomelli, 2003). CB1

receptors are primarily located on central and peripheral neurons (Tsou, Brown, Sanudo-

Pena, Mackie, & Walker, 1998) and are highly expressed throughout the brain. These brain

regions, including, the hippocampus, cortex, cerebellum, basal ganglia, PFC, and anterior

cingulate, are thought to be associated with cognition and mood regulation (Herkenham,

1991).

At the molecular level, CB1 receptors are localized presynaptically on excitatory and

inhibitory neurons, primarily on the axon terminals of GABA interneurons. These receptors

are thought to mediate inhibition and thus maintain homeostasis by preventing excessive

release of excitatory neurotransmission within the CNS (Eggan & Lewis, 2007). Evidence

suggests that endocannabinoids act as retrograde signal messengers, whereby increases in

postsynaptic intracellular calcium stimulated by specific neurotransmitters can trigger the

Page 28: Evaluating The Effects of Cannabis Dependence and ...

15  

synthesis and release of endocannabinoids. These molecules are then thought to activate

presynaptic CB1 receptors and inhibit the further release of GABA (Pertwee, 2008; Szabo &

Schlicker, 2005).

CB1 activation through THC and other psychoactive cannabinoids enhances

mesolimbic dopaminergic neurotransmission, which terminates at the PFC (Pistis et al.,

2002), striatum (Bossong et al., 2009), and nucleus accumbens (Gardner, 2005). Converging

clinical and preclinical research has implicated this increase in dopamine in both the

cognitive impairments (Pistis et al., 2002; Pistis, Porcu, Melis, Diana, & Gessa, 2001) and

reinforcing properties associated with cannabis use (Ameri, 1999; Bossong et al., 2009). The

divergent effects of cannabis on GABA and dopamine may also underlie the subjective

opposing stimulant and depressant effects experienced by individual users.

Similarly, the cannabinoids, specifically THC and CBD, have also been shown to

possess divergent properties. THC acts as a partial CB1 receptor agonist with relatively low

receptor efficacy (Compton, Johnson, Melvin, & Martin, 1992; Pertwee, 1997). In contrast,

CBD indirectly stimulates endocannabinoid signalling through supressing fatty acid amide

hydrolase (FAAH), an enzyme involved in the inactivation of endogenous cannabinoids

(Piomelli, Beltramo, Giuffrida, & Stella, 1998). The molecular mechanism underlying CBD

activity is not well understood, however it has been suggested that CBD has the ability to

non-competitively antagonized CB1 and CB2 receptors (Murray, Morrison, Henquet, & Di

Forti, 2007; Thomas et al., 2007). In this capacity, CBD has been shown to influence the

pharmacological activity of THC, thus countering some of the undesirable side effects

associated with THC administration (Englund et al., 2013; Zuardi, Crippa, Hallak, Moreira,

& Guimaraes, 2006). This opposing molecule activity is reflected in the behavioural effects

Page 29: Evaluating The Effects of Cannabis Dependence and ...

16  

of THC and CBD. For example, THC is known to induce paranoia in individuals with prior

paranoid ideation (Freeman et al., 2015) as well as exacerbate pre-existing psychotic and

affective symptoms in those diagnosed with schizophrenia (D'Souza et al., 2005). In contrast,

CBD has been shown to possess antidepressant (Zanelati, Biojone, Moreira, Guimaraes, &

Joca, 2010), anxiolytic (Bergamaschi et al., 2011; Zuardi, Shirakawa, Finkelfarb, & Karniol,

1982) and neuroprotective properties (Hayakawa et al., 2007).

1.4 Cannabis Use in Schizophrenia

Recent research has focused on cannabis use among individuals with schizophrenia

due in part to its worldwide standing as the most commonly used illicit substance in this

population. With approximately one-third of persons with schizophrenia and other psychoses

reporting daily use (Jablensky, 2000) and one-quarter meeting criteria for a cannabis use

disorders (CUD) (Koskinen, Lohonen, Koponen, Isohanni, & Miettunen, 2010), it is clear

that understanding the molecular and behavioural effects of cannabis use in this population is

essential.

1.4.1 Effects on Prognosis and Course

The presence of co-morbid cannabis use in schizophrenia is associated with

devastating effects on prognosis and clinical severity. These individuals face symptom

exacerbation, higher rates of relapse and poorer treatment compliance and success (D'Souza

et al., 2005; Foti, Kotov, Guey, & Bromet, 2010; Linszen, Dingemans, & Lenior, 1994;

Manrique-Garcia et al., 2014). Interestingly, studies that employ self-report measures

generally find that patients use cannabis to relieve boredom, provide stimulation to feel good,

to get high, or to relax and socialize with peers (Addington & Duchak, 1997; Fowler, Carr,

Page 30: Evaluating The Effects of Cannabis Dependence and ...

17  

Carter, & Lewin, 1998; Goswami, Mattoo, Basu, & Singh, 2004). Of note, these self-report

findings are susceptible to denial and rationalization. Yet these findings suggest that many

patients are unaware of or may downplay the clear detrimental effects of cannabis use

(McGee, Williams, Poulton, & Moffitt, 2000).

1.4.2 Effects on Cognition

Given the evident harmful effects of cannabis on the clinical course, it would follow

that cannabis may have equally detrimental effects of cognitive functioning; however, current

literature provides contradictory findings. A recent meta-analysis including 10 studies and

572 patients with schizophrenia with and without cannabis use, found that those with a

history of cannabis use demonstrated better cognitive functioning compared to non-users.

Furthermore, in the second study included in this meta-analysis, cannabis using first episode

patients with schizophrenia performed better on several domains including visual memory,

working memory and executive function when compared to first episode non-using patients

(Yucel et al., 2012). Similar results were demonstrated in another meta-analysis focusing

more specifically on the effects of cannabis by controlling for confounding poly substance

use (Rabin, Zakzanis, & George, 2011). Researchers have suggested that these findings may

be attributed to the neurochemical effects of cannabis (Coulston, Perdices, Henderson, &

Gin, 2011), given that the neuronal pathways regulated by the eCB system are thought to

underlie neurocognitive functioning (Gerdeman, Partridge, Lupica, & Lovinger, 2003).  

Alternatively, others have proposed that these cannabis-using patients may represent a

subgroup of patients with better premorbid cognitive functioning and social skills (Dixon,

Haas, Weiden, Sweeney, & Frances, 1991; Leeson, Harrison, Ron, Barnes, & Joyce, 2011;

Rodriguez-Sanchez et al., 2010). As such, individuals in the subgroup are thought to possess

Page 31: Evaluating The Effects of Cannabis Dependence and ...

18  

a potentially less severe form of the disorder, developing psychosis following the initiation of

cannabis use (Yucel et al., 2012).

In contrast, several studies have demonstrated that cannabis use may further impair

cognitive performance in patients with schizophrenia. This finding has been replicated in

studies employing lab-based (D'Souza et al., 2005), cross-sectional (Ringen et al., 2010), and

longitudinal approaches (Mata et al., 2008). Finally, several studies have been unable to

show that this comorbidity has a significant or consequential effect on cognition (Jockers-

Scherubl et al., 2007; Sevy et al., 2007). These divergent findings may be attributed in part,

to a lack of control for confounding variables within studies as well as methodological

discrepancies between studies.

1.4.3 Underlying Neurobiological Connection

The once popular hypothesis of ‘self-medication’ suggested the psychiatric illnesses

put those at risk for later cannabis use as a means to ‘self-medicate’ and alleviate unpleasant

symptoms of the disorder and medication side effects (Wittchen et al., 2007; Musty &

Kaback, 1995). However, more recent evidence has shown that self-medication does not

adequately account for the pattern of cannabis use among patients with schizophrenia

(Kolliakou et al., 2015). As such, researchers have begun to investigate other potential

explanations for this common co-morbidity.

In spite of its prevalence, many unknowns still exist regarding the neurophysiological

impact of cannabis use among individuals with schizophrenia. Aberrant eCB system

functioning has been implicated in the detrimental effects of cannabis use on the course of

illness. Genetic variants of cannabinoid-related genes, including the cannabinoid receptor

Page 32: Evaluating The Effects of Cannabis Dependence and ...

19  

gene (CNR1), represent likely candidates underlying the interaction between cannabis use

and schizophrenia (Martinez-Gras et al., 2006). Furthermore, disturbances in the eCB system

have been demonstrated through elevated anandamide levels in the cerebrospinal fluid in

both prodromal and untreated patients with schizophrenia (De Marchi et al., 2003; Koethe et

al., 2009). Additionally, increases in cannabinoid type 1 receptor (CB1R) densities have been

observed in this population (Ceccarini et al., 2013; Dean, Sundram, Bradbury, Scarr, &

Copolov, 2001).

The GABAergic system has also been implicated in this co-morbidity. Aberrant

GABA functioning seen in patients with schizophrenia may be further exacerbated by the

inhibitory influence of cannabis on this neurotransmitter. In fact, it has been suggested the

deficits in GABAergic activity in patients, may increase the likelihood of engaging in

cannabis use (Rabin, 2014). Evidence supporting this suggestion comes from the overlapping

influence of GABA on cognitive function both in individuals with schizophrenia and

cannabis users (Benes, 1998; Di Lazzaro, Ziemann, & Lemon, 2008; Hill, Froc, Fox,

Gorzalka, & Christie, 2004).

1.5 Cortical Inhibition

1.5.1 Indexing Cortical Inhibition using Transcranial Magnetic

Stimulation

Cortical inhibition refers to the neurophysiological process whereby GABA

inhibitory interneurons selectively attenuate the activity of pyramidal neurons in the cortex.

One technique used to evaluate cortical inhibition is transcranial magnetic stimulation

(TMS). TMS was developed as a non-invasive technique used to assess the function of the

corticospinal tract in vivo (Kobayashi & Pascual-Leone, 2003). TMS is based on the concept

Page 33: Evaluating The Effects of Cannabis Dependence and ...

20  

of electromagnetic induction as outlined by Faraday’s Law, whereby an intense current

pulses within the coil, producing a magnetic field orthogonal to the plane of the coil. This

field passes unimpeded through the skull and in turn induces an electric field tangential to the

skull and in an opposite direction to that of the current in the coil. The horizontal orientation

of the current favorably activates horizontally-oriented interneurons. Thus, pyramidal

neurons are activated transynaptically (Rothwell & Rosenkranz, 2005).

Clinicians and researchers have used TMS for both therapeutic and diagnostic

applications. In terms of its diagnostic ability, combining TMS with electromyography

(EMG) allows for the exploration of cortical GABAergic function. Stimulation in the motor

cortex by a single TMS pulse produces a corresponding contralateral motor evoked potential

(MEP). The ultimate effect of the stimulation depends on several important factors, including

the shape of the magnetic coil, the intensity of the current pulse driven through the coil and

the interstimulus interval separating the various pulses. Commonly used stimulation

parameters include the focal figure-of-eight coil with stimulation intensities corresponding to

the 1 millivolt (mV), i.e. the intensity needed to produce a MEP of 1 mV peak to peak

amplitude. These TMS paradigms have been shown to reliably assess the activity of the

GABAA and GABAB receptor functioning in the motor cortex in human subjects (Chen,

2000; Fitzgerald et al, 2002a).

More specifically, paired pulse TMS (ppTMS) has been used to reliably assess both

cortical inhibition and excitation and thus the function of several neurotransmitter systems.

For example, cortical excitation can be indexed through the resting motor threshold (RMT)

and intracortical facilitation (ICF). The RMT is defined as the minimal intensity that

produces a MEP of >50 μV in 5 of 10 trials in the relaxed muscle of interest. In contrast, ICF

Page 34: Evaluating The Effects of Cannabis Dependence and ...

21  

is measured by administering a subthreshold pulse prior to the onset of the test pulse by 7 to

20 milliseconds (Kujirai et al., 1993; Ziemann, Rothwell, & Ridding, 1996). There is strong

evidence to suggest that cortical facilitation is mediated in part, by glutamatergic

neurotransmission (Clark, Randall, & Collingridge, 1994).

Cortical inhibition is also assessed through paired pulse paradigms, differing

primarily in interstimulus intervals. For example, short-interval intracortical inhibition

(SICI), a measure of GABAA activity, is measured by administering a subthreshold pulse

proceeded by a suprathreshold test pulse within 1 to 5 milliseconds. GABAB in contrast, can

be measured through two different paradigms. The first is a ppTMS called long interval

cortical inhibition (LICI), which utilizes two suprathreshold stimuli at interstimulus intervals

ranging from 100 to 200 milliseconds. The second paradigm, cortical silent period (CSP),

incorporates motor cortical stimulation superimposed on background EMG activity resulting

in a cessation of EMG activity. Thus the duration of this silent period represents an index of

GABAB activity (Cantello, Gianelli, Civardi, & Mutani, 1992).

Evidence regarding the involvement of GABAA and GABAB underlying SICI and

LICI, respectively, comes from the time course of effects associated with these

neurotransmitters. Cortical stimulation has been shown to produce disynaptic slow and fast

inhibitory postsynaptic potentials (IPSPs) (Davies, Davies, & Collingridge, 1990). These

fast IPSPs are thought to be mediated by GABAA activity as they last approximately 20 ms,

whereas slow IPSPs, mediated by GABAB receptors, peak around 150–200 ms. This time

course is reflected in the interstimulus intervals associated with SICI (1-5 ms) and LICI (100-

200)(Sanger, Garg, & Chen, 2001). Additional support from pharmacological studies has

revealed that administration of GABAA receptor agonists, specifically lorazepam, reliably

Page 35: Evaluating The Effects of Cannabis Dependence and ...

22  

enhance SICI whereas baclofen, a GABAB receptor agonist prolongs CSP (Di Lazzaro et al.,

2008).

1.5.2 Cortical Inhibitory Deficits in Schizophrenia

While TMS has provided a reliable index of cortical inhibition and excitation in the

motor cortex in healthy controls (Chen, 2000) its more important use has been to better

understand changes in cortical functioning in individuals with mental illness (Daskalakis,

Christensen, Chen, et al., 2002; Di Lazzaro et al., 2004; Maeda, Keenan, & Pascual-Leone,

2000). Aberrant cortical inhibition has been implicated in the pathophysiology of

schizophrenia (Lewis, Pierri, Volk, Melchitzky, & Woo, 1999). Many early studies

investigating the physiological impact of schizophrenia in the motor cortex have reliably

demonstrated dysfunctional inhibition, yet their initial understanding of the mechanisms

associated with these deficits was limited (Davey, Puri, Lewis, Lewis, & Ellaway, 1997;

Puri, Davey, Ellaway, & Lewis, 1996).

Advances in our understanding of the specific neurotransmitters indexed through each

TMS paradigm has allowed for a better understanding of the neurotransmitter deficits

underlying schizophrenia as well as the impact of antipsychotic medication on these deficits.

For example, Daskalakis and colleagues compared the RMT, CSP, SICI and ICF in 15

unmedicated patients with schizophrenia, 15 medicated patients with schizophrenia, and 15

healthy controls. The researchers found that the unmedicated patients exhibited the most

profound cortical deficits on measures of CSP and SICI. Additionally, fewer deficits were

observed among the medicated group when compared to the healthy controls (Daskalakis,

Christensen, Chen, et al., 2002). These findings have been replicated in first episode patients

as well as those with chronic schizophrenia (Fitzgerald, Brown, Daskalakis, & Kulkarni,

Page 36: Evaluating The Effects of Cannabis Dependence and ...

23  

2002a; Fitzgerald et al., 2003; Wobrock et al., 2009). One of the more reliable and interesting

findings regarding CSP comes from studies investigating the effects of antipsychotic

medication on GABAB-mediated inhibition (Kaster et al., 2015). One such study found a

significant lengthening of the CSP in patients treated with clozapine when compared to both

unmedicated patients and controls (Daskalakis, Christensen, et al., 2008). Of note, a similar

study demonstrated that this CSP finding was clozapine specific, as patients treated with any

other antipsychotic medications failed to demonstrate this lengthened silent period (Liu,

Fitzgerald, Daigle, Chen, & Daskalakis, 2009). These results have significant implications

for the treatment of schizophrenia and highlight the importance of underlying GABAA and

GABAB abnormalities in the pathophysiology of this disorder.

Reduced SICI among individuals with schizophrenia remains the most consistently

replicated finding. These deficits have been demonstrated in patients with varying levels of

illness severity and duration, including those at risk for developing this disorder (Hasan et al.,

2012), first episode patients (Wobrock et al., 2008; Wobrock et al., 2009) and individuals

with chronic schizophrenia (Daskalakis, Christensen, Chen, et al., 2002; Fitzgerald, Brown,

et al., 2002a; Pascual-Leone, Manoach, Birnbaum, & Goff, 2002). In a recent meta-analysis

conducted by Radhu and colleagues (2013), researchers found significant deficits in SICI in

patients with schizophrenia, after controlling for age and medication, using a meta-

regression. Furthermore, this finding showed specificity as a characteristic of schizophrenia,

when compared to both patients with major depression and those with obsessive-compulsive

disorder (Radhu et al., 2013).

However, some studies have been unable to find significant diagnostic differences in

SICI (Daskalakis, Christensen, et al., 2008; Eichhammer et al., 2004; Liu et al., 2009) and

Page 37: Evaluating The Effects of Cannabis Dependence and ...

24  

ICF (Eichhammer et al., 2004; Hasan et al., 2012; Wobrock et al., 2008) when comparing

patients to controls. Finally, there are inconsistencies in the current literature regarding

GABAB-mediated inhibitory control in patients. For example, studies have revealed

decreased (Daskalakis, Christensen, Chen, et al., 2002; Fitzgerald, Brown, Daskalakis,

deCastella, & Kulkarni, 2002) increased (Bajbouj et al., 2004; Soubasi et al., 2010) and no

differences (Davey et al., 1997; Frantseva et al., 2008; Puri et al., 1996) on measures of LICI

and CSP.

Importantly, there is strong evidence to suggest that these cortical inhibitory deficits

are associated with symptoms of schizophrenia. Liu and colleagues found that CSP duration

was significantly inversely associated to negative symptoms, while positive symptoms were

correlated with SICI. This finding was most pronounced in the unmedicated group (Liu et al.,

2009). Thus these findings highlight the differential role that GABAA and GABAB likely

have on the symptoms of schizophrenia. In a related study, researchers investigated the

underlying association between abnormal cortical inhibition and social cognitive deficits.

SICI and LICI and several dimensions of social cognition including, social perception and

emotion processing, were evaluated in 33 unmedicated patients, 21 medicated patients and 45

controls. Inline with previous studies, SICI deficits were observed in the unmedicated group

indicating less inhibitory response. Additionally, among the unmedicated group, deficits in

SICI were associated with poorer scores on the measures of global social cognition and

emotion processing, thus demonstrating the potential contribution of aberrant cortical

inhibition to deficits of executive function (Mehta, Thirthalli, Basavaraju, & Gangadhar,

2014).

Page 38: Evaluating The Effects of Cannabis Dependence and ...

25  

A major limitation to the above-mentioned TMS findings is that such measures were

recorded in the motor cortex. Recent technological advances have allowed for the evaluation

of cortical inhibition directly from the DLPFC through combined TMS with EEG

(Daskalakis, Farzan, et al., 2008; Fitzgerald et al., 2008). Assessing cortical inhibition in the

DLPFC is of particular importance in the field of mental illness research, as this region is

associated with the pathophysiology, and cognitive deficits underlying SCZ and is the region

where CBI receptors are densely populated.

Although this method is fairly novel, its validity and reliability have been well

established in healthy subjects by comparing cortical evoked activity as measured through

TMS-EEG with EMG activity from motor cortical inhibition (Farzan et al., 2010b). Using a

pharmaco-TMS-EEG approach, researchers have been able to study the direct role of

GABAergic neurotransmission through TMS-evoked EEG potentials (Premoli et al., 2014).

Furthermore, this technique allows for the decomposition of the EEG signal into five well-

established frequency bands to evaluate their inhibitory activity. Using this technique in

patients with schizophrenia, previous research has demonstrated that DLPFC gamma

inhibitory activity (30-50 Hz) is selectively impaired in individuals with schizophrenia

compared to patients with bipolar disorder (Farzan et al., 2010b), obsessive-compulsive

disorder and healthy individuals (Radhu et al 2015). This finding, along with additional

evidence of decreased cortical inhibition in schizophrenia, provides further support for the

role of the possibility that altered cortical connectivity may underlie the pathophysiology of

schizophrenia (Frantseva et al., 2014).

Page 39: Evaluating The Effects of Cannabis Dependence and ...

26  

1.5.3 Cortical Inhibitory Deficits with Cannabis Use

There is currently a paucity of research investigating the functional effects of

cannabis use on cortical inhibition. Only one study has sought to determine whether

individuals with chronic cannabis use demonstrate abnormalities in cortical inhibition.

Fitzgerald et al (2009) examined the level of use (heavy versus light users) in forty-two

cannabis users compared to 19 non-using controls on measures of cortical inhibition and

excitation. Heavy cannabis use was defined as those who used 7 or more times a week and

had consumed within 48 hours prior to testing. In contrast, light users were those who had

used between 1 to 4 times a week, and had used sometime within the last 7 days of testing

(Block & Ghoneim, 1993). TMS paradigms included SICI, ICF, LICI and CSP and THC

plasma levels were measured prior to the TMS procedure. The researchers found that both

heavy and light cannabis users demonstrated reductions in SICI compared to healthy

controls, with no other differences on the additional measures of inhibition or excitation.

Given that both heavy and light users exhibited comparable SICI deficits, the researchers

suggest that this finding is likely dependent on long term neurobiological changes associated

with lifetime cannabis use rather than the direct effect of cannabis on GABAA activity. Thus,

long-term cannabis use likely results in the down regulation of cortical inhibitory activity

(Fitzgerald, Williams, & Daskalakis, 2009).

1.5.4 The Combined Effects of Cannabis and Schizophrenia on

Cortical Inhibition

To date, only one study has investigated the effects of cannabis use on cortical

inhibition in patients with schizophrenia. Twelve first-episode patients with a history of co-

morbid cannabis abuse and 17 first-episode patients with no history of cannabis use

Page 40: Evaluating The Effects of Cannabis Dependence and ...

27  

completed several TMS motor cortical paradigms including RMT, SICI, ICF, and CSP.

Researchers found that stimulation to the right motor cortex in the cannabis-using group

evoked lower cortical inhibition through SICI and enhanced facilitation as measured by ICF.

In line with Fitzgerald et al. (2009), no group differences were observed in RMT or CSP

duration. Interestingly, there were no significant correlations between psychopathology,

disease characteristics, or the extent of cannabis abuse, with TMS parameters. Researchers

suggested that their results likely reflect the ability of cannabis to exacerbate reduced cortical

inhibition and enhanced facilitation in patients with schizophrenia (Wobrock et al., 2010). It

is clear that continued research is needed to verify these results and determine if these same

findings can be replicated in individuals with varying illness severity and duration.

1.6 Working Memory

1.6.1 The Neurophysiology of Working Memory

A key component of cognition that has garnered significant attention is working

memory. Working memory is commonly defined by the ability to maintain and manipulate

information for short periods of time (Baddeley & Hitch, 1974b; Barak & Tsodyks, 2014).

As a higher order cognitive function, working memory is thought to be central to, and

interact with several additional cognitive domains including learning, comprehension and

reasoning (D'Esposito, 2007). As a theoretical construct, working memory has been defined

through multiple models, based primarily on the discipline it is associated with, including

cognitive psychology and cognitive neuroscience (Baddeley, 2003). One of the most

significant contributions to the field was put forth by Baddeley and Hitch, who in 1974,

described a model that combined the ability to both mediate executive control and maintain

temporarily stored information (Baddeley & Hitch, 1974a).

Page 41: Evaluating The Effects of Cannabis Dependence and ...

28  

Current working memory models came about following early preclinical

observations, which guided their understanding of the brain regions associated with this

cognitive domain. In 1936, Carlyle Jacobsen proposed that the PFC was integral to working

memory functioning, following the observation that damage to this area in primates led to

significant memory deficits. Interestingly, he found that the monkeys could complete

complex cognitive tasks as long as the stimulus of interest remained present. However, the

introduction of a delay period or the removal of the stimulus significantly decreased

performance to chance (Jacobsen, 1936). Later work conducted by Joaquin Fuster supported

this initial observation, whereby prefrontal neurons in monkeys were found to fire most

rapidly during the delay period (Fuster, 1973). These early findings posed as the foundation

for later working memory models, which suggest that the PFC contributes to working

memory by filtering out extraneous information as well as maintaining and organizing key

information in order to aid in goal directed behaviours (Cohen et al., 1997).

Methodological advancements have revealed that working memory function is not

based solely on the PFC, but rather a complex network spanning frontal and parietal areas

(Honey et al., 2002). However, there still exists debate regarding the role of each brain

region. It has been suggested that the PFC may be involved in attentional activation, selecting

strategies, and the manipulation of memory, where as the posterior areas are thought to play a

role in the maintenance of information (Curtis & D'Esposito, 2003; Postle, 2006). However,

neuroimaging and position emission tomography (PET) studies have provided a different

method of categorization suggesting that the PFC may be associated with more specific types

of memory, for example verbal working memory (Jonides et al., 1998; Smith, Jonides,

Marshuetz, & Koeppe, 1998) Such lack of consensus regarding the functional brain regions

Page 42: Evaluating The Effects of Cannabis Dependence and ...

29  

associated with working memory may be attributed to the type of working memory assessed

or task difficulty.

Further complicating our understanding of the physiology of working memory lies

within the complex coordination between excitatory and inhibitory neurotransmitter circuits.

Goldman-Rakic and others have shown that neuronal prefrontal circuits aid in informational

maintenance through recurrent excitatory glutamate outputs that fire throughout the delay

period (Goldman-Rakic, 1995). These circuits are thought to be “tuned” by inhibitory

GABAergic interneurons (Rao, Williams, & Goldman-Rakic, 2000). Thus GABA is believed

to synchronize neuronal activity during working memory processes (Lewis, Hashimoto, &

Volk, 2005). In support of this, Sawaguchi et al (1989) injected bicuculline, a GABA

antagonist, into the DLPFC of monkeys, which lead to disrupted working memory

performance (Sawaguchi, Matsumura, & Kubota, 1989). However, this finding is not specific

to GABA; rather, research suggests that the coordination of several key neurotransmitter

systems are necessary for optimal working memory performance (Robbins & Arnsten, 2009).

1.6.2 Indexing Working Memory

1.6.2.1 Neurocognitive Assessments

There are numerous well-established neuropsychological batteries used to assess

working memory, which allow for the assessment of several memory domains both verbal

and non-verbal (e.g., visuospatial, and audiospatial). Common non-verbal tasks include the

delayed match to sample task and the spatial delay response. These non-verbal tasks are

generally comprised of a brief presentation of a stimulus followed by a delay period. The

participant is then required to either determine the original position of the stimulus (i.e.,

Page 43: Evaluating The Effects of Cannabis Dependence and ...

30  

spatial delay response) or determine if an additional stimulus presented is the same as the

initial one (i.e. delayed match to sample). In contrast, common verbal tasks include the

number-letter-span, the Sternberg task and the N-back task. These tasks require participants

to either maintain and manipulate a string of numbers or letters, through repetition and

reordering (i.e., number-letter-span) or determine if the current letter/number presented was

included in the original string (i.e. the Sternberg task). Finally, in the N-back task, letters are

presented sequentially and participants are asked to determine if the same letter was

presented N (i.e. 0-3) trials back, where 0 represents a control condition measuring reaction

time only. Importantly, the N-back requires continuous updating with each trial,

differentiating itself from other working memory tasks. Furthermore, the N-back task is

commonly used in neurophysiologic-based studies given that it requires minimal movement

for responses, thus decreasing the amount of muscle movement noise in the data.

1.6.2.2 Neurophysiological Assessments

Advances in brain imaging and recording techniques have provided novel insight into

the neurophysiological underpinnings associated with working memory.

Electroencephalography (EEG) is a commonly used technique, due to its high temporal

resolution, ease of use, and low costs. This technique records brain activity from electrodes

placed on the scalp, thus measuring a summation of synaptic activity from thousand of

neurons in a similar spatial orientation from the outer layers of the brain. EEG is able to

capture oscillatory activity in five discrete frequency bands including delta (1-4 Hz), theta (4-

7 Hz), alpha (8-12 Hz), beta (12-30 Hz), and gamma (30-50 Hz). Each oscillatory frequency

is associated with specific brain states. For example, higher frequencies have been shown to

be associated with the awake brain including cognitive tasks such as working memory, while

Page 44: Evaluating The Effects of Cannabis Dependence and ...

31  

lower oscillatory activities are associated with the different stages of sleep (Buzsaki, 2006;

Buzsaki & Draguhn, 2004). Synchrony among large neuronal networks is thought to be a

critical component of healthy cognitive function (Engel, Fries, & Singer, 2001; Uhlhaas &

Singer, 2006).

Gamma activity specifically, has garnered significant attention due to its involvement

in working memory processes, as demonstrated in vivo through EEG and MEG studies

(Mainy et al., 2007; Tallon-Baudry, Bertrand, Peronnet, & Pernier, 1998). Increases in

gamma power are associated with increasing working memory load, most noticeably in the

frontal brain region encompassing the DLPFC (Howard et al., 2003; Meltzer et al., 2008;

Pesaran, Pezaris, Sahani, Mitra, & Andersen, 2002). Evidence suggests that GABAA receptor

mediated IPSPs influence the generation of gamma oscillations (Bartos, Vida, & Jonas, 2007;

Wang & Buzsaki, 1996; Whittington, Traub, & Jefferys, 1995), whereas GABAB is involved

in the modulation of gamma oscillations (Brown, Davies, & Randall, 2007; Leung & Shen,

2007).

1.6.3 Working Memory Deficits in Schizophrenia

Working memory deficits among individuals with schizophrenia are one of the most

replicable and robust findings in this population. A meta-analysis was conducted across 187

studies in patients with schizophrenia on three heterogeneous domains of working memory

including verbal working memory, visuospatial memory and executive working memory.

Researchers found clear deficits across all memory domains in the patient group compared to

controls. Of note, no relationship was found between IQ and working memory performance,

thus confirming that these deficits are not simply a function of low IQ (Forbes, Carrick,

McIntosh, & Lawrie, 2009).

Page 45: Evaluating The Effects of Cannabis Dependence and ...

32  

Neurophysiological research has suggested that in individuals with schizophrenia,

working memory deficits may be attributed to dysfunction of the DLPFC, aberrant brain

network connectivity, as well as disruptions in neurotransmitter signalling and neuronal

synchrony (Cho, Konecky, & Carter, 2006; Lewis, Curley, Glausier, & Volk, 2012; Lewis &

Moghaddam, 2006; Sun et al., 2011; Uhlhaas & Singer, 2010). However, inconsistent

findings exist in the literature. For example, a meta-analysis including 12 neuroimaging

studies in patients with schizophrenia found significantly lower DLPFC activation in patients

during the N-back task (Glahn et al., 2005). However, a less conclusive meta-analysis

including 29 studies failed to find a significant difference in frontal activation in patients

compared to controls during a memory task (Van Snellenberg, Torres, & Thornton, 2006).

Additionally, several studies have demonstrated abnormal neural oscillatory activity

in individuals with schizophrenia, predominantly in gamma frequency band (Farzan et al.,

2010a; Gandal et al., 2010; Uhlhaas et al., 2006). EEG studies in epileptic patients (Howard

et al., 2003) and healthy subjects (Barr et al., 2009; Cho et al., 2006) have reported increases

in the modulation of gamma oscillations in the DLPFC with increased working memory load.

In contrast, patients with schizophrenia seem to exhibit little to no gamma modulation in

response to changing cognitive demands (Barr et al., 2010; Cho et al., 2006). It has been

suggested that patients therefore inefficiently enlist complex cognitive processes to carry out

tasks with low cognitive demands whereas healthy controls vary cognitive strategies in

response to task difficulty (Basar-Eroglu et al., 2007). However, inconsistencies in the

literature exist regarding gamma oscillatory activity evoked during working memory tasks in

patients (Basar-Eroglu et al., 2007; Cho et al., 2006; Haenschel et al., 2009; Lewis et al.,

2008). For example, Barr et al., (2010) demonstrated that compared to healthy controls,

Page 46: Evaluating The Effects of Cannabis Dependence and ...

33  

patients with schizophrenia elicited excessive frontal gamma power, specifically during the

3-back condition of the N-back task. Such excessive gamma power was observed at

equivalent performance levels between patients and healthy controls. Interesting, the authors

also found that task performance was inversely correlated to negative symptoms but not with

positive symptoms (Barr et al., 2010). In contrast, a recent study conducted by Chen et al.,

(2014), found that deficits in gamma band amplitude in patients with schizophrenia during a

modified Sternberg working memory task was associated with poorer performance.

Moreover, working memory-induced gamma oscillations were dependent on baseline GABA

levels across all participants (Chen et al., 2014). These findings provide further support for

the importance of GABA function in the production of working memory specific gamma

oscillations, even in those with aberrant system and cognitive functioning. While

discrepancies in the research exist, taken together these findings suggest that neurobiological

alterations in PFC activation, neurotransmitters and neural oscillations contribute to working

memory deficits in schizophrenia.

1.6.4 Cannabis’ Effects on Working Memory

Only recently have researchers begun to accurately assess the effects of cannabis on

working memory, better understanding and controlling for important confounding factors

including frequency of previous cannabis use (Gruber, Sagar, Dahlgren, Racine, & Lukas,

2012; Solowij et al., 2011), and strain of cannabis consumed (i.e., the ratio of THC:CBD)

(Morgan, Schafer, Freeman, & Curran, 2010). These well controlled studies in conjunction

with advances in neuroimaging methodologies have provided a more conclusive

understanding of the acute and long-term effects of cannabis as well as the potential

mechanisms underlying this relationship.

Page 47: Evaluating The Effects of Cannabis Dependence and ...

34  

There is an abundance of research demonstrating that acute administration of THC

has a negative, dose dependent impact on working memory (Ilan, Gevins, Coleman, ElSohly,

& de Wit, 2005; Silva de Melo et al., 2005). Importantly, these findings suggest that despite

acute impairments following cannabis administration, long-term memory function seems to

remain unaffected. Furthermore, it appeared that acute cannabis intoxication did not

significantly impair working memory in heavy users, unless they were administered high

doses of potent cannabis (i.e., high THC content) (Curran, Brignell, Fletcher, Middleton, &

Henry, 2002). This divergent effect in experienced users implies that drug tolerance is

associated with less impairing side effects.

In contrast to the acute effects, conclusions regarding the long-term impact of chronic

cannabis use on working memory are less consistent, due in part to interindividual and

intraindividual variability in cannabis use. More recent evidence has suggested that memory

deficits persist in individuals who use cannabis frequently and over a long period (for full

review see (Schoeler & Bhattacharyya, 2013)). Moreover, early initiation of cannabis use

(prior to the age of 17) and strains high in THC, seem to increase the risk of long-term

memory dysfunction. These findings are especially troubling given the younger onset of

cannabis use as well as recent increases in cannabis potency (Mehmedic et al., 2010).

In line with this research, neurobiological and neuroimaging studies have

demonstrated that long-term, early cannabis use lead to the most significant adverse and

persistent effects on morphology and brain function, beyond that of acute intoxication

(Becker, Wagner, Gouzoulis-Mayfrank, Spuentrup, & Daumann, 2010; Lopez-Larson et al.,

2011; Wilson et al., 2000; Yucel et al., 2010). These neuroimaging studies have also found

that normal levels of working memory accuracy were associated with a common pattern of

Page 48: Evaluating The Effects of Cannabis Dependence and ...

35  

increased overall brain activation, following cannabis use. This enhanced activity in

combination with accurate performance suggests neurophysiological inefficiency, whereby

increased neural effort is needed in regions normally recruited for task completion. However,

it may also suggest that there is a compensatory mechanism involving the recruitment of

regions not normally associated with the specific task. Researchers have suggested that this

may be indicative of a change in strategy to meet task demands (Bossong, Jager,

Bhattacharyya, & Allen, 2014).

One way in which researchers have attempted to better understand the long-term

effects of cannabis use on neurophysiological and cognitive function is by employing

abstinence paradigms as these studies allow for the assessment of reversibility. In a

longitudinal study, Pope and colleagues compared neurocognitive function in heavy cannabis

users compared to controls, over 1 month of abstinence. As hypothesized, significant

memory impairments in the cannabis-using group on days 0, 1, and 7 were observed;

however, the groups were no longer significantly different at day 28, suggesting that the

effects of cannabis on memory may not persist following the complete elimination of

cannabis from the body. In line with this initial observation, several studies have reported

that memory functioning is restored in “regular” cannabis users following abstinence periods

ranging from 48 hours to one month (Jager, Kahn, Van Den Brink, Van Ree, & Ramsey,

2006; Schweinsburg et al., 2005; Schweinsburg et al., 2010).

It is important to take into consideration the potential moderating effects of factors in

addition to current cannabis use on working memory, including previous exposure to

cannabis and additional drugs, frequency and recency of cannabis use, dose of THC

administered, THC:CBD ratio, and the particular assessment of memory function. Further

Page 49: Evaluating The Effects of Cannabis Dependence and ...

36  

difficulty in interpreting these results lies within the heterogeneity of the selected samples,

especially in terms of the criteria used to categorize participants. Previous studies have

included groups of former cannabis users, current regular cannabis users, or heavy cannabis

users, long-term and short-term cannabis users, or early-onset and late-onset cannabis users

(Schoeler & Bhattacharyya, 2013). Overall, it is clear that further research employing diverse

methodologies and controlling for confounding variables is necessary for elucidating the

acute and long-term effects of cannabis use on working memory.

1.6.5 The Combined Effects of Cannabis Use and Schizophrenia on

Working Memory

The relationship between cannabis use and working memory in individuals with

schizophrenia has proven to be more complex than was initially assumed, as current research

provides inconsistent evidence. D’Souza and colleagues (2005) examined the dose-related

effects of intravenous THC using a double blind, randomized, placebo-controlled design in

patients with schizophrenia and controls. Compared to those given the placebo, both patients

and controls given THC exhibited impaired verbal memory, with more profound deficits

observed in the patient group (D'Souza et al., 2005). Similarly, Ringen et al., (2010)

evaluated the relationship between acute cannabis use within the last six months and

cognitive function including verbal and working memory in 140 patients with schizophrenia.

Consistent with previous findings, the researchers found that cannabis use lead to poor verbal

learning and working memory performance compared to abstainers (Ringen et al., 2010).

In contrast, there is a growing body of evidence suggesting that chronic cannabis

users with schizophrenia may perform better on memory tasks than patients with no history

of drug use. Schnell and colleagues (2009) sought to investigate both the enduring effects of

Page 50: Evaluating The Effects of Cannabis Dependence and ...

37  

cannabis consumption and the specific patterns of use on cognition. As such, a cognitive test

battery was administered to 35 patients with schizophrenia with a lifetime diagnosis of co-

morbid cannabis use disorder and 34 patients with no current or past history of cannabis use.

Interestingly, cannabis-using patients performed better on the memory tasks, and more

frequent cannabis use was associated with better performance, dose-dependently (Schnell,

Koethe, Daumann, & Gouzoulis-Mayfrank, 2009). This finding has been replicated in first-

episode patients with schizophrenia (Yucel et al., 2012). This body of work was further

supported by a longitudinal study conducted by Stirling and colleagues (2005). The

researchers found that patients who continued cannabis use during the follow-up period

demonstrated a “sparing” of neurocognitive functioning (Stirling, Lewis, Hopkins, & White,

2005). Taken together, these findings outline the divergent and complex effects of acute and

chronic cannabis use in this patient population and highlight the need for continued research.

1.7 Overview of Current Study Objectives and Hypotheses

Given the high rates of cannabis abuse and dependence among patients with

schizophrenia and the harmful effects of cannabis on illness onset, prognosis and treatment

success (D'Souza et al., 2005; Foti et al., 2010; Linszen et al., 1994; Manrique-Garcia et al.,

2014), research investigating the neurophysiological effects of cannabis use in patients with

schizophrenia is of great importance. Previous neurochemical evidence has suggested that the

effects of cannabis on the human brain lie within the complex interaction between the

cannabinoid system and inhibitory neuronal networks. More specifically, cannabis acts

through the GABAergic system (Eggan & Lewis, 2007), a system vital to inhibitory control

and working memory associated gamma oscillations (Bartos et al., 2007; Brown et al., 2007;

Fitzgerald et al., 2009). In support of this, previous research has demonstrated that cannabis

Page 51: Evaluating The Effects of Cannabis Dependence and ...

38  

impairs cortical inhibition and working memory in otherwise healthy subjects (Ilan et al.,

2005; Silva de Melo et al., 2005) and further exacerbates already present inhibitory deficits in

patients with schizophrenia (Wobrock et al., 2010). Given that the majority these studies are

only conducted cross-sectionally, our understanding is limited to the acute effects of cannabis

in these individuals. Thus it is clear that longitudinal studies, employing abstinence

paradigms among currently dependent patients, are needed to evaluate the long-term

modulatory effects of cannabis on measures of cortical inhibition and working memory. To

our knowledge, this is the first study to address the complex relationship between cannabis,

cognition, and cortical inhibition in patients with schizophrenia using a within- and between-

subjects longitudinal design. The findings of this study may further our understanding of the

mechanisms underlying cannabis use among patients will likely help guide novel treatment

approaches.

Therefore, the aim of this current study is to examine the neurophysiological effects

of cannabis dependence and abstinence in cannabis dependent patients with schizophrenia

and non-psychiatric controls. Firstly, at baseline, we will be evaluating the differential effects

of cannabis dependence cross-sectionally, on cortical inhibition in patients compared to

controls. Through several well-established TMS paradigms, including SICI, LICI, CSP and

ICF, we will be able to differentially index GABAA, GABAB and NMDA receptor function

of the motor cortex. We hypothesize that patients will demonstrate reduced cortical inhibition

and excitation indexed through SICI, LICI and ICF measures, as well as a shorter silent

period, as assessed through CSP. The second objective of the current study is to evaluate the

differential effects of cannabis dependence on working memory performance and gamma

oscillatory activity in patients and controls, cross-sectionally, using the N-back task while

Page 52: Evaluating The Effects of Cannabis Dependence and ...

39  

EEG is recorded. We hypothesize that patients will demonstrate poorer working memory

performance compared to controls. Moreover, we expect to see excessive gamma oscillatory

activity specifically in the frontal region in patients with schizophrenia.

As an exploratory analysis, an additional objective of the current study is to evaluate

the changes, longitudinally, in cortical inhibition and working memory following a 28-day

abstinence period. Thus we will compare TMS measures prior to and following abstinence in

the patients and controls who are able to successfully abstain from cannabis use. We

hypothesize that following abstinence, both patients and controls will exhibit improved

cortical inhibition, with the greatest improvements in patients. Furthermore, we will compare

working memory performance and gamma oscillatory activity, prior to and following the

cannabis abstinence period. We hypothesize that abstinence will improve working memory

performance in both groups compared to baseline, and reduce gamma activity in the patient

population.

Page 53: Evaluating The Effects of Cannabis Dependence and ...

40  

Chapter 2

Methods

2.1 Study Design

This study is a cross-sectional and longitudinal human laboratory study investigating

the effects of cannabis dependence and abstinence on cortical inhibition and working

memory associated gamma oscillations in cannabis dependent patients with schizophrenia

compared to cannabis dependent non-psychiatric controls. A total of 13 participants (7

controls and 6 patients) completed both the pre and post abstinence TMS testing sessions,

while an additional 10 participants (13 controls and 10 patients) completed both working

memory testing session (see figure 3.2). The study duration was approximately 4 weeks; this

included a screening visit, baseline TMS and working memory visit, and a final TMS

working memory test session following the 28-day abstinence period. Included in this

abstinence period was weekly TMS testing along with twice-weekly urine drug testing (see

figure 2.1). In depth procedural details are provided in section 2.4.

Page 54: Evaluating The Effects of Cannabis Dependence and ...

41  

Figure 2.1

Study Design

Figure 2.1: Outline of overall study design including screening assessments along with pre and post TMS and working memory assessments and weekly motor CI measures. Additionally, urine collection took place twice weekly throughout the abstinence period.

This study was approved by the Centre for Addiction and Mental Health’s (CAMH)

Research Ethics Board (REB #169/2011) and in accordance to the declaration of Helsinki.

Written informed consent was obtained for each participant as approved by the CAMH

Research Ethics Board (see Appendix B). Patients with schizophrenia were recruited through

CAMH via flyers, word-of-mouth, various outpatient clinics, and referrals. Healthy controls

were recruited through flyers, referrals from other studies and online advertisements on

Craigslist, and Kijiji.

Page 55: Evaluating The Effects of Cannabis Dependence and ...

42  

2.2 Participants

2.2.1 Power analysis

Initially, we aimed to assess cannabis abstinence in 12 patients with schizophrenia

and 12 healthy controls, based on an anticipated 25% improvement (Cohen’s d=0.73) in

cortical inhibition in patients compared to controls at the end of 28 days of cannabis

abstinence (with α=0.05 and power=80%). Furthermore, an estimated attrition rate of 20%

per group is expected resulting in 10 completers for each group.

2.2.2 Sample

All participants were between the ages of 16 to 55. Additionally, participants had to

meet criteria for cannabis dependence, as confirmed by the Structured Clinical Interview of

the Diagnostic and Statistical Manual of Mental Disorders Fourth Edition (SCID for DSM-

IV-TR, (APA, 2000a) and could not be treatment seeking for such dependence. Current and

lifetime cannabis use and dependence were assessed using several self-report and clinical

interview measures. Current cannabinoid levels were assessed using the Narcocheck, a semi

quantitative urine drug screen, which detects the presence of cannabis, ranging from 25 to

150 ng/ml. All participants were males, allowing us to focus on the most representative

sample of the target population (Goldstein, Seidman, Santangelo, Knapp, & Tsuang, 1994).

Further, all participants were current daily cigarette smokers, given that cigarette use is

common among cannabis users (Patton, Coffey, Carlin, Sawyer, & Wakefield, 2006) and

patients with schizophrenia (Kalman, Morissette, & George, 2005; Lasser et al., 2000),

including co-morbid nicotine use was essential. Tobacco smoking status was assessed via

self-report and was biochemically verified with expired breath carbon monoxide levels

(Vitalograph, Lenexa, KS).

Page 56: Evaluating The Effects of Cannabis Dependence and ...

43  

A diagnosis of schizophrenia or schizoaffective disorder was confirmed using the

criteria outlined in the SCID for DSM-IV-TR (APA, 2000a). Additionally, inclusion criteria

outlined that schizophrenia patients had to be psychiatrically stable, requiring a score <70 on

the Positive and Negative Syndrome Scale (Kay, Fiszbein, & Opler, 1987) (PANSS), and on

a stable dose of antipsychotic medication for a least one month prior to the time of their

assessment. In contrast, healthy controls could not meet SCID for DSM-IV TR criteria for

any current or past Axis I psychiatric disorder except for cannabis dependence or past major

depression, with at least one year of symptom remission. Additionally, healthy controls could

not be taking any psychotropic medications, or have a first-degree relative with psychosis.

Participants were excluded if they met criteria for abuse or dependence for alcohol or

any illicit substances within the past 6 months, with the exception of cannabis, nicotine or

caffeine. The absence of additional substances was verified using Medtox urine toxicology

screens (7-panel-Cannabinoids, Opiates, Amphetamine, Cocaine, Phencyclidine,

Barbiturates, and Benzodiazepines). All measures will be discussed in detail in the following

section. Additional general exclusion criteria included a history of head injury or loss of

consciousness, a neurological or medical condition deemed to affect cognitive function and a

personal or first-degree relative with a history of seizures or syncope. Furthermore, all

participants had to have a Full Scale IQ ≥ 80 as determined by the WTAR (Wechsler, 2001)

and a Test of Memory Malingering (TOMM) score ≥ 45.

Page 57: Evaluating The Effects of Cannabis Dependence and ...

44  

2.3 Measures

2.3.1 Clinical interview measures

Structured Clinical Interview for the DSM-IV-TR (SCID-IV)

The SCID-IV (First, 2002) is a semi-structured interview used to diagnose a current

or lifetime axis I disorder. In the current study, the SCID was used to verify a diagnosis of

schizophrenia or schizoaffective disorder in our patient group and subsequently confirm that

the control group did not meet criteria for any axis I disorder. The SCID also allowed for a

definitive diagnosis for cannabis dependence in all participants.

Positive and Negative Syndrome Scale (PANSS)

The PANSS (Kay et al., 1987) is a 30-item medical scale used to evaluate symptom

severity in individuals with schizophrenia. Through clinical interview, the occurrence and

severity of positive symptoms, negative symptoms and general psychopathology are

assessed. All items are rated using a 7-point scale (from 1=absent to 7=extreme). The PANSS

was chosen due to its high inter-rater reliability (Kay, Opler, & Lindenmayer, 1988) and

suitable construct validity (Kay et al., 1987). Administration occurred at screen and weekly

throughout the abstinence period.

Addiction Severity Index (ASI)

The ASI (McLellan, Alterman, Cacciola, Metzger, & O'Brien, 1992) is a semi-

structured interview used to assess substance abuse and outline the need for new or additional

treatment approaches based on the duration, severity and number of symptoms. The ASI

evaluates several potential problem areas referencing both the past 30 days and one’s

lifetime. These problem domains include psychiatric and medical status, employment and

Page 58: Evaluating The Effects of Cannabis Dependence and ...

45  

support, drug and alcohol use, family/social status, and legal status. Using a ten point scale

(from 0-1 No real problem, treatment not indicated to 8-9 Extreme problem, treatment

absolutely necessary) interviewer severity ratings (ISR) are assigned regarding the severity of

problems in each of the seven domains. The ISRs are used to calculate composite scores

based on the individuals’ responses corresponding to their experiences in the last 30 days.

The ASI was administered at screen in all potential participants.

Calgary Depression Scale for Schizophrenia (CDSS)

The CDSS is semi-structured interview, developed to assess the severity of

depression symptomatology in individuals with schizophrenia (McLellan et al., 1992). The

CDSS is a 9-item measure rated from 0 (absent) to 3 (severe). A total score greater than 4

suggests the presence of minor depression, whereas scores greater than 7 are indicative of

major depression. The CDSS was administered at screen in the patient group only.

2.3.2 Premorbid IQ

Wechsler Test of Adult Reading (WTAR)

The WTAR (Wechsler, 2001) was developed to assess premorbid intelligence. This

assessment is comprised of 50 irregularly spelled words, and each correctly pronounced word

is given a score of 1. The irregularity of the words makes it difficult to correctly pronounce

without having previous knowledge of the words. Thus the participant’s vocabulary, and by

extension IQ, can be assessed. Raw scores were standardized by age.

Page 59: Evaluating The Effects of Cannabis Dependence and ...

46  

2.3.3 Extrapyramidal Side Effects

Abnormal Involuntary Movement Scale (AIMS)

The AIMS (Guy & Cleary, 1976) was designed to measures dyskinetic symptoms, a

common side effect associated with long-term antipsychotic treatment in patients with

schizophrenia. The AIMS allows for early detection and ongoing surveillance of tardive

dyskinesia (TD). This 12-item assessment of involuntary movements is rated on a 5-point

scale ranging from 0 (none) to 4 (severe), except for items 11 and 12 (dental care) which are

answered with either a “yes” or a “no”. A score of 2 or higher, i.e. mild movements in two

categories or moderate movements in one area, (Barnes, 1989) suggest the presence of TD.

Barnes Akathisia Rating Scale (BARS)

The BARS (Barnes, 1989) is used to assess neuroleptic-induced akathisia, a

syndrome of motor restlessness, in individuals with schizophrenia. The BARS incorporates

subjective reports of awareness and distress related to akathisia, along with objective motor

restlessness observations rated using a 4-point scale. Finally, global clinical assessment is

rated using a 5-point scale.

Simpson Angus Rating Scale (SARS)

The SARS (Simpson & Angus, 1970) is used to assess neuroleptic-induced

parkinsonism, evaluating gait (hypokinesia), rigidity and glabella tap, as well as tremors and

salivation. The SARS is a 10-item measurement rated on a 4-point scale.

Page 60: Evaluating The Effects of Cannabis Dependence and ...

47  

2.3.4 Substance Use Measures

The Alcohol Use Disorders Identification Test (AUDIT)

The AUDIT (Saunders, Aasland, Babor, de la Fuente, & Grant, 1993) was developed

by the World Health Organization to quickly screen for excessive alcohol use. The ten-item

measure assesses alcohol consumption, alcohol dependence and other alcohol related

problems. Men who achieve a score of 8 or more (7 for women) out of a possible score of 40,

likely partake in hazardous or harmful drinking behaviours, whereas a score of 20 or more is

indicative of alcohol dependence.

Timeline Follow Back (TLFB)

The TLFB (Sobell, Sobell, Leo, & Cancilla, 1988) is used as an estimate of alcohol,

marijuana, tobacco and caffeine use over a one-week period. Participants were asked to

retrospectively assess the frequency of substance use on a day-to-day basis. Given the

importance of cannabis use in this study specifically, participants were also asked to estimate

the number of times they had purchased cannabis in the last week, as well as the number or

grams and amount of money spent on cannabis.

Fagerstrom Test of Nicotine Dependence (FTND)

The FTND (Heatherton, Kozlowski, Frecker, & Fagerstrom, 1991) is one of the most

commonly used measures, developed to assess self-reported nicotine dependence. This brief

6-item questionnaire ranges in scores from 0 to 10, where higher scores correspond to more

severe nicotine dependence. Importantly, the FTND has been shown to have adequate

reliability in smokers with and without schizophrenia (Weinberger et al., 2007).

Page 61: Evaluating The Effects of Cannabis Dependence and ...

48  

Joint Years

Joint years was used as a measure of cumulative exposure to cannabis. This measure

was adapted from a commonly used measure in nicotine research called cigarette years. A

joint-year was defined as smoking on average one joint per day for 1 year (e.g., 1 joint year =

365 joints smoked in one year).

Urine Drug Screen

The Narcocheck is a semi-quantitative urine drug-screening test that has 5 levels of

cannabis detection ranging from 25 to 150 ng/ml. This test is able to detect marijuana,

hashish and cannabis resin present in the users urine. In this study, the Narcocheck was used

once weekly throughout the abstinence period to assess for changes in cannabis using

behaviours and verify abstinence at day 28.

2.3.5 Cortical Inhibition

Participants were instructed to abstain from cannabis 12 hours prior to the TMS

session; however, ad libitum cigarette use was allowed throughout the test session to avoid

nicotine withdrawal. During testing, participants were seated in a comfortable chair with their

arms supported passively by a pillow, and were asked to maintain relaxation throughout the

session. Surface EMG was recorded from the abductor pollicis brevis (APB) muscle of the

right hand with disposable disc electrodes placed in a tendon-belly arrangement over the bulk

of the muscle. An additional metal ground electrode was placed on the inside of the forearm

distal to the wrist. EMG was monitored on a computer screen and the signal was amplified

(Intronix Technologies Corporation Model 2024F, Bolton, ON, Canada), filtered (band pass

Page 62: Evaluating The Effects of Cannabis Dependence and ...

49  

2 Hz–2.5 kHz), digitized at 5 kHz (Micro 1401, Cambridge Electronics Design, Cambridge,

UK) and stored in a laboratory computer for offline analysis.

TMS was applied to the hand area of the left motor cortex with a 7 cm figure-of-eight

magnetic coil and two Magstim 200 magnetic stimulators (Magstim, Whitland, Dyfed,

Wales). The coil was held tangentially on the head with the handle pointing backward

midline at a 45° lateral angle. The optimal coil position was determined for each subject

individually and was marked to ensure consistent positioning. The RMT was defined as the

minimal intensity that produced a MEP of >50 μV in 5 of 10 trials in relaxed the APB muscle

(Kujirai et al., 1993). The output intensity required to elicit a MEP 1 mV peak to peak was

determined (Kujirai et al., 1993).

SICI and ICF were assessed using a standard paired pulse procedure (Kujirai et al.,

1993). A subthreshold conditioning stimulus (CS), which was set at 80% of RMT, preceded a

suprathreshold test stimulus (TS). The TS was delivered at an intensity that produced an

average 1mV peak-to-peak amplitude (Kujirai et al., 1993). The CS was applied to the motor

cortex before the TS at one of five randomized ISIs: 2 ms and 4 ms for SICI and 10 ms,

15 ms, and 20 ms for ICF along with the TS alone. Seventy-two trials were performed, 12 for

each condition. Changes in the TS MEP amplitude at each ISI were expressed as a

percentage of the mean unconditioned MEP amplitude (Daskalakis, Christensen, Fitzgerald,

& Chen, 2002).

For LICI, a suprathreshold CS preceded a suprathreshold TS (set at the adjusted 1 mv

peak-to-peak) at one of three randomized ISIs: 100 ms, 150 ms and 200 ms. Forty trials were

performed, 10 for each ISI as well as 10 for the TS alone. Finally, the measurement of the

Page 63: Evaluating The Effects of Cannabis Dependence and ...

50  

CSP duration was obtained in moderately tonically active APB muscle (i.e., 20% of

maximum contraction) by stimulating the motor cortex with an intensity of 140% of the

RMT. Ten trials were performed. CSP duration was measured as the time from the MEP

onset to the return of any voluntary EMG activity, referred to as the absolute CSP. The CSP

was determined with our previously published automated method (Daskalakis et al., 2003).

Using CFSview in CED Signal 2.0, the 10 trials were averaged off- line and CSP duration

was measured on an averaged recording. The absolute CSP value was calculated as the time

of offset of the period of EMG activity suppression minus the time of onset. The order of

administration of SICI/ICF, LICI and CSP were counterbalanced for each participant (refer

to figure 2.2).

Page 64: Evaluating The Effects of Cannabis Dependence and ...

51  

Figure 2.2

Motor Cortical Inhibition Paradigms

Figure 2.2. Adapted from (Barr, Fitzgerald, Farzan, George, & Daskalakis, 2008). EMG recordings following: A) SICI a conditioning stimulus (CS) precedes the TS by 2–5 ms to and inhibits corresponding MEP B) LICI, the CS precedes the TS by approximately 100–200 ms and inhibits the corresponding MEP C) CSP induced following a 140% suprathreshold TS while the right hand muscle is tonically activated D) ICF a conditioning stimulus (CS) precedes the TS by 10-20 sec.

A

B

C

D

Page 65: Evaluating The Effects of Cannabis Dependence and ...

52  

2.3.6 Working Memory

N-Back Task

Subjects performed the N-back task while EEG activity was recorded (STIM2,

Neuroscan, U.S.A.). Black capital letters were presented on a computer monitor one at a time

in a continuous sequence. Participants were instructed to respond by pushing a button (2;

target) if the present stimulus was identical to the stimulus presented N (1 or 3) trials back;

otherwise, participants pushed a different button (1; non-target). Each letter was present on

the screen for 250 ms, followed by a 3000 ms time frame to respond. Stimuli were presented

for 15 minutes in the 1-back condition and 30 minutes in the 3-back, ensuring that there were

a satisfactory number of correct responses for data analysis (refer to figure 2.3). The number

of target letters in each condition was: 46 of 198 (23.2%) in the 1-back and 59 of 400 trials

(14.6%) in the 3-back condition. Outcome measures included task accuracy and response

time. The order of conditions were randomized and counterbalanced across subjects to

prevent order effects.

EEG Measurement of Induced γ Oscillatory Activity

Induced oscillatory responses are non-phase-locked to the stimulus onset with a

latency within the first 500 ms of stimulus onset, which varied trial-by-trial (Tallon-Baudry,

Kreiter, & Bertrand, 1999). In contrast to induced activity, evoked oscillatory activity is

phase-locked to the stimulus onset, with a fixed latency within 100 ms of the stimulus onset

(Tallon-Baudry et al., 1999). Although both evoked and induced gamma activities are

associated with working memory (Barr et al., 2008; Basar-Eroglu et al., 2007; Howard et al.,

2003), it has been suggested that induced gamma band activity may be more closely related

to the maintenance of information in working memory, whereas evoked oscillations are

Page 66: Evaluating The Effects of Cannabis Dependence and ...

53  

considered less related to higher cognitive functions (Tallon-Baudry et al., 1999). Several

studies have demonstrated that evoked oscillations are more strongly influenced by attention

(Tiitinen et al., 1993) and task instructions (Hermann, 1993). As such, we measured mean

induced gamma power from frontal electrodes (Fpz, Fp1, Fp2, AFz, AF3, AF4, AF7, AF8,

Fz, F1, F2, F3, F4, F5, F6, F7, F8) while subjects completed the N-back task.

Figure 2.3

N-Back Task

Figure 2.3. Adapted from (Barr et al., 2013) A) An example of the 1- and 3-back conditions that were completed by patients and controls. Participants were asked to push a button if the current letter was identical to the letter presented N trials back and push a different button for the non-targets. Only correct responses for target (TC) were included in the data analysis. B) The timing of one trial for a total time of 3000 ms including the presentation of a letter separated by a + sign followed by a subsequent letter. The data gamma power were measured following the presentation of the stimulus (250 ms) until 750 ms.

 

Page 67: Evaluating The Effects of Cannabis Dependence and ...

54  

EEG Recording

EEG data were acquired using a 64-electrode cap and Synamps2 DC-coupled EEG

system (Compumedics, U.S.A.). Data were recorded at a rate of 1000 Hz DC and with a 0.3

to 200 Hz band pass hardware filter. Electrode impedances were lowered to < 5 kΩ. All

channels were referenced to the mastoid electrodes, TP7 and TP8.

Offline EEG Processing

EEG recordings were processed offline using MATLAB (The MathWorks Inc.

Natick, MA, USA) and EEGLAB toolbox (Delorme reference). Data were down sampled to

1 kHz and filtered by using second order, Butterworth, zero-phase shift 1–55 Hz band pass

filter. Second, EEG data was segmented -1400 ms to +3100 ms relative to the stimulus onset.

Next, a channels-by-trials matrix of all ones was created and assigned the value to zero if an

epoch had: (1) amplitude larger than +/- 150 µV; (2) power spectrum that violated 1/f power

law; or (3) standard deviation 3 times larger than average of all trials. A channel was rejected

if its corresponding row had more than 60% of columns (i.e., trials) coded as zeros and an

epoch was removed if its corresponding column had more than 20% of rows (i.e., channels)

coded as zeros. Epochs were then manually inspected and trials containing irregularities were

removed. Finally, an independent component analysis (ICA) (EEGLAB toolbox; Infomax

algorithm) was performed to remove eye-blink traces, muscle artifacts, and other noise from

the EEG data.

Gamma power was analyzed for the target trials that were responded to correctly

across all N-back conditions. The raw data was filtered for gamma (30-50 Hz) frequency

ranges with zero-phase shift. We then calculated the time series for gamma amplitude using

the Hilbert transform. Additionally, we created concatenated signal of 5000 ms using

Page 68: Evaluating The Effects of Cannabis Dependence and ...

55  

randomly selected epochs for each subject, each trial type, and at each electrode, as this

corresponds with induced gamma power.

2.3.7 Cannabis Abstinence Paradigm

Abstinence was assessed weekly through the administration of NarcoCheck.

However, to our knowledge this test has not been measured in a clinical research setting and

thus the NarcoCheck was only used to capture decreasing THC levels or alternatively any

sudden increases throughout the 28-day abstinence period. End-point abstinence was

assessed using MEDTOX urine toxicology. Abstinent individuals were those who did not use

cannabis within the 28-day period and demonstrated endpoint detection THC-COOH levels

below 50ng/ml, which is the accepted cut-off level in the literature (Huestis, Mitchell, &

Cone, 1995). If participants did not meet these criteria, they were classified as relapsers or

non-abstainers. All stored urine from those classified as relapses was sent to our CAMH

clinical laboratory and was further analyzed by gas chromatography-mass spectroscopy to

obtain quantitative THC-COOH and creatinine concentrations. This analysis confirmed that

relapsers did in fact use cannabis over the abstinence period.

Contingency management and brief behavioural therapy were administered weekly to

aid in cannabis abstinence. The basic model of contingency managment promotes

behavioural change through the administration of positive or negative contingencies. In the

current study, we used a cost-effective strategy called the “fishbowl” technique whereby

participants who demonstrated decreasing THC levels through urinalysis, were able to select

a slip of paper from a fish bowl. The bowl will contain 250 slips of paper, half of which were

nonwinning slips that said Sorry, try again while the winning slips could be exchanged for

prizes that varied in value. Of the winning slips, 109 were small prizes consisting of $1-5

Page 69: Evaluating The Effects of Cannabis Dependence and ...

56  

coupons to local restaurants, 15 were large prizes, worth up to a maximum of $20 in value

and 1 of the slips was a jumbo prize valued at $100. In addition to the weekly contingency

management, on day 29, participants received a $3000 bonus if urine results were indicative

of cannabis abstinence.

Furthermore, individual weekly therapy sessions were administered on days 1, 8, 15

and 22 in order to aid in study attendance and abstinence. A combination of psychoeducation,

motivational interviewing, coping skills therapy, and cognitive-behavioural relapse

prevention were implemented, given research suggesting that a combination of interventions

leads to the highest rates of abstinence success (Budney et al., 2000). Early sessions focused

on building a rapport and provided psychoeducation in order to heighten awareness of the

personal consequences associated with cannabis use. Additionally, motivation interviewing

was used to increase participants’ willingness to stop using. Finally, later sessions focused on

coping strategies and relapse prevention techniques. These therapy sessions lasted

approximately 30 minutes and were conducted by trained research assistants in the CAMH

Schizophrenia Program. .

2.4 Study Procedure

Once potential participants were identified, a telephone screen was completed, which

briefly assessed eligibility based on current and past medical and substance use. Those who

were deemed eligible were invited into the Biobehavioural Addictions and Concurrent

Disorders Research Laboratory (BACRDL; Principal Investigator: Dr. Tony P. George,

M.D., FRCPC) at CAMH for an additional screening assessment. Upon arrival for a

screening assessment, participants were asked to complete the informed consent as well as a

post-consent quiz to verify their understanding of the consent. Following this, demographic

Page 70: Evaluating The Effects of Cannabis Dependence and ...

57  

information, IQ testing, as well as measures of depression (HAM-D), drug use (ASI, TLFB,

AUDIT), carbon monoxide levels and Medtox™ urine toxicology were administered. For

schizophrenia patients, clinical assessments were conducted including the SCID-IV, PANSS,

with the AIMS, SARS, and BARS. The candidate, Michelle Goodman conducted screening

measures along with Rachel Rabin, who primarily conducted the SCID, PANSS, and ASI

assessments.

Data presented are a subset of a larger study, therefore only baseline and post

abstinence testing sessions will be included. All testing sessions were conducted at the

Temerty Centre for Therapeutic Intervention, CAMH. Participants were instructed to abstain

from using cannabis 12 hours prior to baseline testing. During the baseline visit, motor

cortical inhibition was administered including SICI, LICI, ICF and CSP. Following this, N-

Back EEG was collected for the 1 and 3-Back conditions. This testing session was repeated

following the 28-day abstinence period. Participants were compensated for their time at a rate

of $10 per hour, which they received at the end of every testing session, after approximately

3 hours.

2.5 Data Analysis

2.5.1 Study Sample

Independent t-tests and chi-squared tests were used to analyze differences between

the two diagnostic groups on demographic variables including age, years of education, IQ,

daily cannabis and cigarette use and joint years.

Page 71: Evaluating The Effects of Cannabis Dependence and ...

58  

2.5.2 Cortical Inhibition

All data were analyzed using the Statistical Program for Social Sciences (SPSS)

version 21.0 (SPSS Inc., Chicago, Ill). All tests were two-tailed and the level of significance

was set at α = 0.05. Separate repeated measures ANOVAs were conducted for SICI, LICI

and ICF with diagnosis (patients versus controls) as the between subjects factor and ISI (SICI

ISI: 2 ms and 4 ms, ICF ISI: 10 ms, 15 ms and 20 ms, LICI ISI: 100 ms, 150 ms and 200 ms)

as the within subjects factor. Independent t-tests were conducted for RMT and CSP. Post-hoc

tests were conducted where appropriate with an adjusted Bonferroni corrected alpha level to

account for multiple comparisons. Pearson correlation analyses were conducted on each of

the TMS paradigms and demographic variables, and were analyzed within groups when

significant.

2.5.3 Working Memory

Baseline working memory performance (accuracy and reaction time) and gamma

oscillatory activity were analyzed using separate repeated measures ANOVAS with diagnosis

(schizophrenia vs. healthy controls) as the between-subjects factor and N-back load (1- and

3-back) as the within-subjects factor. Pearson correlation analyses were conducted on

performance and oscillatory activity on the 1-back and 3-back condition and demographic

variables, and were analyzed within groups when significant.

Page 72: Evaluating The Effects of Cannabis Dependence and ...

59  

Chapter 3 Results

3.1 Baseline Results

3.1.1 Demographics

A total sample of 26 individuals participated in the baseline testing session of this

study, recruited over a 24-month period. Twelve of these participants met diagnostic criteria

for schizophrenia or schizoaffective disorder and fourteen were non-psychiatric healthy

controls. Of these participants, all completed baseline N-Back testing. However, 3 patients

with schizophrenia and 5 healthy controls failed to meet eligibility for TMS testing due to

history of concussions and other head injuries, history of seizures, as well as 1 control

participant who did not want to take part in this portion of the study (refer to figure 3.2).

All patients were clinically stable, with PANSS scores below 70. Ten of these

patients were on atypical antipsychotic medications including: risperidone (4), quetiapine (3)

olanzapine (1), clozapine (1), and paliperidone (1). Furthermore, 1 patient was on the typical

antipsychotics flupentixol, and 1 patient received both fluphenazine and quetiapine. In

addition to antipsychotic medications, several patients were on antidepressants for symptoms

of depression or anxiety including escitalopram, lorazepam and citalopram and trazodone.

Finally, 1 of these patients was on the anticholigeric medication benzotropine as well as

several additional cholesterol and diabetes medications including metformin, nicotinic acid,

atorvastatin, indapamide, and glyburide.

Page 73: Evaluating The Effects of Cannabis Dependence and ...

60  

Given that sample sizes differed between cortical inhibition and working memory

assessments, separate demographic tables were presented for our TMS and working memory

findings. The following demographics included all baseline participants who completed the

working memory assessment (for baseline TMS demographics refer to table 3.1). As

expected, group differences were revealed on measures of IQ as measured by the WTAR,

with higher average IQ scores in the control group (t(25) = -3.061, p =.005). Group

differences were revealed on years of education, with fewer years seen in the patient group

(t(25) = -2.116, p = .044). Importantly, there were no significant differences in daily cigarette

use (p = .520), daily cannabis use (p = .971) and joint years (p =.157) (refer to table 3.3).

Interestingly, IQ was negatively correlated with joint years (r (25) = -.393, p = .043). This

correlation only remained significant when all participants were included.

Figure 3.1. Significant negative correlation between IQ and joint years r (25) = -.393, p =.043, Thus higher IQ was associated with fewer joint years.

0

5

10

15

20

25

30

60 70 80 90 100 110 120

Join

t Yea

rs

IQ

Page 74: Evaluating The Effects of Cannabis Dependence and ...

61  

Figure 3.2

Consort Diagram for patients with schizophrenia (SZ) and healthy controls (HC)

 

 

 

 

 

 

 

 

 Assessed for eligibility via phone screen (N=123)

 Baseline TMS sample SZ= 9 HC= 10

 Baseline N-Back sample SZ= 12 HC= 14

  Initially eligible (N=71)

 Total sample (N= 26)

 

Excluded (n = 47): Concurrent Substance use or Axis I, PANSS > 70, FSIQ < 80, or Negative for THC

 Completed TMS sample SZ= 6 HC= 7

 Completed N-Back sample SZ= 9 HC= 13

Page 75: Evaluating The Effects of Cannabis Dependence and ...

62  

3.1.2 Baseline TMS Assessments Table 3.1 Baseline TMS demographics, clinical means, and standard deviations by diagnosis

_______________________________________________________________________ HC (N = 10) SZ (N = 9) p-value . Age (years) 30.54 ± 6.98 30.33 ± 9.63 .955 Race Caucasian 70% (7) 11.1% (1) skskdn African American 20% (2) 66.7% (6) Other 10% (1) 22.2% (2) Education (years) 12.3 ± 2.00 11.0 ± 2.18 .191 IQ 99.54 ± 9.43 91.67 ± 1.01 .089 PANSS positive - 13.78 ± 2.95 negative - 11.89 ± 2.71 general - 27.44 ± 4.5 total - 53.11 ± 8.31 CPZ equivalent (mg/day) - 531.92 ± 262.65 CPD 11.99 ± 1.02 9.52 ± 5.91 .592 GPD 1.86 ± 1.27 1.30 ± .88 .276 Joint years 8.75 ± 4.8 12.35 ± 9.36 .281 ________________________________________________________________________

Values are expressed as mean ± SD HC = Healthy controls, SZ = patient with schizophrenia CPZ = chlorpromazine, CPD = cigarettes per day, GPD = grams per day

3.1.2.1 Baseline Inhibitory Assessments: SICI, LICI, and CSP

To evaluate baseline differences in GABAA inhibitory activity between patients and

controls (SICI), a repeated measures ANOVA was conducted with ISI (2 ms versus 4 ms) as

the within-subjects variable and diagnosis (patients and controls) as the between-subjects

variable. This analysis revealed a significant main effect of ISI (F(1,17) = 9.244, p = .007)

(see figure 3.3). A paired samples t-test revealed greater inhibition at 2 ms compared to 4 ms

(t(18)   = 3.123, p =0.006). No significant ISI x diagnosis interaction was found (p = .888),

Page 76: Evaluating The Effects of Cannabis Dependence and ...

63  

however there was a trend towards a significant diagnosis effect (F(1,17) = 3.473, p = 0.080),

with greater inhibition in patients.

Figure 3.3. Baseline short interval cortical inhibition (SICI) between patients (SZ) and healthy controls (HC) across 2 ms (SICI2) and 4 ms (SIC4) ISIs. Results revealed a significant main effect of ISI F(1,17) = 9.244, p = .007. Bars represent (±) 1 standard deviation. Asterisks (*) represent significance.

To evaluate baseline differences in GABAB inhibitory activity between patients and

controls measured through LICI, a repeated measures ANOVA was conducted with ISI (100

ms versus 150 ms versus 200 ms) as the within-subjects variable and diagnosis (patients

versus controls) as the between-subjects variable. Results revealed a significant main effect

of ISI (F(2,38) = 8.876, p = .001)  (see figure 3.4). A paired samples t-test found significant

differences between150 and 200 ms (t(20) = 3.180, p = 0.005) and 100 and 200 ms (t(2) =

2.366, p = 0.028). Only the first comparison remained significant following an adjustment for

multiple comparisons (Bonferroni correction) with greater inhibition in the 150 ms condition.

No significant diagnosis effect was found (p = .928), however a trend towards a significant

ISI x diagnosis interaction (F(2,38) = 2.778, p = 0.075) was revealed.

-­‐80  

-­‐60  

-­‐40  

-­‐20  

0  

20  

40  

60  

80  

100  

120  

SICI2 SICI4

HC

SZ

Interstimulus Interval

*

Cor

tical

Inhi

bitio

n (%

ME

P re

duct

ion)

Page 77: Evaluating The Effects of Cannabis Dependence and ...

64  

          Figure 3.4. Baseline long interval cortical inhibition (LICI) between patients (SZ) and healthy controls (HC) across 100 ms (LICI100), 150 ms (LICI150) and 200 (LICI200). Results revealed a significant main effect of ISI (F(2,38) = 8.876, p = .001). Bars represent (±) 1 standard deviation. Asterisks (*) represent significance.

-­‐60  

-­‐40  

-­‐20  

0  

20  

40  

60  

80  

100  

120  

140  

LICI100 LICI150 LICI200

HC

SZ

Cor

tical

Inhi

bitio

n (%

ME

P re

duct

ion)

Interstimulus Interval

* *

Page 78: Evaluating The Effects of Cannabis Dependence and ...

65  

An additional measure of GABAB inhibitory activity was assessed using an

independent t-test with CSP as the test variable and diagnosis as the grouping variable.

Results revealed significantly longer baseline CSP in patients (M = 0.167, SD = 0.030)

compared to controls (M = 0.135, SD = 0.010 SD) (t(18) = 2.257, p = 0.037).  

Figure 3.5. Baseline cortical silent period (CSP) for patients (SZ) and healthy controls (HC). Significant differences were found between diagnosis regarding the lengths of silent periods (t(18) = 2.257, p = 0.037). Bars represent (±) 1 standard deviation. Asterisks (*) represent significance.

0  

0.02  

0.04  

0.06  

0.08  

0.1  

0.12  

0.14  

0.16  

0.18  

0.2  

HC SZ

HC

SZ

Cor

tical

Sile

nt P

erio

d D

urat

ion

(sec

onds

) *

Page 79: Evaluating The Effects of Cannabis Dependence and ...

66  

3.1.2.2 Baseline Excitatory Assessments: RMT and ICF

To examine potential baseline differences in cortical excitatory activity between

patients and controls, we conducted an independent t-test on RMT. Results revealed no

significant diagnostic differences (p = 0.207). Additionally, a repeated measures ANOVA

was conducted on ICF to assess NMDA cortical excitation. There were also no significant

differences in ICF between controls and patients (p = 0.278); however, a large effect size

was revealed for the 20 ms interval (Cohen’s d = .951;refer to table 3.2).

Figure 3.6. Baseline cortical facilitation (ICF) between patients (SZ) and healthy controls (HC) across 10 ms (ICF10), 15 ms (ICF15) and 20 ms (ICF20). No significant differences were found at any ISI. Bars represent (±) 1 standard deviation.

0  

50  

100  

150  

200  

250  

ICF10 ICF15 ICF20

HC

SZ

Interstimulus interval

Cor

tical

Fac

ilita

tion

(%M

EP e

nhan

cem

ent)

Page 80: Evaluating The Effects of Cannabis Dependence and ...

67  

Table 3.2 Cohen’s D and p-values for TMS Measures

.

TMS Measure ISI HC (N = 10) SZ (N = 9) Cohen’s d p-value . .

SICI 2 ms 29.5 ± 62.8 72.2 ± 24.0 .915 .065 4 ms 14.9 ± 70.3 54.1 ± 27.5 .794 .109

LICI 100 ms 58.3 ± 43.1 58.2 ± 70.4 .065 .881 150 ms 61.4 ± 38.9 67.6 ± 63.9 .177 .683 200 ms 50.2 ± 39.3 24.0 ± 66.2 .342 .429

CSP - 0.13 ± 0.03 0.16 ± 0.03 .926 .037

ICF 10 ms 130.2 ± 64.7 148.2 ± 57.8 .198 .109 15 ms 128.4 ± 54.1 150.7 ± 52.7 .336 .668 20 ms 103.3 ± 44.8 147.9 ± 51.1 .951 .054

___________________________________________________________________________ Bolded values indicate a medium to large effect size (>.5)

Percent inhibition or facilitation are expressed as mean ± SD. Cohen’s D values and p-values comparing short interval cortical inhibition (SICI), long interval cortical inhibition (LICI), cortical silent period (CSP) and intracortical facilitation (ICF) between patients (SZ) and healthy controls (HC). Bolded results indicate that there is a strong or significant effect for SICI (2 ms and 4 ms ISI), CSP and ICF (20ms ISI).

Page 81: Evaluating The Effects of Cannabis Dependence and ...

68  

3.1.3 Baseline Working Memory Assessments

Table 3.3

Baseline N-Back demographics, clinical means, and standard deviations by diagnosis     _______________________________________________________________________

HC (N = 14) SZ (N = 12) p-value . Age (years) 28.93 ± 6.62 32.00 ± 1.02 .353 Race Caucasian 78.6% (11) 25.1% (3) skskdn African American 14.3%(2) 50.0% (6) Other 7.1% (1) 16.7% (2) Education (years) 13.1 ± 2.67 11.13 ± 2.02 .044* IQ 102.40 ± 9.43 91.17 ± 9.37 .005* PANSS positive 13.67 ± 3.06 negative 11.58 ± 2.61 general 27.00 ± 4.77 total 52.25 ± 9.01 CPZ equivalents (mg/day) 463.21 ± 302.06 CPD 10.43 ± 9.05 12.56 ± 7.55 .520 GPD 1.55 ± 1.20 1.53 ± .89 .971 Joint years 7.94 ± 4.50 11.68 ± 8.60 .157 ________________________________________________________________________

Values are expressed as mean ± SD Results that are significant at the 0.05 level are marked with an (*) HC = healthy controls, SZ = patients with schizophrenia, CPZ = chlorpromazine, CPD = cigarettes per day, GPD = grams per day

3.1.3.1 Baseline Working Memory Performance

In total, 12 patients with schizophrenia and 14 healthy controls were included in our

baseline analysis. As expected, a repeated measures ANOVA revealed a main effect of N-

back load, between the 1-back condition (M = 85.45, SD = 12.09) and the 3-back condition

(M = 49.67, SD = 23.09) (F(1,24) = 97.021, p > 0.001). Both groups demonstrated

significantly lower scores on the 3-back, as measured by a paired t-test (t (25) = 10.019, p <

0.001)(refer to figure 3.7). No significant N-back load x diagnosis interaction was found (p =

Page 82: Evaluating The Effects of Cannabis Dependence and ...

69  

0.695). No significant effect of diagnosis was revealed (p = 0.186). A repeated measures

ANOVA on reaction time (ms) found a main effect of load, between the 1-back condition (M

= 821.23, SD = 166.76) and the 3-back condition (M = 1041.40, SD = 258.80) (F(1,24) =

23.835, p > 0.001). As expected, paired samples t-test revealed longer reaction times in the 3-

back condition (t (25) = -5.056, p < 0.001).

Figure 3.7. Baseline N-back performance between patients (SZ) and healthy controls (HC) and the 1-back and 3-back conditions. Performance is significantly lower in the 3-back condition across both groups F(1,24) = 23.835, p > 0.001. No performance differences were found in the 1-back condition or 3-back condition between groups. Bars represent (±) 1 standard deviation. Asterisks (*) represent significance.

3.1.3.2 Baseline Gamma Power

To evaluate baseline gamma oscillatory activity elicited during a working memory

task, a repeated measures ANOVA was conducted for the frontal electrodes encompassing

the DLPFC (Fpz, Fp1, Fp2, AFz, AF3, AF4, AF7, AF8, Fz, F1, F2, F3, F4, F5, F6, F7, F8)

and for the target correct responses only, with N-back condition (1 and 3-back) as the within-

0  

20  

40  

60  

80  

100  

120  

1-Back 3-Back

HC

SZ

N-Back Condition

Mea

n Ta

rget

Cor

rect

Res

pons

e (%

)

*

Page 83: Evaluating The Effects of Cannabis Dependence and ...

70  

subjects variable and diagnosis (patients versus controls) as the between-subjects variable.

Results did not revealed a significant main effect of memory load (p = .589) nor did we find

a significant N-back load x diagnosis interaction (p = .390). Similarly no significant effect of

diagnosis was revealed (p = .823).

1-Back 3-Back

   

HC SZ HC SZ

Figure 3.8. Mean gamma (30-50 Hz) oscillatory power from target correct (TC) responses elicited across 1-back and 3-back conditions in patients (SZ) compared to healthy controls (HC). Bar graphs represent mean gamma power across the frontal electrodes (Fpz, Fp1, Fp2, AFz, AF3, AF4, AF7, AF8, Fz, F1, F2, F3, F4, F5, F6, F7, F8). Bars represent (±) 1 standard deviation. Topographical illustration of mean gamma power elicited during 1- and 3-back in patients with SCZ compared to HS. Greater gamma power is represented by hot or red colours.

0  

5000  

10000  

15000  

20000  

25000  

30000  

35000  

40000  

HC SZ

Mea

n G

amm

a P

ower

µV

2

HC

SZ

0  

5000  

10000  

15000  

20000  

25000  

30000  

35000  

40000  

HC SZ

HC

SZ

Mea

n G

amm

a P

ower

µV

2

Page 84: Evaluating The Effects of Cannabis Dependence and ...

71  

3.2 Exploratory Longitudinal Results

The following section is exploratory given that this study is too underpowered to

assess the effects of cannabis abstinence in patients and controls. Abstinent individuals were

defined as those who did not use cannabis within the 28-day period and who demonstrated

endpoint THC-COOH levels below 50ng/ml (for more details refer to section 2.3.7).

Urinalysis was conducted for most of the participants in the study and reported as

normalized-creatinine THC-COOH levels. THC-COOH is the main non-psychoactive

metabolite detected in the urine of cannabis users. Normalizing THC-COOH concentrations

to urinary creatinine controls for individual variability in hydration and urine output (Lafolie

et al., 1991). It should be noted that at baseline, healthy control abstainers had higher levels

of THC-COOH as compared to patient abstainers. Regardless of diagnosis, at baseline,

relapsers demonstrated higher average THC-COOH levels than the abstainer.

Table 3.4 Creatinine-normalized THC-COOH levels in patient and control abstainers and relapsers

. Group Diagnosis Baseline THCCOOH Post THCCOOH

(ng/ml) (ng/ml) .

Abstainers HC (N = 7) 77.68 ± 110.70 2.83 ± 5.40

SZ (N = 3) 21.9 ± 9.53 1.13 ± 1.19

Relapsers HC (N = 2) 157.35 ± 68.93 127.45 ± 130.74

SZ (N = 3) 86.9 ± 66.93 50.17 ± 78.2 ___________________________________________________________________________

Urinalysis of creatinine-normalized TH-CCOOH levels prior to and following a 28-day abstinence period in patients (SZ) and healthy controls (HC). THC-COOH is the main non-psychoactive metabolite, detected in the urine of cannabis users. Values above 50ng/ml are representative of “relapse” or new cannabis use.

Page 85: Evaluating The Effects of Cannabis Dependence and ...

72  

3.2.1 Longitudinal TMS Assessments Six patients with schizophrenia and 7 controls participated in the abstinence TMS

testing session. Of the patients who participated, 4 successfully abstained, while 2 relapsed

and for the controls, 3 successfully abstained and 4 relapsed (see Table 3.4).

Table 3.5 Post TMS demographics, clinical means, and standard deviations of the sample by diagnosis   _______________________________________________________________________ HC abst HC non-abst SZ abst SZ non-abst

(N = 3) (N = 4) (N = 4) (N = 2) . Age (years) 29.0 ± 5.57 32.0 ± 6.16 32.75± 13.2 26.5 ± .707 Race Caucasian 66.7% (2) 66.7% (2) - 50% (1) skskdn African American - 33.3% (1) 75% (3) 50% (1) Other 33.3% (1) - 25% (1) - Education (years) 12.0 ± 3.0 12.0 ± 1.41 12.75 ± 1.5 11.0 ± 1.41 IQ 98.67 ± 8.08 95.75 ± 8.90 92.25 ± 11.4 92.0 ± 5.6 PANSS positive - - 14.75 ± 3.10 14.5 ± 4.95 negative - - 11.0 ± 2.83 11.0 ± 2.83 general - - 29.5 ± 4.20 25.0 ± 5.66 total - - 55.25 ± 9.29 50.5 ± 13.0 CPZ equivalents (mg/day) - - 549.7 ± 4111.9 250 ± 353. 5 CPD 18.86 ± 18.41 9.43 ± 6.31 7.22 ± 3.96 16.0 ± 5.6 GPD 1.83 ± 1.56 2.74 ± 1.30 .748 ± .227 2.31 ± 1.60 Joint years 12.11 ± 4.99 10.35 ± 3.15 13.65 ± 13.12 9.66 ± 4.73 __________________________________________________________________________

Values are expressed as mean ± SD HC = healthy controls, SZ = patients with schizophrenia Abst = abstainer and non-abst = relapse CPZ = chlorpromazine, CPD = cigarettes per day, GPD = grams per day

3.2.1.1 Longitudinal Inhibitory Assessments: SICI, LICI, and CSP

We aimed to evaluate the potential longitudinal changes in GABAA inhibitory activity

through SICI. We compared the reduction in cortical inhibition at baseline and following 28

Page 86: Evaluating The Effects of Cannabis Dependence and ...

73  

days of abstinence in patients with schizophrenia and healthy controls. Furthermore, we

separately graphed patients and controls who successfully abstained (figure 3.9 A) from

those who relapsed (figure 3.9 B).

                         

Figure 3.9. Longitudinal data for short interval cortical inhibition (SICI) between patients (SZ) and healthy controls (HC) across 2 ms (SICI2) and 4 ms (SIC4) ISIs at baseline and at following 28-days of abstinence. A represents a graph of abstainer HC and SZ, B represents a graph of relapser HC and SZ. Bars represent (±) 1 standard deviation.

To evaluate longitudinal changes in GABAB inhibitory activity, we administered

LICI and CSP in patients and controls. We compared the reduction in cortical inhibition

(figure 3.10) as well as the change in silent period length (figure 3.11) at baseline and

-­‐80  

-­‐60  

-­‐40  

-­‐20  

0  

20  

40  

60  

80  

100  

120  

SICI 2 SICI 4 SICI 2 SICI 4

HC SZ

Baseline

Day-28

-­‐100  

-­‐80  

-­‐60  

-­‐40  

-­‐20  

0  

20  

40  

60  

80  

100  

120  

SICI 2 SICI 4 SICI 2 SICI 4

HC SZ

Baseline

Day-28

A.

B.

Diagnosis and Interstimulus interval

A.

B.

Diagnosis and Interstimulus interval

Cor

tical

Inhi

bitio

n (%

ME

P re

duct

ion)

C

ortic

al In

hibi

tion

(%M

EP

redu

ctio

n)

Page 87: Evaluating The Effects of Cannabis Dependence and ...

74  

following 28 days of abstinence separately in patients and controls who successfully

abstained (figure 3.10 A, 3.11A) and those who relapsed (figure 3.10 B, 3.11 B).

             

  Figure 3.10. Longitudinal data comparing long interval cortical inhibition (LICI) between patients (SZ) and healthy controls (HC) across 100 ms (LICI100), 150 ms (LICI150) and 200 (LICI200) at baseline and following 28-days of abstinence. A represents a graph of abstainer HC and SZ, B represents a graph of relapser HC and SZ. Bars represent (±) 1 standard deviation.

-­‐100  

-­‐50  

0  

50  

100  

150  

LICI 100 LICI 150 LICI 200 LICI 100 LICI 159 LICI 200

HC SZ

Baseline

Day-28

-­‐150  

-­‐100  

-­‐50  

0  

50  

100  

150  

LICI 100 LICI 150 LICI 200 LICI 100 LICI 159 LICI 200

HC SZ

Baseline

Day-28

A.

B.

Diagnosis and Interstimulus interval

Diagnosis and Interstimulus interval

Cor

tical

Inhi

bitio

n (%

ME

P re

duct

ion)

C

ortic

al In

hibi

tion

(%M

EP

redu

ctio

n)

Page 88: Evaluating The Effects of Cannabis Dependence and ...

75  

 

 

Figure 3.11. Longitudinal data comparing cortical silent period (CSP) between patients (SZ) and healthy controls at baseline and following 28-days of abstinence. A represents a graph of abstainer HC and SZ, B represents a graph of relapser HC and SZ. Bars represent (±) 1 standard deviation.

3.2.1.2 Longitudinal Excitatory Assessment: ICF

We also evaluated the potential longitudinal changes in NMDA excitatory activity

through ICF. We compared the enhancement in cortical excitation at baseline and following

28 days of abstinence in patients with schizophrenia and healthy controls. We separately

0  

0.05  

0.1  

0.15  

0.2  

0.25  

CSP CSP

HC SZ

Baseline

Day-28

0  

0.05  

0.1  

0.15  

0.2  

0.25  

CSP CSP

HC SZ

Baseline

Day-28

B.

A.

Cor

tical

Inhi

bitio

n (s

econ

ds)

Cor

tical

Inhi

bitio

n (s

econ

ds)

Cor

tical

Sile

nt P

erio

d D

urat

ion

(sec

onds

)

Cor

tical

Sile

nt P

erio

d D

urat

ion

(sec

onds

)

Page 89: Evaluating The Effects of Cannabis Dependence and ...

76  

graphed patients and controls who successfully abstained (figure 3.12 A) from those who

relapsed (figure 3.12 B).

Figure 3.12. Longitudinal data comparing intracortical faciliatation ICF) between patients (SZ) and healthy controls (HC) across 10 ms (ICF10), 15 ms (ICF15) and 20 ms (ICF20) at baseline and following 28-day abstinence. A represents a graph of abstainer HC and SZ, B represents a graph of relapser HC and SZ. Bars represent (±) 1 standard deviation.

0  

50  

100  

150  

200  

250  

ICF 10 ICF 15 ICF 20 ICF 10 ICF 15 ICF 20

HC SCZ

Baseline

Day-28

0  

50  

100  

150  

200  

250  

300  

ICF 10 ICF 15 ICF 20 ICF 10 ICF 15 ICF 20

HC SZ

Baseline

Day-28

A.

B. Diagnosis and Interstimulus interval

Diagnosis and Interstimulus interval

Cor

tical

Fac

ilita

tion

(%M

EP

enh

ance

men

t) C

ortic

al F

acili

tatio

n (%

ME

P e

nhan

cem

ent)

Page 90: Evaluating The Effects of Cannabis Dependence and ...

77  

3.2.2 Longitudinal Working Memory Assessments Ten patients with schizophrenia and 13 controls participated in the longitudinal TMS

testing session. Of the patients who participated, 6 successfully abstained while 4 relapsed,

and for the controls, 8 successfully abstained and 5 relapsed (see Table 3.5).

Table 3.6 Post WM demographics, clinical means, and (±) 1 standard deviations by diagnosis

_________________________________________________________________________ HC abst HC non-abst SZ abst SZ non-abst

(N = 8) (N = 5) (N = 6) (N = 4) . Age (years) 26.25 ± 5.20 31.2 ± 5.63 32.2± 11.50 33.5 ± 11.79 Race Caucasian 87.5% (7) 66.7% (2) 16.7% (1) 50% (1) skskdn African American - 33.3% (1) 50% (3) 50% (1) Other 33.3% (1) - 16.7% (1) - Education (years) 12.5 ± 3.35 12.8 ± 2.17 12.90 ± 1.34 10.75 ± .957 IQ 105.88 ± 8.03 97.8 ± 9.04 90.6 ± 10.57 92.25 ± 6.24 PANSS positive - - 14.0 ± 3.16 14.5 ± 4.04 negative - - 10.4 ± 2.79 11.5 ± 1.91 general - - 28.0 ± 4.95 26.25 ± 5.74 total - - 55.4 ± 10.26 52.25 ± 11.44 CPZ equivalents (mg/day) - - 651.33 ± 238.1 412.5 ± 287.2 CPD 10.93 ± 11.89 9.03 ± 5.54 10.77 ± 8.66 18.0 ± 4.0 GPD 1.24 ± .998 2.39 ± 1.37 1.13± .869 2.17 ± .98 Joint years 7.03 ± 5.35 8.99 ± 4.08 11.81 ± 12.07 10.97 ± 5.33 __________________________________________________________________________

Values are expressed as mean ± SD HC = healthy controls and SZ = patients with schizophrenia Abst = abstainer and non-abst = relapse CPZ = chlorpromazine, CPD = cigarettes per day, GPD = grams per day

Page 91: Evaluating The Effects of Cannabis Dependence and ...

78  

3.2.2.1 Longitudinal Working Memory Performance

The N-back task was administered at baseline and following 28 days of cannabis

abstinence in patient and control abstainers and relapsers. Accuracy for target correct

responses on the 1 and 3-back conditions were graphed separately.

                                   

 

Figure 3.13. Longitudinal N-back performance between patients (SZ) and healthy controls (HC) abstainers and relapsers at baseline and following 28-day abstinence on the 1-back and 3-back conditions. Bars represent (±) 1 standard deviation

0  

20  

40  

60  

80  

100  

120  

Abstainer Relapser Abstainer Relapser

HC SZ

Baseline

Day-28

0  

20  

40  

60  

80  

100  

120  

Abstainer   Relapser     Abstainer   Relapser    

HC   SZ  

Baseline  

Day-­‐28  

Mea

n Pe

rcen

tage

of

Tar

get

Cor

rect

R

espo

nses

1-Back

3-Back

5 Second Time Window, Mastoid Reference

Group

Group

Mea

n Ta

rget

Cor

rect

Res

pons

e (%

)

Mea

n Ta

rget

Cor

rect

Res

pons

e (%

)

Page 92: Evaluating The Effects of Cannabis Dependence and ...

79  

3.2.2.2 Longitudinal Gamma Power

Mean gamma oscillatory power was measured at baseline and following abstinence in

patient and control abstainers and relapsers. Topographs illustrated the mean gamma power

elicited during the 1- and 3-back conditions.

Mean Gamma 1-Back (µV2)

HC

SZ

Baseline Abstinence Relapse

Mean Gamma 3-Back (µV2)

HC

SZ

Baseline Abstinence Relapse

Figure 3.14. Topographical illustration of mean gamma power elicited during 1- and 3-back in patients (SZ) and healthy control (HC). Included are topo plots at baseline and following a 28-day abstinence period in abstainers and relapsers. Greater gamma power is represented by hot or red colours.

N = 13 N = 8 N =5

Basel Abstinent Relapse

5 Second Time Window, Mastoid Reference N = 10 N = 6 N =4

N = 13 N = 8 N =5

N = 10 N = 6 N =4

Page 93: Evaluating The Effects of Cannabis Dependence and ...

80  

Chapter 4 Discussion

4.1 General Discussion

Given the high rates of cannabis abuse and dependence among patients with

schizophrenia and the detrimental effects of cannabis on illness onset, prognosis and

treatment success (D'Souza et al., 2005; Foti et al., 2010; Linszen et al., 1994; Manrique-

Garcia et al., 2014), uncovering the mechanism underlying this common co-morbidity is of

great value. As such, the aim of this study was to investigate the neurophysiological effects

of cannabis dependence in patients with schizophrenia and non-psychiatric controls. To our

knowledge, this was the first study to address the complex relationship between cannabis,

cognition, and cortical inhibition in patients with schizophrenia using a within- and between-

subjects longitudinal design. Given that cannabis inhibits GABAergic neurotransmission,

which is necessary for both cortical inhibition and working memory, we hypothesized that

cannabis dependence would further impair GABAergic deficits present in patients with

schizophrenia. We assessed GABA inhibitory neurotransmission through several well-

established TMS paradigms as well as indirectly through working memory performance and

gamma oscillatory activity. As an exploratory secondary analysis, we evaluated longitudinal

changes in cortical inhibition and working memory following a 28-day cannabis abstinence

period. We hypothesized that cortical inhibition and working memory deficits would be

potentiated with cannabis abstinence in both patients and non-psychiatric controls.

The main finding in the current study was greater GABAB mediated inhibitory

neurotransmission, as measured by CSP, in cannabis dependent patients compared to

dependent healthy controls. In regards to SICI, while no significant effect of diagnosis was

Page 94: Evaluating The Effects of Cannabis Dependence and ...

81  

found, due in part to the large within-group variability, our results suggest that cannabis

dependent patients may have more GABAA mediated inhibition compared to controls. No

significant differences were found on LICI or on our measures of cortical excitation,

including ICF and RMT. With regards to our working memory findings, results suggest that

patient and control cannabis users perform similarly on the N-back task, along with

comparable levels of frontal gamma oscillatory activity. Taken together, these results suggest

that cannabis dependence has differential effects on cortical inhibition in controls compared

to patients with schizophrenia. While these contradict our initial hypotheses, this preliminary

research provides novel insight into the mechanisms underlying co-morbid cannabis use

among individuals with schizophrenia.

4.1.1 Study Sample

In the current study, our patient group demonstrated a significantly lower IQ and

fewer years of education as compared to the control group. This finding is in accordance with

previous cross-sectional studies both in patients with schizophrenia (Kremen, Seidman,

Faraone, & Tsuang, 2001) and in chronic cannabis users (Arseneault, Cannon, Witton, &

Murray, 2004); therefore, this sample seems to be representative of the target population.

Additionally, all participants were male. While this does limit the generalizability of the

study, cannabis use has been shown to be especially common in young males experiencing

their first-episode of psychosis (Koskinen et al., 2010).

Of note, this was one of the first studies conducted in cannabis dependent individuals,

to employ a longitudinal abstinence paradigm, with twice-weekly urinalysis. Additionally,

we accounted for important confounding variables associated with substance use. This

Page 95: Evaluating The Effects of Cannabis Dependence and ...

82  

included, frequency and recency of use as well as the amount of money spent weekly on

cannabis, which is thought to indirectly index of the quality of cannabis consumed.

Furthermore, unlike previous research in this field, we implemented a clear definition of

cannabis dependence, whereby all participants had to be daily cannabis users for a minimum

of 1 year. Additionally, participants had to meet DSM-IV criteria for cannabis dependence

and could not currently consume or meet dependence for, any additional illicit substances

within the past 6 months. Finally, we also controlled for co-morbid cigarette use unlike

previous research, given its common co-occurrence in this population (Kalman et al., 2005;

Patton et al., 2006). Importantly, daily cannabis and cigarette consumption did not differ

between controls and patients.

4.1.2 TMS Findings

The main finding in the current study was increased GABAB mediated inhibitory

control, as measured by CSP, in cannabis dependent patients compared to dependent healthy

controls. This finding contradicts the previous two studies investigating the effects of

cannabis use on cortical inhibition in first episode patients with schizophrenia (Wobrock et

al., 2010) and controls (Fitzgerald et al., 2009), as neither study found significant differences

on this measure. CSP is considered to be one of the more robust and consistent TMS

measures; however recent observations have brought to light large variability between

studies in terms of the effects of schizophrenia on CSP. For example, early research

demonstrated GABAB specific CSP deficits in patients with schizophrenia (Eichhammer et

al., 2004; Fitzgerald, Brown, et al., 2002a). However, consistent with the current study, more

recent research has found a lengthening of the CSP length in patients with schizophrenia

(Hasan et al., 2012; Strube et al., 2014; Wobrock et al., 2009).

Page 96: Evaluating The Effects of Cannabis Dependence and ...

83  

In comparison to previously published CSP values, our cannabis dependent controls

and patients displayed both longer than “average” (Hasan et al., 2012) and shorter than

“average” (Strube et al., 2014) CSP lengths. Therefore, in the context of previous research,

this suggests that no clear unidirectional comparisons regarding the effects of cannabis use

on cortical silent period in patients or controls can be drawn. Methodological differences,

including severity and duration of illness, stimulation intensity and the effects of medication,

likely contribute to these divergent findings. For example, stimulation intensity in both of

these previous studies was set to the participant’s 1mV (which is typically 120% of their

RMT), whereas in the current study, stimulation intensity was set to 140% of the RMT

(Hasan et al., 2012; Strube et al., 2014). Additionally, clozapine has been shown to

significantly lengthen CSP in patients with schizophrenia (Daskalakis, Christensen, et al.,

2008). In the current study, we included one patient who was treated with clozapine, and

given that no outliers were removed from analysis, we cannot rule out the possibility that this

participant is driving the current finding. In summary, our results suggest that cannabis

dependent patients with schizophrenia exhibit greater GABAB mediated inhibition compared

to cannabis dependent controls.

Regarding our SICI findings, unlike previous research and contrary to our hypothesis,

we did not find reduced SICI in cannabis dependent patients. In fact, with a trend towards a

significant diagnosis effect, as well as a large effect size, our results suggest the opposite

effect of cannabis on GABAA mediated cortical inhibition, with enhanced inhibition in

patients. This large effect size yet lack of statistical significance is likely due to small sample

sizes and large variability within groups. Interestingly, when compared to the heavy using

dependent controls in Fitzgerald’s study, our current control group demonstrated similarly

Page 97: Evaluating The Effects of Cannabis Dependence and ...

84  

low levels of GABAA mediated cortical inhibition (Fitzgerald et al., 2009). Additionally,

when compared to previously published values regarding the percentage of inhibition in

Fitzgerald’s healthy, non-cannabis using sample, controls in the current study demonstrate

lower levels of inhibition (Fitzgerald et al., 2009).

In contrast, when compared to Wobrock’s previously published SICI findings in first-

episode cannabis-using patients, cannabis dependent patients in the current study

demonstrated a greater amount of cortical inhibition. Similarly, when compared to published

findings in both recent onset and chronically ill non–using patients with schizophrenia

(Strube et al., 2014), cannabis dependent patients in the current study also demonstrated more

cortical inhibition. Of note, these comparisons must be interpreted with caution, given that

they were made across studies, which introduces a number of uncontrolled variables. For

example, in Wobrock’s study, dependence was assessed with the European Addiction

Severity Index as well as the DSM-IV criteria of substance abuse/dependence, however,

these cannabis users only had to use weekly and more than 20 times in their lifetime. Given

such divergence in cannabis dependence criteria, it is not possible to draw direct comparisons

regarding cortical inhibitory results from the current study with that of the previous findings.

In the context of previously published data (Fitzgerald et al., 2009; Strube et al.,

2014; Wobrock et al., 2010), these SICI results suggest that cannabis may reduce GABAA

inhibition in controls whereas cannabis may have the opposite effect on inhibition in patients.

In support of this, our group recently demonstrated this finding with preliminary data. This

data showed that cannabis using patients had more cortical inhibition as measured by SICI

compared to non-using patients, whereas control cannabis users demonstrated significantly

Page 98: Evaluating The Effects of Cannabis Dependence and ...

85  

less inhibition as compared to non-using controls (Goodman, 2015). While the previous

studies mentioned above all assessed SICI, these findings must be considered with caution,

given that differences within these SICI paradigms, participant pool and study designs likely

exist. Thus, these comparisons go beyond the scope of the current project.

In line with previous findings in cannabis using controls compared to non-users

(Fitzgerald et al., 2009) as well as in medicated and unmedicated patients compared to

controls (Fitzgerald et al., 2003), no LICI diagnostic differences were revealed in the current

study. Given that both LICI and CSP are thought to index GABAB mediated inhibition, and

significant differences in CSP were observed in the current study, it would suggest that LICI

might not be sensitive enough to detect these potential diagnostic differences. These

divergent effects may also be due to the differential mechanisms underling these TMS

paradigms. For example, CSP assesses the duration of inhibition, whereas LICI indexes the

magnitude of inhibition at a single time point (McDonnell, Orekhov, & Ziemann, 2006).

Furthermore, CSP has been shown to be associated with inhibition at the level of the spinal

cord, whereas long interval inhibition may take place more so at the level of the cortex

(Chen, Lozano, & Ashby, 1999).

While no significant diagnostic differences were observed on measures of cortical

excitation including RMT and ICF, a large effect size at 20 milliseconds for ICF was

revealed. The previous study conducted by Wobrock and colleagues (2010) found enhanced

ICF in cannabis dependent first-episode patients with schizophrenia (Wobrock et al., 2010).

In the current study, we found a trend towards higher facilitation in the cannabis dependent

patient group, although these differences were not significant. It has been suggested that

enhanced ICF in these patients might best be explained by an increased glutamatergic input

Page 99: Evaluating The Effects of Cannabis Dependence and ...

86  

along with disturbances in cholinergic intracortical neuronal circuits (Wobrock et al., 2010).

While no study has looked specifically at the effects of chronic cannabis use on cortical

facilitation, previous research has shown that nicotine, alcohol and cocaine users demonstrate

cortical hypoexcitability (Barr et al., 2008; Boutros et al., 2005; Conte et al., 2008; Lang,

Hasan, Sueske, Paulus, & Nitsche, 2008). Given that these previous studies were conducted

in otherwise healthy controls, we cannot rule out the possibility that these divergent findings

may be due, in part, to the underlying brain abnormalities in NMDA receptor functioning

seen in patients with schizophrenia (Falkai, Wobrock, Schneider-Axmann, & Gruber, 2008).

Additionally, several of these previous studies only examined the acute drug effects on

cortical facilitation and did not employ the appropriate abstinence period, thus testing

participants while intoxicated. As such, it is clear that continued research investigating the

effects of cannabis use on cortical excitability is necessary.

In summary, these findings suggest that cannabis has differential effects in controls

and patients in terms of GABAB inhibitory functioning, demonstrated through a significantly

longer CSP in patients. The large effect sizes and trends towards significant diagnostic

effects on measures of GABAA and NMDA, suggest that with a larger sample size, these

differences may become more evident. While these findings oppose our initial hypotheses,

they depict an interesting picture of the neurophysiological effects of cannabis dependence

among patients with schizophrenia. Given the paucity of research in this field, it is clear that

further research confirming this finding is necessary.

Page 100: Evaluating The Effects of Cannabis Dependence and ...

87  

4.1.3 Working Memory Findings

Contrary to our initial hypothesis, cannabis dependent patients with schizophrenia

performed as well as cannabis dependent controls on both the 1 and 3-back conditions of the

N-back verbal working memory task. Previous research has suggested that patients with

schizophrenia consistently perform worse than controls on working memory tasks. In fact,

working memory deficits are among the most replicable and robust findings in this

population (Forbes et al., 2009). Comparisons have been drawn between the cognitive

impairments seen in patients with schizophrenia with that of cannabis users; these cognitive

domains included attention, learning and inhibition (Radhakrishnan et al., 2014; Solowij &

Michie, 2007). Thus it would follow that cannabis use among patients with schizophrenia

would exacerbate already present working memory deficits. However, there is a growing

body of evidence suggesting that chronic cannabis users with schizophrenia perform better

on memory tasks than patients with no history of drug use (Rabin et al., 2011; Schnell et al.,

2009; Stirling et al., 2005; Yucel et al., 2012). Thus, in order to determine exactly how

cannabis dependence impacts working memory deficits in patients with schizophrenia, the

current sample must be compared to a non-dependent patient sample.

Current research has suggested that under low working memory load conditions (i.e.,

the 1-back), accuracy in patients and controls is comparable; however, increased memory

load was associated with a deterioration in patients’ performance (Barr et al., 2013; Carter et

al., 1998). Unfortunately, few studies have administered the most challenging 3-back

condition to patients with schizophrenia. In one study conducted by Barr et al. (2013),

researchers aimed to evaluate the effects of a non-invasive brain stimulation technique called

repetitive transcranial magnetic stimulation (rTMS) on working memory performance in

Page 101: Evaluating The Effects of Cannabis Dependence and ...

88  

schizophrenia patients. Prior to and following sham stimulation, 3-back accuracy slightly

decreased from 36 to 33 percent (Barr et al., 2013). In an earlier study conducted by Jansma

et al. (2004), researchers found that half of their patient sample performed near chance on the

3-back condition, with scores lower than 35 percent (Jansma, Ramsey, van der Wee, & Kahn,

2004). In contrast, cannabis dependent patients in the current study scored above average at

82 percent on the 1-back and 44 percent on the 3-back. It should be noted that in the current

study, no outliers were removed, thus we included one extreme outlier who performed well

below chance on the 3-back condition. Excluding this patient’s data increased 3-back

performance to 49 percent. Taken together, these findings suggest that cannabis dependent

patients may perform better on the 3-back condition than non-using patients.

Similarly to the cognitive deficits observed in non-using patients with schizophrenia,

converging neuroimaging, neurobiological and longitudinal research has suggested that

memory deficits persist in otherwise healthy individuals who use cannabis at an early age,

frequently, and over a long period of time (Becker et al., 2010; Lopez-Larson et al., 2011;

Mehmedic et al., 2010; Yucel et al., 2010). To our knowledge, only one study has

administered the 3-back condition in cannabis using non-psychiatric controls. In this previous

study, N-back accuracy was compared among daily cannabis users (>7 joints per week for a

minimum duration of 3 years) (Verdejo-Garcia et al., 2013). Unlike the current study,

researchers reported probability of hit (i.e., number of hits/number of hits+number of

misses). The 1-back probability of hit was 91 percent in cannabis users, while their 3-back

probability of hit was significantly lower in at 61 percent. These scores were similar to, yet

slightly better than that of the cannabis dependent controls in the current study, who scored

an 89 percent on the 1-back and a 54 percent on the 3-back. It should be noted that the

Page 102: Evaluating The Effects of Cannabis Dependence and ...

89  

accuracy from these two studies cannot be directly compared, given the different methods of

reporting performance (i.e., % accuracy versus probability of hit) and number of available

target correct responses. Unlike the current study, the cannabis-using group had to abstain for

72 hours, which coincides with the height of cannabis withdrawal symptoms (Budney,

Moore, Vandrey, & Hughes, 2003); however, these withdrawal symptoms were medically

controlled (Verdejo-Garcia et al., 2013). Thus these slight differences in performance could

be due to the period of cannabis abstinence, as well as the divergent methods of reporting

performance.

Compared to non-using controls, cannabis dependent controls in the current study

seemed to performe worse on the more challenging 3-back condition. In healthy controls, 3-

back accuracy has been shown to range from above 80% (Prilipko et al., 2011; Sanchez-

Carrion et al., 2008) to approximately 70% (Barr et al., 2010; Rose & Ebmeier, 2006).

Therefore, these results suggest that cannabis dependence in healthy controls impairs

working memory performance as compared to non-using controls. This phenomenon is

generally supported in the literature, whereby memory deficits persist beyond acute

intoxication in chronic users (for full review see (Schoeler & Bhattacharyya, 2013)).

Regarding the effects of cannabis dependence on baseline gamma oscillatory activity,

we initially hypothesized that cannabis dependence among patients would further exacerbate

their excessive gamma activity. However, our results revealed that patients and controls

exhibited similarly high level of gamma activity in the prefrontal cortex. One study

conducted by our group revealed excessive oscillations in patients compared to controls

during the 3-back condition (Barr et al., 2010). The researchers suggested that this finding

may be due in part to GABAergic deficits, specifically through the action of GABAB,

Page 103: Evaluating The Effects of Cannabis Dependence and ...

90  

ultimately leading to a lack of inhibition of gamma oscillations (Farzan et al., 2010a). In the

current study, a similar lack of modulation was observed in both controls and patients, given

that gamma oscillatory activity was as high in the 1-back condition as the 3-back. This

suggests that cannabis may have similar underlying impairing effects on neural oscillations

as those seen in patients with schizophrenia. Specifically this lack of gamma modulation may

be due, in part to GABAB dysfunction, which was observed among our control cannabis

using participants.

Similarly to the excessive oscillatory activity among patients, cannabis use has been

shown to increase gamma oscillations in healthy controls. The CB1R receptor is primarily

located on GABAergic inhibitory interneurons, and exogenous activation of these receptors

through THC, for example, suppresses GABA (Freund et al., 2003). Thus, it has been

suggested that cannabis may increase neuronal synchrony similarly to that of a GABA

receptor antagonist. These antagonists have been shown to lead to convulsive seizures

(Steriade, Amzica, Neckelmann, & Timofeev, 1998), due to excessive or hypersynchronous

neuronal activity in the brain (Shusterman & Troy, 2008). Moreover, models of epilepsy

utilize cannabinoids due to their pro-convulsant side effects (Clement, Hawkins, Lichtman, &

Cravatt, 2003). In line with this research, cannabis dependent controls in the current study

demonstrated increased levels of gamma oscillatory activity compared to previously

published results in non-using controls (Barr et al., 2010). Thus, it is possible that the lack of

diagnostic differences in gamma activity in the current study could be attributed to excessive

oscillations both in control cannabis users and patients with schizophrenia.

Thus taken together, these findings suggest that aberrant gamma oscillatory activity

may underlie cognitive deficits associated with both schizophrenia and cannabis use. Based

Page 104: Evaluating The Effects of Cannabis Dependence and ...

91  

on our working memory performance results, it may seem as though there are no differential

diagnostic effects of cannabis dependence, however the potential effects of cannabis become

clearer when compared to previously published 3-back accuracy data conducted in a group of

non-cannabis using patients and controls. As such, these findings suggest that cannabis use

may be more impairing in control users, where as cannabis dependent patients with

schizophrenia seem to performed better than non-using patients. Given that this is the first

study evaluating working memory performance combined with gamma activity in this

population, it is clear that further research is warranted.

4.1.4 Exploratory Findings To date, all studies that have investigated the effects of cannabis either on cortical

inhibition or working memory associated oscillatory activity in patients with schizophrenia

have employed cross-sectional designs. As such, the current study aimed to investigate the

longitudinal neurophysiological effects of cannabis throughout a 28-day cannabis abstinence

period. Employing an abstinence paradigm allows for the evaluation of the long-term effects

of cannabis, beyond that of acute intoxication and withdrawal. Given that cannabis likely

differentially modulates cortical functioning in patients and controls, we directly compared

these effects in cannabis dependent individuals with and without schizophrenia. Additionally,

within each of these groups, valuable information was also drawn from comparing those who

successfully abstained and those who relapsed.

In the current study, the small sample size limited our ability to draw conclusions

regarding the longitudinal effects of cannabis abstinence. This was especially true for our

TMS findings, given the large within group variability and lack of clear or consistent

inhibitory patterns. However, there is one interesting preliminary LICI finding in abstaining

Page 105: Evaluating The Effects of Cannabis Dependence and ...

92  

patients versus those who relapsed. Results revealed an increase in GABAB mediated

inhibition following 28-day abstinence in patients who successfully abstained. In contrast,

the reverse was found in patients who relapsed, whereby, on day 28, relapsers demonstrated a

decrease in inhibition. While it is an interesting finding, it is clear that continued research is

needed in order to validate this potential pattern with a greater number of participants.

Regarding the working memory findings, although subtle, results revealed a decrease

in accuracy on both the 1 and 3-back conditions among patient abstainers. In line with this

finding, these patients also demonstrated slight decreases in frontal gamma oscillatory

activity. While this finding may suggest that patient’s optimal level of gamma activity and

accuracy is present during acute cannabis abstinence (i.e. at baseline), continued research is

needed, as these findings are very preliminary. An additional interesting finding within the 3-

back condition in both patients and controls, was that those who successfully abstained from

cannabis performed better at baseline than those who went on to relapse (refer to figure

3.13). This may suggest that N-back accuracy, or more broadly cognitive functioning, may

predict one’s ability to successfully abstain from cannabis. While this relationship has never

been studied in cannabis users, in cigarette smokers with schizophrenia, poor working

memory performance has been shown to predict relapse (Dolan et al., 2004). These

preliminary findings highlight the need for continued research on the effects of both cannabis

dependence and abstinence in order to better understand the long-term neurophysiological

impact of cannabis on the brain.

4.1.5 Potential Mechanisms While the exact mechanisms underlying the effects of cannabis in individuals with

schizophrenia are not well understood, overlapping neurocognitive and neurophysiological

Page 106: Evaluating The Effects of Cannabis Dependence and ...

93  

research suggests that interferences in the homeostatic role of the eCB system may underlie

this common co-morbidity. The eCB system acts through synaptic retrograde signalling to

ultimately control the release of other neurotransmitters, including GABA, glutamate and

dopamine (Schlicker and Kathmann, 2001). Activation of the CB1 receptor, through either its

endogenous ligands anandamide and 2-AG, or exogenous cannabis use, mediates the

production and release of inhibitory and excitatory neurotransmitters key to cortical

inhibition, higher order cognitive functions and synaptic plasticity (i.e., learning and

memory) (Chevaleyre, Takahashi, & Castillo, 2006; Howlett et al., 2004; Piomelli, 2003).

Exposure to exogenous cannabinoids, especially during adolescence, has been shown to

significantly disrupt the protective effect of the eCB system (James, James, & Thwaites,

2013). Chronic cannabis use leading to the development of dependence and tolerance has

been shown to alter the functionality of the eCB system; however the exact neurochemical

and neurophysiological effects of chronic cannabis use are still largely unknown (Hoffman,

Oz, Caulder, & Lupica, 2003; Sim-Selley, 2003). Given that cannabis is known to suppress

GABAergic inhibitory neurotransmission (Iversen, 2003) we hypothesized that, cannabis

dependence would lead to deficits in cortical inhibition. In line with this hypothesis, cannabis

dependent controls in the current study demonstrated less GABAB mediated cortical

inhibition when compared to cannabis dependent patients with schizophrenia.

Interest in the involvement of the eCB system underlying cognitive processes was

motivated by evidence demonstrating that CB1 receptors are highly expressed in the

hippocampus (Herkenham, 1991), a brain region vital to learning and memory. Evidence has

consistently demonstrated that cannabinoids manipulate the various stages of learning and

memory, including acquisition, consolidation, and retrieval (Ilan et al., 2005; Silva de Melo

Page 107: Evaluating The Effects of Cannabis Dependence and ...

94  

et al., 2005). These behavioural effects are thought to be produced, in part, by an imbalance

in GABAergic neuronal transmission in the PFC (Pistis et al., 2002). As such, we

hypothesized that cannabis dependence in patients and controls would increase gamma

oscillatory activity and lead to poorer working memory performance. Based on previously

published 3-back accuracy results in healthy controls (Barr et al., 2010), we found that

cannabis dependent controls in the current study demonstrated lower working memory

accuracy as well as high frontal gamma oscillatory activity. Thus we propose that these

cortical inhibitory and working memory deficits seen in non-psychiatric cannabis dependent

controls are likely due to the suppressing effects of cannabis use on GABA inhibitory

neurotransmission.

Regarding the patient sample, we hypothesized that cannabis dependence would

further exacerbate GABAergic deficits, leading to aberrant cortical inhibition and poor

working memory performance. However, our results revealed that cannabis dependent

patients actually demonstrated improved cortical inhibition and similar working memory

performance to our control group. Thus this implies that chronic cannabis use does not

necessarily have the same neurophysiological effects in controls and patients and highlights

the complex relationship between schizophrenia and cannabis dependence. There are a

number of plausible reasons as to why we did not confirm our hypotheses. Firstly, there is

large body of evidence illustrating alterations in the eCB system in patients with

schizophrenia, regardless of cannabis consumption. For example, post-mortem studies in

patients demonstrated an increased density of CB1 receptors in DLPFC (Dean et al., 2001)

and anterior cingulate (Zavitsanou, Garrick, & Huang, 2004) as well as elevated levels of

anandamide in the cerebrospinal fluid (CSF) in acute schizophrenia (Giuffrida et al., 2004).

Page 108: Evaluating The Effects of Cannabis Dependence and ...

95  

Thus these pre-existing eCB alterations likely influence the “normal” effects of cannabis use

among patients.

Secondly, it is possible that patients develop a compensatory mechanism in response

to these underlying eCB system deficits. This may therefore account for the improved

cortical inhibition and working memory performance in cannabis dependent patients

observed in the current study. The existence of a compensatory mechanism in patients has

previously been suggested. For example, one study found that in the DLPFC of patients with

schizophrenia, GABAA receptors were up-regulated at pyramidal axons, which was thought

to develop in response to deficient GABA release from chandelier axon terminals (Jensen,

Chiu, Sokolova, Lester, & Mody, 2003). Researchers have interpreted this finding to

represent a compensatory response to GABA deficits (Lewis et al., 2005). Similarly, Bossong

and colleagues proposed that chronic cannabis use might lead to a compensatory mechanism,

involving the recruitment of regions not normally associated with a working memory task

(Bossong et al., 2014). Thus these findings suggest that chronic cannabis using patients

develop a GABA specific compensatory mechanism, which may in part, account for our

current unexpected findings in patients.

An additional potential reason as to why cannabis dependent patients demonstrated

better cortical inhibition and working memory performance compared to non-using patients

is drawn from social cognitive research. Several researchers have suggested that cannabis-

using patients with schizophrenia may represent a subset individuals with this disorder,

following the observation that these patients reliably performed better on cognitive tasks than

non-using patients (Rodriguez-Sanchez et al., 2010; Yucel et al., 2012). It has been suggested

that cannabis-using patients may be cognitively less vulnerable given that their onset of

Page 109: Evaluating The Effects of Cannabis Dependence and ...

96  

psychosis may not have developed without the initiation of cannabis use. These individuals

may therefore represent a ‘healthier’ group (Arndt, Tyrrell, Flaum, & Andreasen, 1992), who

are able to lead the complex life of a drug user (Joyal, Halle, Lapierre, & Hodgins, 2003;

Rodriguez-Sanchez et al., 2010). Given that these cannabis-using patients may have better

premorbid social and cognitive functioning, it is possible that they may not have as profound

GABAergic deficits as their non-using counterparts. In line with this hypothesis, preliminary

data from our group have demonstrated that cannabis dependent patients have better GABAA

mediated cortical inhibition than non-cannabis dependent patients (Goodman, 2015).

Therefore, the current findings of enhanced cortical inhibition and working memory

performance in cannabis dependent patients may provide preliminary support for the

hypothesis that patients who present with co-morbid cannabis use represent a subset of the

patient population with better premorbid social functioning may.

The fact that this GABAergic mechanism cannot adequately account for all of the

observed findings in patients and controls highlights the complex neurophysiological

interactions between mental illness and drug use. Of note, this current mechanism does not

take into consideration the widespread effects of cannabis on dopamine, glutamate, and other

neurotransmitters. Similar to the pathophysiological theories underlying schizophrenia,

deficits in one neurotransmitter alone may not sufficiently account for this complex co-

morbidity. Furthermore, the majority of neurochemical and neurophysiological research is

based on the acute effects of cannabis among patients with schizophrenia and many

unknowns still exist regarding the long-term effects of chronic cannabis use. Thus, without a

better understanding of these unknowns, we are limited in our interpretation of the

underlying neurophysiological and neurochemical mechanisms.

Page 110: Evaluating The Effects of Cannabis Dependence and ...

97  

4.2 Limitations These results should be interpreted in light of several limitations. One of the most

noticeable weaknesses in the current study was the small sample size. This was especially

true for our longitudinal analysis, given that our small sample was further subdivided into

abstainers and relapses. Nevertheless, the 3-back working memory load condition may

capture the divergent neurophysiological effects in abstainers versus relapsers with continued

testing. With such a small sample size, we chose not to remove outliers from the study,

which likely contributed to the large variability seen within our TMS measures. An

additional and significant limitation was the lack of a non cannabis-using control group, for

both our patients and controls. This lack of a true control group likely masked the true

neurophysiological effects of cannabis use.

Although our patient and control groups were well matched in terms of age, cigarette

use as well as daily and lifetime cannabis use, there were several limitations within our

sample. Firstly, the study included only male participants, thus reducing the generalizability

of our findings. A key reason why only males were selected for this study was due to

previous research suggesting that cannabis is especially common in young males

experiencing their first-episode of psychosis (Koskinen et al., 2010). Thus, these findings

highlight the difficulties in recruiting cannabis dependent female patients. Further differences

in our sample were observed in the breakdown of racial backgrounds between our two

groups, with a higher percentage of Caucasian controls and non-Caucasian patients. While

these differences were not significant, it is possible that cannabis metabolism may be

influenced by race. To date, no studies have looked into this specifically in cannabis users

however, research has suggested that the pharmacodynamics and pharmacokinetics of other

Page 111: Evaluating The Effects of Cannabis Dependence and ...

98  

drugs vary based on race (Chen, 2006).

Secondly, while we collected data regarding current and past patterns of cannabis use,

we did not differentiate between the strains of cannabis nor did we chemically verify the

potency of cannabis used by our participants. It has been well-documented that the

neuropharmacological effects of cannabis vary widely across the different strains due to the

varying ratios of the cannabinoid constituents (Potter, Clark, & Brown, 2008). It should be

noted, however, that other than the joint year and IQ negative correlation, we did not find a

significant correlation between the TMS or working memory assessments and cannabis

consumption, including grams per day and joint years.

Thirdly, it should be considered that while participants were excluded if they met

criteria for other substance abuse or dependence 6 months prior to testing, several

participants had a past diagnosis of an alcohol or substance use disorder. Lifetime substance

dependence has been shown to have long-term effects on neurocognition (Potter, Downey, &

Stough, 2013; Verdejo-Garcia, Bechara, Recknor, & Perez-Garcia, 2006) and cortical

functioning (Bolla et al., 2003; Ersche et al., 2005) even following the complete elimination

of the substance from the body. Additionally, all participants were current cigarette users,

given that nicotine dependence commonly co-occurs in cannabis users (Patton et al., 2006)

and patients with schizophrenia (Kalman et al., 2005; Lasser et al., 2000). In patients,

nicotine use is known to selectively enhance neurocognitive function (Sacco et al., 2005),

thus the presence of nicotine in our participants likely confounds our working memory

performance results and potentially our neurophysiological findings. Importantly, in the

current study, the frequency of past substance dependence and daily cigarette use did not

significantly differ between patients and controls. However, in order to truly elucidate the

Page 112: Evaluating The Effects of Cannabis Dependence and ...

99  

effects of cannabis, future studies should better control for these confounding variables.

Additionally, in the patient group specifically, it should be noted that antipsychotic

medications pose as a considerable confounding factor (Davey et al., 1997). Differences in

RMT have been found in patients treated with risperidone compared to those on olanzapine

(Fitzgerald, Brown, Daskalakis, & Kulkarni, 2002b). Similarly, for paired-pulse

measurements, studies have shown that clozapine significantly lengthens CSP (Daskalakis,

Christensen, et al., 2008) where as benzodiazepines alter GABA specific TMS paradigms (Di

Lazzaro et al., 2008). Furthermore, these medications likely influenced our working memory

findings. For example, studies have shown enhanced 40 Hz oscillatory activity (Hong et al.,

2004) and cognitive performance (Bilder et al., 2002; Meltzer & McGurk, 1999; Sharma,

Hughes, Soni, & Kumari, 2003) in patients on second-generation antipsychotics. In response

to these findings, we did not include any patient on benzodiazepines for the TMS portion of

the study; however, participants on a variety of antipsychotics were included and therefore

pose as a limitation to the current study.

One final limitation of the current study was the task used to assess working memory

functioning. The N-back task does not allow for the evaluation of specific working memory

subprocesses and previous research has demonstrated oscillatory alterations within these

subprocesses (Haenschel et al., 2009). Therefore, we were unable to determine if cannabis

differentially modulated the encoding, maintenance, manipulation, or retrieval stages of

memory. Additionally, repeated working memory and EEG testing has been shown to be

influenced by within-subject variability. A recent study revealed that while the overall score,

cortical activation, and alertness, showed long-term stability within subjects, these measures

were sensitive at the level of the individual. Cognitive function generally varied due to

Page 113: Evaluating The Effects of Cannabis Dependence and ...

100  

within-day fluctuations including alcohol, marijuana and caffeine use; these fluctuations

were greatest when testing sessions took place in the afternoon (Gevins et al., 2012). While

participants in the current study were instructed not to use alcohol, caffeine, or marijuana on

the testing day, these factors as well as variability within testing times likely influence the

current findings.

4.3 Conclusions To our knowledge, this was the first study to address the complex relationship

between cannabis, cognition, and cortical inhibition in cannabis dependent patients with

schizophrenia using a within- and between-subjects longitudinal design. We hypothesized

that patients would demonstrate reduced cortical inhibition and excitation indexed through

SICI, LICI and ICF measures, as well as a shorter silent period, as assessed through CSP. We

also hypothesized that patients would demonstrate poorer working memory performance

compared to controls as well as excessive gamma oscillatory activity specifically in the

frontal region in patients with schizophrenia. Finally, in regards to our longitudinal data, we

hypothesized that following abstinence, both patients and controls would exhibit improved

cortical inhibition and working memory performance, with the greatest improvements in

patients.

However, contrary to previous findings (Fitzgerald et al., 2009; Wobrock et al., 2010)

at baseline, cannabis dependent patients demonstrated significantly greater GABAB function,

as measured by CSP. Additionally in the current study, no diagnostic differences were found

on baseline measures of working memory accuracy or gamma oscillatory activity. When

compared to a non-using sample, our current dependent controls performed worse on the 3-

back condition of the N-back whereas cannabis dependent patients performed better than

Page 114: Evaluating The Effects of Cannabis Dependence and ...

101  

non-using patients. Taken together, these findings would suggest that cannabis dependence

might have more impairing effects, specifically on GABAB mediated inhibition, in controls

compared to patients.

Given that cannabis reduces GABA function, and GABAergic neurotransmission

underlies both cortical inhibition and cognition, it follows that cannabis dependence would

lead to deficits in these domains. This pattern was clearly observed in our control group.

However, this mechanism cannot account for our findings in cannabis dependent patients.

Converging evidence suggests that cannabis-using patients may represent a subset of

schizophrenia patients with better premorbid cognitive and social functioning, and therefore,

potentially less impaired cortical inhibition. It is clear that continued research is necessary in

order to better understand the mechanism underlying this common co-morbidity to work

towards developing more specialized treatment approaches.

4.4 Future Directions Given that this is the first study evaluating the effects of cannabis dependence and

abstinence on cortical inhibition and working memory in patients with schizophrenia, it is

clear that future investigation is needed in order to address the aforementioned limitations.

Other than our CSP findings, the baseline analysis did not yield statistically significant

differences between controls and patients, suggesting that continued research is needed in

order to elucidate the potential differential effects of cannabis. This also highlights the

heterogeneity of both schizophrenia (Davidson & McGlashan, 1997) and cannabis use

(Iversen, 2003). Given the large interindividual and intraindividual variation over time in this

population, much larger samples may be needed in order to elucidate group differences.

Furthermore, future research would benefit from enlisting a true control group of non-using

Page 115: Evaluating The Effects of Cannabis Dependence and ...

102  

patients or controls. This four-way comparison would provide additional insight into the

differential effects of cannabis use on patients with schizophrenia and non-psychiatric

controls.

The current study sought to investigate the effects of the cannabis-psychosis co-

morbidity primarily on GABAergic functioning, however, both cannabis and schizophrenia

are known to influence a range of neurotransmitters, especially dopamine. For example, CB1

activation through THC is known to enhance mesolimbic dopaminergic neurotransmission.

Converging clinical and preclinical research has implicated this increase in dopamine in both

the cognitive impairments (Pistis et al., 2002; Pistis et al., 2001) and reinforcing properties

associated with cannabis use (Ameri, 1999; Bossong et al., 2009). While PET studies

investigating the effects of chronic and recreational cannabis use exist, studies focusing on

the long-term neurophysiological effects in abstinent users are limited. Thus, given the

widespread neurochemical effects of both cannabis use and schizophrenia, in order to gain a

true understanding of the neurophysiological effects in this population, future research should

take into consideration additional neurotransmitter systems.

Similarly, future research should also focus on additional oscillatory frequencies

including theta gamma coupling, which has also been implicated in working memory

functioning (Canolty et al., 2006). There is a paucity of research investigating the

functionality of theta-gamma coupling in patients with schizophrenia, and to our knowledge,

this topic has not been investigated in cannabis users. In the current study, we chose the

prefrontal cortex as the optimal site to investigate gamma oscillatory activity from, given its

involvement in working memory processes. However, neuroimaging studies have shown that

working memory functioning relies on a complex network spanning frontal and posterior

Page 116: Evaluating The Effects of Cannabis Dependence and ...

103  

parietal areas (Honey et al., 2002). Research has suggested that the prefrontal cortex may be

involved in retrieval whereas the posterior regions are associated with information storage

(Curtis & D'Esposito, 2003). Unfortunately, the N-back task used in the current study does

not allow for the investigation of the distinct subprocesses associated with working memory

functioning. Taken together, this highlights the need for future research examining additional

oscillatory frequencies spanning the frontal and parietal working memory networks. In order

to better understand the exact influence of cannabis and schizophrenia on working memory,

future studies should employ a memory task that allows for such assessments.

An additional and extremely important future direction would be to assess cortical

inhibition from the DLPFC, given its direct involvement in the pathophysiology of

schizophrenia (Barch et al., 2001) and evidence revealing eCB alterations in this region in

patients (Dean et al., 2001). Moreover, many of the cognitive side effects associated with

cannabis use are thought to be derived from CB1 receptors in the DLPFC. Recent

technological advances have allowed for the combination of TMS with EEG. Using a

pharmaco-TMS-EEG approach, researchers have been able to study the direct role of

GABAergic neurotransmission through TMS-evoked EEG potentials (Premoli et al., 2014).

Therefore, this technique would likely provide valuable insight into the mechanism

underlying co-morbid cannabis use in patients with schizophrenia.

Future research would also benefit from employing longitudinal abstinence

paradigms, which not only provide insight into the long-term neurophysiological effects of

cannabis but also control for some of the confounding variables associated with between-

subject study designs. Employing a longitudinal design may provide clarification as to

whether the observed results were due to the residual effects of THC in the body, acute

Page 117: Evaluating The Effects of Cannabis Dependence and ...

104  

cannabis withdrawal, long-term cannabis consumption, or premorbid neurodevelopmental

dysfunction seen in patients with schizophrenia.

Furthermore, in order to understand its effects within complex co-morbid populations,

we must first better understand the neurochemical and neurophysiological impact associated

with cannabis use. Additionally, further research specifically focusing on the long-term

effects of chronic use, and early initiation of cannabis use is needed as the clinical

importance of cannabis research lies within this problematic pattern of use. Thus longitudinal

research should also focus more so on naturalistic substance use in the general population

rather than acute administration of THC using lab-based methodologies. While these

naturalistic studies introduce variability, in terms of the strain and pattern of cannabis used as

well as history of cannabis and other drug use, they also allow for more generalizable and

relevant findings. This variability within the cannabis using population highlights the fact

that cannabis research is still in its infancy and continued investigation is needed.

Elucidating the neurophysiological effects of cannabis dependence would provide

novel and valuable insight into this common co-morbidity. This field would benefit from

multidisciplinary research both neurophysiological and cognitive based, in order to

ultimately catalyze future research regarding intervention and treatment strategies. It is

evident that there is a need for biologically-based investigation of mechanisms underlying

this common co-morbidity. As such, more funding is needed in order to allow for a more

intense investigation in this area. While the findings from the current study are preliminary,

they provide a first step towards better understanding the impact of cannabis on patients with

schizophrenia. Whether continued research ultimately determines that cannabis use has more

impairing neurophysiological effects on patients with schizophrenia or seemingly positive

Page 118: Evaluating The Effects of Cannabis Dependence and ...

105  

effects, the findings will be of value. However, it is clear that given the widespread use of

cannabis among patients with schizophrenia, the known negative effects on illness course

and the lack of specific treatment approaches targeted at cannabis dependent patients, further

investigation if of great importance.

Page 119: Evaluating The Effects of Cannabis Dependence and ...

106  

Literature Cited Adams, I. B., & Martin, B. R. (1996). Cannabis: pharmacology and toxicology in animals and

humans. Addiction, 91(11), 1585-1614. Addington, J., & Duchak, V. (1997). Reasons for substance use in schizophrenia. Acta Psychiatr

Scand, 96(5), 329-333. Adlaf, E. M., Begin, P., and Sawka, E. (2005). Canadian Addiction Survey (CAS): A National survey

of Canadians' Use of Alcohol and Other Drugs: Prevalence of Use and Related Harms: Detailed report. Ottawa: Canadian Centre on Substance Abuse.

Agurell, S., Halldin, M., Lindgren, J. E., Ohlsson, A., Widman, M., Gillespie, H., & Hollister, L. (1986). Pharmacokinetics and metabolism of delta 1-tetrahydrocannabinol and other cannabinoids with emphasis on man. Pharmacol Rev, 38(1), 21-43.

Akbarian, S., & Huang, H. S. (2006). Molecular and cellular mechanisms of altered GAD1/GAD67 expression in schizophrenia and related disorders. Brain Res Rev, 52(2), 293-304.

Allsop, D. J., Copeland, J., Lintzeris, N., Dunlop, A. J., Montebello, M., Sadler, C., . . . McGregor, I. S. (2014). Nabiximols as an agonist replacement therapy during cannabis withdrawal: a randomized clinical trial. JAMA Psychiatry, 71(3), 281-291.

Ameri, A. (1999). The effects of cannabinoids on the brain. Prog Neurobiol, 58(4), 315-348. American Psychiatric Association. (2013). Diagnostic and statistical manual of mental disorders (5th

ed). Washington, DC: American Psychiatric Association: Author. Andreasen, N. C., Arndt, S., Alliger, R., Miller, D., & Flaum, M. (1995). Symptoms of schizophrenia.

Methods, meanings, and mechanisms. Arch Gen Psychiatry, 52(5), 341-351. APA. (2000). Treatment of patients with schizophrenia: practice guidelines for the treatment

of psychiatric disorders. Washington: . Arndt, S., Tyrrell, G., Flaum, M., & Andreasen, N. C. (1992). Comorbidity of substance abuse and

schizophrenia: the role of pre-morbid adjustment. Psychol Med, 22(2), 379-388. Arseneault, L., Cannon, M., Witton, J., & Murray, R. M. (2004). Causal association between cannabis

and psychosis: examination of the evidence. Br J Psychiatry, 184, 110-117. Asai, Y., Takano, A., Ito, H., Okubo, Y., Matsuura, M., Otsuka, A., . . . Suhara, T. (2008).

GABAA/Benzodiazepine receptor binding in patients with schizophrenia using [11C]Ro15-4513, a radioligand with relatively high affinity for alpha5 subunit. Schizophr Res, 99(1-3), 333-340.

Ashton, C. H. (1999). Adverse effects of cannabis and cannabinoids. Br J Anaesth, 83(4), 637-649. Baddeley, A. (2003). Working memory: looking back and looking forward. Nat Rev Neurosci, 4(10),

829-839. Baddeley, A. D., & Hitch, G. (1974a). Working Memory. In G. H. Bower (Ed.), The psychology of

learning and motivation: Advances in research and theory.

(Vol. 8, pp. 47-89). New York: Academic Press. Baddeley, A. D., & Hitch, G. (1974b). Working Memory. The psychology of learning and motivation:

Advances in research and theory New York: Academic Press. Bajbouj, M., Gallinat, J., Niehaus, L., Lang, U. E., Roricht, S., & Meyer, B. U. (2004). Abnormalities

of inhibitory neuronal mechanisms in the motor cortex of patients with schizophrenia. Pharmacopsychiatry, 37(2), 74-80.

Barak, O., & Tsodyks, M. (2014). Working models of working memory. Curr Opin Neurobiol, 25, 20-24.

Barch, D. M., Carter, C. S., Braver, T. S., Sabb, F. W., MacDonald, A., 3rd, Noll, D. C., & Cohen, J. D. (2001). Selective deficits in prefrontal cortex function in medication-naive patients with schizophrenia. Arch Gen Psychiatry, 58(3), 280-288.

Page 120: Evaluating The Effects of Cannabis Dependence and ...

107  

Barnes, T. R. (1989). A rating scale for drug-induced akathisia. Br J Psychiatry, 154, 672-676. Barr, M. S., Farzan, F., Rajji, T. K., Voineskos, A. N., Blumberger, D. M., Arenovich, T., . . .

Daskalakis, Z. J. (2013). Can repetitive magnetic stimulation improve cognition in schizophrenia? Pilot data from a randomized controlled trial. Biol Psychiatry, 73(6), 510-517.

Barr, M. S., Farzan, F., Rusjan, P. M., Chen, R., Fitzgerald, P. B., & Daskalakis, Z. J. (2009). Potentiation of gamma oscillatory activity through repetitive transcranial magnetic stimulation of the dorsolateral prefrontal cortex. Neuropsychopharmacology, 34(11), 2359-2367.

Barr, M. S., Farzan, F., Tran, L. C., Chen, R., Fitzgerald, P. B., & Daskalakis, Z. J. (2010). Evidence for excessive frontal evoked gamma oscillatory activity in schizophrenia during working memory. Schizophr Res, 121(1-3), 146-152.

Barr, M. S., Fitzgerald, P. B., Farzan, F., George, T. P., & Daskalakis, Z. J. (2008). Transcranial magnetic stimulation to understand the pathophysiology and treatment of substance use disorders. Curr Drug Abuse Rev, 1(3), 328-339.

Bartos, M., Vida, I., & Jonas, P. (2007). Synaptic mechanisms of synchronized gamma oscillations in inhibitory interneuron networks. Nat Rev Neurosci, 8(1), 45-56.

Basar-Eroglu, C., Brand, A., Hildebrandt, H., Karolina Kedzior, K., Mathes, B., & Schmiedt, C. (2007). Working memory related gamma oscillations in schizophrenia patients. Int J Psychophysiol, 64(1), 39-45.

Becker, B., Wagner, D., Gouzoulis-Mayfrank, E., Spuentrup, E., & Daumann, J. (2010). Altered parahippocampal functioning in cannabis users is related to the frequency of use. Psychopharmacology (Berl), 209(4), 361-374.

Benes, F. M. (1998). Model generation and testing to probe neural circuitry in the cingulate cortex of postmortem schizophrenic brain. Schizophr Bull, 24(2), 219-230.

Bergamaschi, M. M., Queiroz, R. H., Chagas, M. H., de Oliveira, D. C., De Martinis, B. S., Kapczinski, F., . . . Crippa, J. A. (2011). Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naive social phobia patients. Neuropsychopharmacology, 36(6), 1219-1226.

Bilder, R. M., Goldman, R. S., Volavka, J., Czobor, P., Hoptman, M., Sheitman, B., . . . Lieberman, J. A. (2002). Neurocognitive effects of clozapine, olanzapine, risperidone, and haloperidol in patients with chronic schizophrenia or schizoaffective disorder. Am J Psychiatry, 159(6), 1018-1028.

Block, R. I., & Ghoneim, M. M. (1993). Effects of chronic marijuana use on human cognition. Psychopharmacology (Berl), 110(1-2), 219-228.

Bolla, K. I., Eldreth, D. A., London, E. D., Kiehl, K. A., Mouratidis, M., Contoreggi, C., . . . Ernst, M. (2003). Orbitofrontal cortex dysfunction in abstinent cocaine abusers performing a decision-making task. Neuroimage, 19(3), 1085-1094.

Bora, E., Yucel, M., & Pantelis, C. (2010). Cognitive impairment in schizophrenia and affective psychoses: implications for DSM-V criteria and beyond. Schizophr Bull, 36(1), 36-42.

Bossong, M. G., Jager, G., Bhattacharyya, S., & Allen, P. (2014). Acute and non-acute effects of cannabis on human memory function: a critical review of neuroimaging studies. Curr Pharm Des, 20(13), 2114-2125.

Bossong, M. G., van Berckel, B. N., Boellaard, R., Zuurman, L., Schuit, R. C., Windhorst, A. D., . . . Kahn, R. S. (2009). Delta 9-tetrahydrocannabinol induces dopamine release in the human striatum. Neuropsychopharmacology, 34(3), 759-766.

Boutros, N. N., Lisanby, S. H., McClain-Furmanski, D., Oliwa, G., Gooding, D., & Kosten, T. R. (2005). Cortical excitability in cocaine-dependent patients: a replication and extension of TMS findings. J Psychiatr Res, 39(3), 295-302.

Brown, J. T., Davies, C. H., & Randall, A. D. (2007). Synaptic activation of GABA(B) receptors regulates neuronal network activity and entrainment. Eur J Neurosci, 25(10), 2982-2990.

Page 121: Evaluating The Effects of Cannabis Dependence and ...

108  

Budney, A. J., Higgins, S. T., Radonovich, K. J., & Novy, P. L. (2000). Adding voucher-based incentives to coping skills and motivational enhancement improves outcomes during treatment for marijuana dependence. J Consult Clin Psychol, 68(6), 1051-1061.

Budney, A. J., Moore, B. A., Rocha, H. L., & Higgins, S. T. (2006). Clinical trial of abstinence-based vouchers and cognitive-behavioral therapy for cannabis dependence. J Consult Clin Psychol, 74(2), 307-316.

Budney, A. J., Moore, B. A., Vandrey, R. G., & Hughes, J. R. (2003). The time course and significance of cannabis withdrawal. J Abnorm Psychol, 112(3), 393-402.

Buzsaki, G. (2006). Rhythms of the Brain. New York, New York: Oxford University Press. Buzsaki, G., & Draguhn, A. (2004). Neuronal oscillations in cortical networks. Science, 304(5679),

1926-1929. Canolty, R. T., Edwards, E., Dalal, S. S., Soltani, M., Nagarajan, S. S., Kirsch, H. E., . . . Knight, R.

T. (2006). High gamma power is phase-locked to theta oscillations in human neocortex. Science, 313(5793), 1626-1628.

Cantello, R., Gianelli, M., Civardi, C., & Mutani, R. (1992). Magnetic brain stimulation: the silent period after the motor evoked potential. Neurology, 42(10), 1951-1959.

Carpenter, W. T., Jr., Buchanan, R. W., Kirkpatrick, B., & Breier, A. F. (1999). Diazepam treatment of early signs of exacerbation in schizophrenia. Am J Psychiatry, 156(2), 299-303.

Carter, C. S., Perlstein, W., Ganguli, R., Brar, J., Mintun, M., & Cohen, J. D. (1998). Functional hypofrontality and working memory dysfunction in schizophrenia. Am J Psychiatry, 155(9), 1285-1287.

Ceccarini, J., De Hert, M., Van Winkel, R., Peuskens, J., Bormans, G., Kranaster, L., . . . Van Laere, K. (2013). Increased ventral striatal CB1 receptor binding is related to negative symptoms in drug-free patients with schizophrenia. Neuroimage, 79, 304-312.

Chen, C. M., Stanford, A. D., Mao, X., Abi-Dargham, A., Shungu, D. C., Lisanby, S. H., . . . Kegeles, L. S. (2014). GABA level, gamma oscillation, and working memory performance in schizophrenia. Neuroimage Clin, 4, 531-539.

Chen, M. L. (2006). Ethnic or racial differences revisited: impact of dosage regimen and dosage form on pharmacokinetics and pharmacodynamics. Clin Pharmacokinet, 45(10), 957-964.

Chen, R. (2000). Studies of human motor physiology with transcranial magnetic stimulation. Muscle Nerve Suppl, 9, S26-32.

Chen, R., Lozano, A. M., & Ashby, P. (1999). Mechanism of the silent period following transcranial magnetic stimulation. Evidence from epidural recordings. Exp Brain Res, 128(4), 539-542.

Chevaleyre, V., Takahashi, K. A., & Castillo, P. E. (2006). Endocannabinoid-mediated synaptic plasticity in the CNS. Annu Rev Neurosci, 29, 37-76.

Cho, R. Y., Konecky, R. O., & Carter, C. S. (2006). Impairments in frontal cortical gamma synchrony and cognitive control in schizophrenia. Proc Natl Acad Sci U S A, 103(52), 19878-19883.

Clark, K. A., Randall, A. D., & Collingridge, G. L. (1994). A comparison of paired-pulsed facilitation of AMPA and NMDA receptor-mediated excitatory postsynaptic currents in the hippocampus. Exp Brain Res, 101(2), 272-278.

Clement, A. B., Hawkins, E. G., Lichtman, A. H., & Cravatt, B. F. (2003). Increased seizure susceptibility and proconvulsant activity of anandamide in mice lacking fatty acid amide hydrolase. J Neurosci, 23(9), 3916-3923.

Cohen, J. D., Perlstein, W. M., Braver, T. S., Nystrom, L. E., Noll, D. C., Jonides, J., & Smith, E. E. (1997). Temporal dynamics of brain activation during a working memory task. Nature, 386(6625), 604-608.

Cohen, M., Solowij, N., & Carr, V. (2008). Cannabis, cannabinoids and schizophrenia: integration of the evidence. Aust N Z J Psychiatry, 42(5), 357-368.

Compton, D. R., Johnson, M. R., Melvin, L. S., & Martin, B. R. (1992). Pharmacological profile of a series of bicyclic cannabinoid analogs: classification as cannabimimetic agents. J Pharmacol Exp Ther, 260(1), 201-209.

Page 122: Evaluating The Effects of Cannabis Dependence and ...

109  

Conte, A., Attilia, M. L., Gilio, F., Iacovelli, E., Frasca, V., Bettolo, C. M., . . . Inghilleri, M. (2008). Acute and chronic effects of ethanol on cortical excitability. Clin Neurophysiol, 119(3), 667-674.

Coulston, C., Perdices, M., Henderson, A., & Gin, S. M. (2011). Cannabinoids for the Treatment of Schizophrenia? A Balanced Neurochemical Framework for Both Adverse and Therapeutic Effects of Cannabis Use. Schizophrenia Research and Treatment, 2011.

Crean, R. D., Crane, N. A., & Mason, B. J. (2011). An evidence based review of acute and long-term effects of cannabis use on executive cognitive functions. J Addict Med, 5(1), 1-8.

Cui, S. S., Bowen, R. C., Gu, G. B., Hannesson, D. K., Yu, P. H., & Zhang, X. (2001). Prevention of cannabinoid withdrawal syndrome by lithium: involvement of oxytocinergic neuronal activation. J Neurosci, 21(24), 9867-9876.

Curran, H. V., Brignell, C., Fletcher, S., Middleton, P., & Henry, J. (2002). Cognitive and subjective dose-response effects of acute oral Delta 9-tetrahydrocannabinol (THC) in infrequent cannabis users. Psychopharmacology (Berl), 164(1), 61-70.

Curtis, C. E., & D'Esposito, M. (2003). Persistent activity in the prefrontal cortex during working memory. Trends Cogn Sci, 7(9), 415-423.

D'Esposito, M. (2007). From cognitive to neural models of working memory. Philos Trans R Soc Lond B Biol Sci, 362(1481), 761-772.

D'Souza, D. C., Abi-Saab, W. M., Madonick, S., Forselius-Bielen, K., Doersch, A., Braley, G., . . . Krystal, J. H. (2005). Delta-9-tetrahydrocannabinol effects in schizophrenia: implications for cognition, psychosis, and addiction. Biol Psychiatry, 57(6), 594-608.

Daskalakis, Z. J., Christensen, B. K., Chen, R., Fitzgerald, P. B., Zipursky, R. B., & Kapur, S. (2002). Evidence for impaired cortical inhibition in schizophrenia using transcranial magnetic stimulation. Arch Gen Psychiatry, 59(4), 347-354.

Daskalakis, Z. J., Christensen, B. K., Fitzgerald, P. B., & Chen, R. (2002). Transcranial magnetic stimulation: a new investigational and treatment tool in psychiatry. J Neuropsychiatry Clin Neurosci, 14(4), 406-415.

Daskalakis, Z. J., Christensen, B. K., Fitzgerald, P. B., Moller, B., Fountain, S. I., & Chen, R. (2008). Increased cortical inhibition in persons with schizophrenia treated with clozapine. J Psychopharmacol, 22(2), 203-209.

Daskalakis, Z. J., Farzan, F., Barr, M. S., Maller, J. J., Chen, R., & Fitzgerald, P. B. (2008). Long-interval cortical inhibition from the dorsolateral prefrontal cortex: a TMS-EEG study. Neuropsychopharmacology, 33(12), 2860-2869.

Daskalakis, Z. J., Molnar, G. F., Christensen, B. K., Sailer, A., Fitzgerald, P. B., & Chen, R. (2003). An automated method to determine the transcranial magnetic stimulation-induced contralateral silent period. Clin Neurophysiol, 114(5), 938-944.

Davey, N. J., Puri, B. K., Lewis, H. S., Lewis, S. W., & Ellaway, P. H. (1997). Effects of antipsychotic medication on electromyographic responses to transcranial magnetic stimulation of the motor cortex in schizophrenia. J Neurol Neurosurg Psychiatry, 63(4), 468-473.

Davidson, L., & McGlashan, T. H. (1997). The varied outcomes of schizophrenia. Can J Psychiatry, 42(1), 34-43.

Davies, C. H., Davies, S. N., & Collingridge, G. L. (1990). Paired-pulse depression of monosynaptic GABA-mediated inhibitory postsynaptic responses in rat hippocampus. J Physiol, 424, 513-531.

De Marchi, N., De Petrocellis, L., Orlando, P., Daniele, F., Fezza, F., & Di Marzo, V. (2003). Endocannabinoid signalling in the blood of patients with schizophrenia. Lipids Health Dis, 2, 5.

Dean, B., Sundram, S., Bradbury, R., Scarr, E., & Copolov, D. (2001). Studies on [3H]CP-55940 binding in the human central nervous system: regional specific changes in density of

Page 123: Evaluating The Effects of Cannabis Dependence and ...

110  

cannabinoid-1 receptors associated with schizophrenia and cannabis use. Neuroscience, 103(1), 9-15.

Dennis, M., Godley, S. H., Diamond, G., Tims, F. M., Babor, T., Donaldson, J., . . . Funk, R. (2004). The Cannabis Youth Treatment (CYT) Study: main findings from two randomized trials. J Subst Abuse Treat, 27(3), 197-213.

Di Lazzaro, V., Oliviero, A., Pilato, F., Saturno, E., Dileone, M., Marra, C., . . . Tonali, P. A. (2004). Motor cortex hyperexcitability to transcranial magnetic stimulation in Alzheimer's disease. J Neurol Neurosurg Psychiatry, 75(4), 555-559.

Di Lazzaro, V., Ziemann, U., & Lemon, R. N. (2008). State of the art: Physiology of transcranial motor cortex stimulation. Brain Stimul, 1(4), 345-362.

Dixon, L., Haas, G., Weiden, P. J., Sweeney, J., & Frances, A. J. (1991). Drug abuse in schizophrenic patients: clinical correlates and reasons for use. Am J Psychiatry, 148(2), 224-230.

Dolan, S. L., Sacco, K. A., Termine, A., Seyal, A. A., Dudas, M. M., Vessicchio, J. C., . . . George, T. P. (2004). Neuropsychological deficits are associated with smoking cessation treatment failure in patients with schizophrenia. Schizophr Res, 70(2-3), 263-275.

Driesen, N. R., McCarthy, G., Bhagwagar, Z., Bloch, M., Calhoun, V., D'Souza, D. C., . . . Krystal, J. H. (2013). Relationship of resting brain hyperconnectivity and schizophrenia-like symptoms produced by the NMDA receptor antagonist ketamine in humans. Mol Psychiatry, 18(11), 1199-1204.

Driesen, N. R., McCarthy, G., Bhagwagar, Z., Bloch, M. H., Calhoun, V. D., D'Souza, D. C., . . . Krystal, J. H. (2013). The impact of NMDA receptor blockade on human working memory-related prefrontal function and connectivity. Neuropsychopharmacology, 38(13), 2613-2622.

Eggan, S. M., & Lewis, D. A. (2007). Immunocytochemical distribution of the cannabinoid CB1 receptor in the primate neocortex: a regional and laminar analysis. Cereb Cortex, 17(1), 175-191.

Eichhammer, P., Wiegand, R., Kharraz, A., Langguth, B., Binder, H., & Hajak, G. (2004). Cortical excitability in neuroleptic-naive first-episode schizophrenic patients. Schizophr Res, 67(2-3), 253-259.

Ellis, G. M., Jr., Mann, M. A., Judson, B. A., Schramm, N. T., & Tashchian, A. (1985). Excretion patterns of cannabinoid metabolites after last use in a group of chronic users. Clin Pharmacol Ther, 38(5), 572-578.

Engel, A. K., Fries, P., & Singer, W. (2001). Dynamic predictions: oscillations and synchrony in top-down processing. Nat Rev Neurosci, 2(10), 704-716.

Englund, A., Morrison, P. D., Nottage, J., Hague, D., Kane, F., Bonaccorso, S., . . . Kapur, S. (2013). Cannabidiol inhibits THC-elicited paranoid symptoms and hippocampal-dependent memory impairment. J Psychopharmacol, 27(1), 19-27.

Ersche, K. D., Fletcher, P. C., Lewis, S. J., Clark, L., Stocks-Gee, G., London, M., . . . Sahakian, B. J. (2005). Abnormal frontal activations related to decision-making in current and former amphetamine and opiate dependent individuals. Psychopharmacology (Berl), 180(4), 612-623.

Falkai, P., Wobrock, T., Schneider-Axmann, T., & Gruber, O. (2008). [Schizophrenia as a brain disorder and its development]. Fortschr Neurol Psychiatr, 76 Suppl 1, S63-67.

Farzan, F., Barr, M. S., Levinson, A. J., Chen, R., Wong, W., Fitzgerald, P. B., & Daskalakis, Z. J. (2010a). Evidence for gamma inhibition deficits in the dorsolateral prefrontal cortex of patients with schizophrenia. Brain, 133(Pt 5), 1505-1514.

Farzan, F., Barr, M. S., Levinson, A. J., Chen, R., Wong, W., Fitzgerald, P. B., & Daskalakis, Z. J. (2010b). Reliability of long-interval cortical inhibition in healthy human subjects: a TMS-EEG study. J Neurophysiol, 104(3), 1339-1346.

First, M. B. (2002). The DSM series and experience with DSM-IV. Psychopathology, 35(2-3), 67-71.

Page 124: Evaluating The Effects of Cannabis Dependence and ...

111  

Fitzgerald, P. B., Brown, T. L., Daskalakis, Z. J., deCastella, A., & Kulkarni, J. (2002). A study of transcallosal inhibition in schizophrenia using transcranial magnetic stimulation. Schizophr Res, 56(3), 199-209.

Fitzgerald, P. B., Brown, T. L., Daskalakis, Z. J., & Kulkarni, J. (2002a). A transcranial magnetic stimulation study of inhibitory deficits in the motor cortex in patients with schizophrenia. Psychiatry Res, 114(1), 11-22.

Fitzgerald, P. B., Brown, T. L., Daskalakis, Z. J., & Kulkarni, J. (2002b). A transcranial magnetic stimulation study of the effects of olanzapine and risperidone on motor cortical excitability in patients with schizophrenia. Psychopharmacology (Berl), 162(1), 74-81.

Fitzgerald, P. B., Brown, T. L., Marston, N. A., Oxley, T. J., de Castella, A., Daskalakis, Z. J., & Kulkarni, J. (2003). A transcranial magnetic stimulation study of abnormal cortical inhibition in schizophrenia. Psychiatry Res, 118(3), 197-207.

Fitzgerald, P. B., Daskalakis, Z. J., Hoy, K., Farzan, F., Upton, D. J., Cooper, N. R., & Maller, J. J. (2008). Cortical inhibition in motor and non-motor regions: a combined TMS-EEG study. Clin EEG Neurosci, 39(3), 112-117.

Fitzgerald, P. B., Williams, S., & Daskalakis, Z. J. (2009). A transcranial magnetic stimulation study of the effects of cannabis use on motor cortical inhibition and excitability. Neuropsychopharmacology, 34(11), 2368-2375.

Forbes, N. F., Carrick, L. A., McIntosh, A. M., & Lawrie, S. M. (2009). Working memory in schizophrenia: a meta-analysis. Psychol Med, 39(6), 889-905.

Foti, D. J., Kotov, R., Guey, L. T., & Bromet, E. J. (2010). Cannabis use and the course of schizophrenia: 10-year follow-up after first hospitalization. Am J Psychiatry, 167(8), 987-993.

Fowler, I. L., Carr, V. J., Carter, N. T., & Lewin, T. J. (1998). Patterns of current and lifetime substance use in schizophrenia. Schizophr Bull, 24(3), 443-455.

Frantseva, M., Cui, J., Farzan, F., Chinta, L. V., Perez Velazquez, J. L., & Daskalakis, Z. J. (2014). Disrupted cortical conductivity in schizophrenia: TMS-EEG study. Cereb Cortex, 24(1), 211-221.

Frantseva, M. V., Fitzgerald, P. B., Chen, R., Moller, B., Daigle, M., & Daskalakis, Z. J. (2008). Evidence for impaired long-term potentiation in schizophrenia and its relationship to motor skill learning. Cereb Cortex, 18(5), 990-996.

Freeman, D., Dunn, G., Murray, R. M., Evans, N., Lister, R., Antley, A., . . . Morrison, P. D. (2015). How Cannabis Causes Paranoia: Using the Intravenous Administration of 9-Tetrahydrocannabinol (THC) to Identify Key Cognitive Mechanisms Leading to Paranoia. Schizophr Bull, 41(2), 391-399.

Freund, T. F., Katona, I., & Piomelli, D. (2003). Role of endogenous cannabinoids in synaptic signaling. Physiol Rev, 83(3), 1017-1066.

Fuster, J. M. (1973). Unit activity in prefrontal cortex during delayed-response performance: neuronal correlates of transient memory. Journal of Neurophysiology.

Galiegue, S., Mary, S., Marchand, J., Dussossoy, D., Carriere, D., Carayon, P., . . . Casellas, P. (1995). Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur J Biochem, 232(1), 54-61.

Gandal, M. J., Edgar, J. C., Ehrlichman, R. S., Mehta, M., Roberts, T. P., & Siegel, S. J. (2010). Validating gamma oscillations and delayed auditory responses as translational biomarkers of autism. Biol Psychiatry, 68(12), 1100-1106.

Gardner, E. L. (2005). Endocannabinoid signaling system and brain reward: emphasis on dopamine. Pharmacol Biochem Behav, 81(2), 263-284.

Gerdeman, G. L., Partridge, J. G., Lupica, C. R., & Lovinger, D. M. (2003). It could be habit forming: drugs of abuse and striatal synaptic plasticity. Trends Neurosci, 26(4), 184-192.

Page 125: Evaluating The Effects of Cannabis Dependence and ...

112  

Gevins, A., McEvoy, L. K., Smith, M. E., Chan, C. S., Sam-Vargas, L., Baum, C., & Ilan, A. B. (2012). Long-term and within-day variability of working memory performance and EEG in individuals. Clin Neurophysiol, 123(7), 1291-1299.

Giuffrida, A., Leweke, F. M., Gerth, C. W., Schreiber, D., Koethe, D., Faulhaber, J., . . . Piomelli, D. (2004). Cerebrospinal anandamide levels are elevated in acute schizophrenia and are inversely correlated with psychotic symptoms. Neuropsychopharmacology, 29(11), 2108-2114.

Glahn, D. C., Ragland, J. D., Abramoff, A., Barrett, J., Laird, A. R., Bearden, C. E., & Velligan, D. I. (2005). Beyond hypofrontality: a quantitative meta-analysis of functional neuroimaging studies of working memory in schizophrenia. Hum Brain Mapp, 25(1), 60-69.

Goldman-Rakic, P. S. (1995). Cellular basis of working memory. Neuron, 14(3), 477-485. Goldstein, J. M., Seidman, L. J., Santangelo, S., Knapp, P. H., & Tsuang, M. T. (1994). Are

schizophrenic men at higher risk for developmental deficits than schizophrenic women? Implications for adult neuropsychological functions. J Psychiatr Res, 28(6), 483-498.

Goodman, M. S., Rabin, R.A., Daskalakis, Z.J., George, T.P., Barr, M.S. (2015). Does 28-Days Abstinence Increase Working Memory Evoked Gamma Oscillations in Cannabis Dependent Schizophrenia Patients and Non-Psychiatric Controls?

. Paper presented at the International Congress on Schizophrenia Research, Colorodo Springs, CO. Goswami, S., Mattoo, S. K., Basu, D., & Singh, G. (2004). Substance-abusing schizophrenics: do

they self-medicate? Am J Addict, 13(2), 139-150. Gottesman, I. (1991). Psychiatric Genesis: The

Origins of Madness. . New York

Green, M. F. (2006). Cognitive impairment and functional outcome in schizophrenia and bipolar disorder. J Clin Psychiatry, 67 Suppl 9, 3-8; discussion 36-42.

Grotenhermen, F. (2003). Pharmacokinetics and pharmacodynamics of cannabinoids. Clin Pharmacokinet, 42(4), 327-360.

Gruber, S. A., Sagar, K. A., Dahlgren, M. K., Racine, M., & Lukas, S. E. (2012). Age of onset of marijuana use and executive function. Psychol Addict Behav, 26(3), 496-506.

Guy, W., & Cleary, P. A. (1976). Pretreatment status and its relationship to the length of drying-out period. Psychopharmacol Bull, 12(2), 20-22.

Haenschel, C., Bittner, R. A., Waltz, J., Haertling, F., Wibral, M., Singer, W., . . . Rodriguez, E. (2009). Cortical oscillatory activity is critical for working memory as revealed by deficits in early-onset schizophrenia. J Neurosci, 29(30), 9481-9489.

Hall, W., Solowij, N., & Lemon, J. T. (1994). The health and psychological consequences of cannabis use National Drug Strategy Monograph Series (Vol. 25). Canberra: Australian Government Publishing Service.

Harrison, P. J., & Weinberger, D. R. (2005). Schizophrenia genes, gene expression, and neuropathology: on the matter of their convergence. Mol Psychiatry, 10(1), 40-68; image 45.

Hartung, B., Kauferstein, S., Ritz-Timme, S., & Daldrup, T. (2014). Sudden unexpected death under acute influence of cannabis. Forensic Sci Int, 237, e11-13.

Harvey, D. J., & Brown, N. K. (1991). Comparative in vitro metabolism of the cannabinoids. Pharmacol Biochem Behav, 40(3), 533-540.

Harvey, D. J., Paton, W.D.M. (1976). Examination of the metabolites of Δ1-tetrahydrocannabinol in mouse, liver, heart and lung by

combined gas chromatography and mass spectrometry. New York: Springer-Verlag. Hasan, A., Wobrock, T., Grefkes, C., Labusga, M., Levold, K., Schneider-Axmann, T., . . . Bechdolf,

A. (2012). Deficient inhibitory cortical networks in antipsychotic-naive subjects at risk of

Page 126: Evaluating The Effects of Cannabis Dependence and ...

113  

developing first-episode psychosis and first-episode schizophrenia patients: a cross-sectional study. Biol Psychiatry, 72(9), 744-751.

Hashimoto, T., Bazmi, H. H., Mirnics, K., Wu, Q., Sampson, A. R., & Lewis, D. A. (2008). Conserved regional patterns of GABA-related transcript expression in the neocortex of subjects with schizophrenia. Am J Psychiatry, 165(4), 479-489.

Hawksworth, G., & McArdle, K. (2004). Metabolism and pharmacokinetics of cannabinoids. In G. Guy, B. Whittle & P. Robson (Eds.), The medicinal uses of cannabis and cannabinoids (pp. 205–228). London: Pharmaceutical Press.

Hayakawa, K., Mishima, K., Nozako, M., Ogata, A., Hazekawa, M., Liu, A. X., . . . Fujiwara, M. (2007). Repeated treatment with cannabidiol but not Delta9-tetrahydrocannabinol has a neuroprotective effect without the development of tolerance. Neuropharmacology, 52(4), 1079-1087.

Heatherton, T. F., Kozlowski, L. T., Frecker, R. C., & Fagerstrom, K. O. (1991). The Fagerstrom Test for Nicotine Dependence: a revision of the Fagerstrom Tolerance Questionnaire. Br J Addict, 86(9), 1119-1127.

Herkenham, M. (1991). Characterization and localization of cannabinoid receptors in brain: an in vitro technique using slide-mounted tissue sections. NIDA Res Monogr, 112, 129-145.

Hermann, C. S. (1993). Gamma response and ERPs in a visual classification task. Clin Neurophysiol, 110, 636-642.

Hill, M. N., Froc, D. J., Fox, C. J., Gorzalka, B. B., & Christie, B. R. (2004). Prolonged cannabinoid treatment results in spatial working memory deficits and impaired long-term potentiation in the CA1 region of the hippocampus in vivo. Eur J Neurosci, 20(3), 859-863.

Hoffman, A. F., Oz, M., Caulder, T., & Lupica, C. R. (2003). Functional tolerance and blockade of long-term depression at synapses in the nucleus accumbens after chronic cannabinoid exposure. J Neurosci, 23(12), 4815-4820.

Honey, G. D., Fu, C. H., Kim, J., Brammer, M. J., Croudace, T. J., Suckling, J., . . . Bullmore, E. T. (2002). Effects of verbal working memory load on corticocortical connectivity modeled by path analysis of functional magnetic resonance imaging data. Neuroimage, 17(2), 573-582.

Hong, L. E., Summerfelt, A., McMahon, R., Adami, H., Francis, G., Elliott, A., . . . Thaker, G. K. (2004). Evoked gamma band synchronization and the liability for schizophrenia. Schizophr Res, 70(2-3), 293-302.

Howard, M. W., Rizzuto, D. S., Caplan, J. B., Madsen, J. R., Lisman, J., Aschenbrenner-Scheibe, R., . . . Kahana, M. J. (2003). Gamma oscillations correlate with working memory load in humans. Cereb Cortex, 13(12), 1369-1374.

Howlett, A. C., Breivogel, C. S., Childers, S. R., Deadwyler, S. A., Hampson, R. E., & Porrino, L. J. (2004). Cannabinoid physiology and pharmacology: 30 years of progress. Neuropharmacology, 47 Suppl 1, 345-358.

Huestis, M. A., Henningfield, J. E., & Cone, E. J. (1992). Blood cannabinoids. I. Absorption of THC and formation of 11-OH-THC and THCCOOH during and after smoking marijuana. J Anal Toxicol, 16(5), 276-282.

Huestis, M. A., Mitchell, J. M., & Cone, E. J. (1995). Detection times of marijuana metabolites in urine by immunoassay and GC-MS. J Anal Toxicol, 19(6), 443-449.

Humphries, C., Mortimer, A., Hirsch, S., & de Belleroche, J. (1996). NMDA receptor mRNA correlation with antemortem cognitive impairment in schizophrenia. Neuroreport, 7(12), 2051-2055.

Hunt, C. A., & Jones, R. T. (1980). Tolerance and disposition of tetrahydrocannabinol in man. J Pharmacol Exp Ther, 215(1), 35-44.

Ilan, A. B., Gevins, A., Coleman, M., ElSohly, M. A., & de Wit, H. (2005). Neurophysiological and subjective profile of marijuana with varying concentrations of cannabinoids. Behav Pharmacol, 16(5-6), 487-496.

Iversen, L. (2003). Cannabis and the brain. Brain, 126(Pt 6), 1252-1270.

Page 127: Evaluating The Effects of Cannabis Dependence and ...

114  

Iversen, L. (2008). The Science of Marijuana (Vol. 2nd edition). Oxford: Oxford University Press. Jablensky, A. (2000). Prevalence and incidence of schizophrenia spectrum disorders: implications for

prevention. Aust N Z J Psychiatry, 34 Suppl, S26-34; discussion S35-28. Jacobsen, C. F. (1936). Studies of cerebral function in primates. Comp Psychol Monogr, 13, 1-68. Jaffe, J. H. (1985). Drug addiction and drug abuse. In A. G. Gilman, L. S. Goodman & F. Murad

(Eds.), The Pharmacological Basis of Therapeutics. USA: Macmillan. Jager, G., Kahn, R. S., Van Den Brink, W., Van Ree, J. M., & Ramsey, N. F. (2006). Long-term

effects of frequent cannabis use on working memory and attention: an fMRI study. Psychopharmacology (Berl), 185(3), 358-368.

James, A., James, C., & Thwaites, T. (2013). The brain effects of cannabis in healthy adolescents and in adolescents with schizophrenia: a systematic review. Psychiatry Res, 214(3), 181-189.

Jansma, J. M., Ramsey, N. F., van der Wee, N. J., & Kahn, R. S. (2004). Working memory capacity in schizophrenia: a parametric fMRI study. Schizophr Res, 68(2-3), 159-171.

Javitt, D. C. (2007). Glutamate and schizophrenia: phencyclidine, N-methyl-D-aspartate receptors, and dopamine-glutamate interactions. Int Rev Neurobiol, 78, 69-108.

Jensen, K., Chiu, C. S., Sokolova, I., Lester, H. A., & Mody, I. (2003). GABA transporter-1 (GAT1)-deficient mice: differential tonic activation of GABAA versus GABAB receptors in the hippocampus. J Neurophysiol, 90(4), 2690-2701.

Jockers-Scherubl, M. C., Wolf, T., Radzei, N., Schlattmann, P., Rentzsch, J., Gomez-Carrillo de Castro, A., & Kuhl, K. P. (2007). Cannabis induces different cognitive changes in schizophrenic patients and in healthy controls. Prog Neuropsychopharmacol Biol Psychiatry, 31(5), 1054-1063.

Johansson, E., Halldin, M. M., Agurell, S., Hollister, L. E., & Gillespie, H. K. (1989). Terminal elimination plasma half-life of delta 1-tetrahydrocannabinol (delta 1-THC) in heavy users of marijuana. Eur J Clin Pharmacol, 37(3), 273-277.

Johnson, J. L. (1972). Glutamic acid as a synaptic transmitter in the nervous system. A review. Brain Res, 37(1), 1-19.

Jones, M. W., Kilpatrick, I. C., & Phillipson, O. T. (1986). The agranular insular cortex: a site of unusually high dopamine utilisation. Neurosci Lett, 72(3), 330-334.

Jonides, J., Schumacher, E. H., Smith, E. E., Koeppe, R. A., Awh, E., Reuter-Lorenz, P. A., . . . Willis, C. R. (1998). The role of parietal cortex in verbal working memory. J Neurosci, 18(13), 5026-5034.

Joy, J. E., Watson, Jr., S.J. and Benson, Jr,J.S. (1999). Marijuana and Medicine: Assessing The Science Base. Washington D.C: National Academy of Sciences Press.

Joyal, C. C., Halle, P., Lapierre, D., & Hodgins, S. (2003). Drug abuse and/or dependence and better neuropsychological performance in patients with schizophrenia. Schizophr Res, 63(3), 297-299.

Julien, R. M. (2001). A Primer of Drug Action (Vol. 9). New York: Worth Publishers. Kadden, R. M., Litt, M. D., Kabela-Cormier, E., & Petry, N. M. (2007). Abstinence rates following

behavioral treatments for marijuana dependence. Addict Behav, 32(6), 1220-1236. Kalman, D., Morissette, S. B., & George, T. P. (2005). Co-morbidity of smoking in patients with

psychiatric and substance use disorders. Am J Addict, 14(2), 106-123. Kaster, T. S., de Jesus, D., Radhu, N., Farzan, F., Blumberger, D. M., Rajji, T. K., . . . Daskalakis, Z.

J. (2015). Clozapine potentiation of GABA mediated cortical inhibition in treatment resistant schizophrenia. Schizophr Res.

Kay, S. R., Fiszbein, A., & Opler, L. A. (1987). The positive and negative syndrome scale (PANSS) for schizophrenia. Schizophr Bull, 13(2), 261-276.

Kay, S. R., Opler, L. A., & Lindenmayer, J. P. (1988). Reliability and validity of the positive and negative syndrome scale for schizophrenics. Psychiatry Res, 23(1), 99-110.

Kobayashi, M., & Pascual-Leone, A. (2003). Transcranial magnetic stimulation in neurology. Lancet Neurol, 2(3), 145-156.

Page 128: Evaluating The Effects of Cannabis Dependence and ...

115  

Koethe, D., Giuffrida, A., Schreiber, D., Hellmich, M., Schultze-Lutter, F., Ruhrmann, S., . . . Leweke, F. M. (2009). Anandamide elevation in cerebrospinal fluid in initial prodromal states of psychosis. Br J Psychiatry, 194(4), 371-372.

Kolliakou, A., Castle, D., Sallis, H., Joseph, C., O'Connor, J., Wiffen, B., . . . Ismail, K. (2015). Reasons for cannabis use in first-episode psychosis: does strength of endorsement change over 12 months? Eur Psychiatry, 30(1), 152-159.

Koskinen, J., Lohonen, J., Koponen, H., Isohanni, M., & Miettunen, J. (2010). Rate of cannabis use disorders in clinical samples of patients with schizophrenia: a meta-analysis. Schizophr Bull, 36(6), 1115-1130.

Krabbendam, L., & van Os, J. (2005). Schizophrenia and urbanicity: a major environmental influence--conditional on genetic risk. Schizophr Bull, 31(4), 795-799.

Kremen, W. S., Seidman, L. J., Faraone, S. V., & Tsuang, M. T. (2001). Intelligence quotient and neuropsychological profiles in patients with schizophrenia and in normal volunteers. Biol Psychiatry, 50(6), 453-462.

Kujirai, T., Caramia, M. D., Rothwell, J. C., Day, B. L., Thompson, P. D., Ferbert, A., . . . Marsden, C. D. (1993). Corticocortical inhibition in human motor cortex. J Physiol, 471, 501-519.

Lafolie, P., Beck, O., Blennow, G., Boreus, L., Borg, S., Elwin, C. E., . . . Hjemdahl, P. (1991). Importance of creatinine analyses of urine when screening for abused drugs. Clin Chem, 37(11), 1927-1931.

Lahti, A. C., Holcomb, H. H., Gao, X. M., & Tamminga, C. A. (1999). NMDA-sensitive glutamate antagonism: a human model for psychosis. Neuropsychopharmacology, 21, S158-S169.

Lahti, A. C., Holcomb, H. H., Weiler, M. A., Corey, P. K., Zhao, M., Medoff, D., & Tamminga, C. A. (1997). Effects of ketamine on behavior and rCBF in schizophrenia and normal individuals. Biol Psychiatry, 41(165 - 169).

Lahti, A. C., Weiler, M. A., Tamara Michaelidis, B. A., Parwani, A., & Tamminga, C. A. (2001). Effects of ketamine in normal and schizophrenic volunteers. Neuropsychopharmacology, 25(4), 455-467.

Lang, N., Hasan, A., Sueske, E., Paulus, W., & Nitsche, M. A. (2008). Cortical hypoexcitability in chronic smokers? A transcranial magnetic stimulation study. Neuropsychopharmacology, 33(10), 2517-2523.

Lasser, K., Boyd, J. W., Woolhandler, S., Himmelstein, D. U., McCormick, D., & Bor, D. H. (2000). Smoking and mental illness: A population-based prevalence study. JAMA, 284(20), 2606-2610.

Leeson, V. C., Harrison, I., Ron, M. A., Barnes, T. R., & Joyce, E. M. (2011). The Effect of Cannabis Use and Cognitive Reserve on Age at Onset and Psychosis Outcomes in First-Episode Schizophrenia. Schizophr Bull.

Leung, L. S., & Shen, B. (2007). GABAB receptor blockade enhances theta and gamma rhythms in the hippocampus of behaving rats. Hippocampus, 17(4), 281-291.

Lewis, D. A., Cho, R. Y., Carter, C. S., Eklund, K., Forster, S., Kelly, M. A., & Montrose, D. (2008). Subunit-selective modulation of GABA type A receptor neurotransmission and cognition in schizophrenia. Am J Psychiatry, 165(12), 1585-1593.

Lewis, D. A., Curley, A. A., Glausier, J. R., & Volk, D. W. (2012). Cortical parvalbumin interneurons and cognitive dysfunction in schizophrenia. Trends Neurosci, 35(1), 57-67.

Lewis, D. A., Hashimoto, T., & Volk, D. W. (2005). Cortical inhibitory neurons and schizophrenia. Nat Rev Neurosci, 6(4), 312-324.

Lewis, D. A., & Lieberman, J. A. (2000). Catching up on schizophrenia: natural history and neurobiology. Neuron, 28(2), 325-334.

Lewis, D. A., & Moghaddam, B. (2006). Cognitive dysfunction in schizophrenia: convergence of gamma-aminobutyric acid and glutamate alterations. Arch Neurol, 63(10), 1372-1376.

Page 129: Evaluating The Effects of Cannabis Dependence and ...

116  

Lewis, D. A., Pierri, J. N., Volk, D. W., Melchitzky, D. S., & Woo, T. U. (1999). Altered GABA neurotransmission and prefrontal cortical dysfunction in schizophrenia. Biol Psychiatry, 46(5), 616-626.

Lewis, D. A., Volk, D. W., & Hashimoto, T. (2004). Selective alterations in prefrontal cortical GABA neurotransmission in schizophrenia: a novel target for the treatment of working memory dysfunction. Psychopharmacology (Berl), 174(1), 143-150.

Linszen, D. H., Dingemans, P. M., & Lenior, M. E. (1994). Cannabis abuse and the course of recent-onset schizophrenic disorders. Arch Gen Psychiatry, 51(4), 273-279.

Liu, S. K., Fitzgerald, P. B., Daigle, M., Chen, R., & Daskalakis, Z. J. (2009). The relationship between cortical inhibition, antipsychotic treatment, and the symptoms of schizophrenia. Biol Psychiatry, 65(6), 503-509.

Lopez-Larson, M. P., Bogorodzki, P., Rogowska, J., McGlade, E., King, J. B., Terry, J., & Yurgelun-Todd, D. (2011). Altered prefrontal and insular cortical thickness in adolescent marijuana users. Behav Brain Res, 220(1), 164-172.

Maeda, F., Keenan, J. P., & Pascual-Leone, A. (2000). Interhemispheric asymmetry of motor cortical excitability in major depression as measured by transcranial magnetic stimulation. Br J Psychiatry, 177, 169-173.

Mainy, N., Kahane, P., Minotti, L., Hoffmann, D., Bertrand, O., & Lachaux, J. P. (2007). Neural correlates of consolidation in working memory. Hum Brain Mapp, 28(3), 183-193.

Manrique-Garcia, E., Zammit, S., Dalman, C., Hemmingsson, T., Andreasson, S., & Allebeck, P. (2014). Prognosis of schizophrenia in persons with and without a history of cannabis use. Psychol Med, 44(12), 2513-2521.

Martinez-Gras, I., Hoenicka, J., Ponce, G., Rodriguez-Jimenez, R., Jimenez-Arriero, M. A., Perez-Hernandez, E., . . . Rubio, G. (2006). (AAT)n repeat in the cannabinoid receptor gene, CNR1: association with schizophrenia in a Spanish population. Eur Arch Psychiatry Clin Neurosci, 256(7), 437-441.

Mata, I., Rodriguez-Sanchez, J. M., Pelayo-Teran, J. M., Perez-Iglesias, R., Gonzalez-Blanch, C., Ramirez-Bonilla, M., . . . Crespo-Facorro, B. (2008). Cannabis abuse is associated with decision-making impairment among first-episode patients with schizophrenia-spectrum psychosis. Psychol Med, 38(9), 1257-1266.

McDonald, J., Schleifer, L., Richards, J. B., & de Wit, H. (2003). Effects of THC on behavioral measures of impulsivity in humans. Neuropsychopharmacology, 28(7), 1356-1365.

McDonnell, M. N., Orekhov, Y., & Ziemann, U. (2006). The role of GABA(B) receptors in intracortical inhibition in the human motor cortex. Exp Brain Res, 173(1), 86-93.

McGee, R., Williams, S., Poulton, R., & Moffitt, T. (2000). A longitudinal study of cannabis use and mental health from adolescence to early adulthood. Addiction, 95(4), 491-503.

McLellan, A. T., Alterman, A. I., Cacciola, J., Metzger, D., & O'Brien, C. P. (1992). A new measure of substance abuse treatment. Initial studies of the treatment services review. J Nerv Ment Dis, 180(2), 101-110.

Mednick, S. A., Machon, R. A., Huttunen, M. O., & Bonett, D. (1988). Adult schizophrenia following prenatal exposure to an influenza epidemic. Arch Gen Psychiatry, 45(2), 189-192.

Mehmedic, Z., Chandra, S., Slade, D., Denham, H., Foster, S., Patel, A. S., . . . ElSohly, M. A. (2010). Potency trends of Delta9-THC and other cannabinoids in confiscated cannabis preparations from 1993 to 2008. J Forensic Sci, 55(5), 1209-1217.

Mehta, U. M., Thirthalli, J., Basavaraju, R., & Gangadhar, B. N. (2014). Association of intracortical inhibition with social cognition deficits in schizophrenia: Findings from a transcranial magnetic stimulation study. Schizophr Res, 158(1-3), 146-150.

Meltzer, H. Y., & McGurk, S. R. (1999). The effects of clozapine, risperidone, and olanzapine on cognitive function in schizophrenia. Schizophr Bull, 25(2), 233-255.

Page 130: Evaluating The Effects of Cannabis Dependence and ...

117  

Meltzer, J. A., Zaveri, H. P., Goncharova, II, Distasio, M. M., Papademetris, X., Spencer, S. S., . . . Constable, R. T. (2008). Effects of working memory load on oscillatory power in human intracranial EEG. Cereb Cortex, 18(8), 1843-1855.

Mittleman, M. A., Lewis, R. A., Maclure, M., Sherwood, J. B., & Muller, J. E. (2001). Triggering myocardial infarction by marijuana. Circulation, 103(23), 2805-2809.

Moore, T. H., Zammit, S., Lingford-Hughes, A., Barnes, T. R., Jones, P. B., Burke, M., & Lewis, G. (2007). Cannabis use and risk of psychotic or affective mental health outcomes: a systematic review. Lancet, 370(9584), 319-328.

Morgan, C. J., Schafer, G., Freeman, T. P., & Curran, H. V. (2010). Impact of cannabidiol on the acute memory and psychotomimetic effects of smoked cannabis: naturalistic study: naturalistic study [corrected]. Br J Psychiatry, 197(4), 285-290.

Murray, R. M., Morrison, P. D., Henquet, C., & Di Forti, M. (2007). Cannabis, the mind and society: the hash realities. Nat Rev Neurosci, 8(11), 885-895.

Pascual-Leone, A., Manoach, D. S., Birnbaum, R., & Goff, D. C. (2002). Motor cortical excitability in schizophrenia. Biol Psychiatry, 52(1), 24-31.

Patton, G. C., Coffey, C., Carlin, J. B., Sawyer, S. M., & Wakefield, M. (2006). Teen smokers reach their mid twenties. J Adolesc Health, 39(2), 214-220.

Perez-Reyes, M., White, W. R., McDonald, S. A., Hicks, R. E., Jeffcoat, A. R., & Cook, C. E. (1991). The pharmacologic effects of daily marijuana smoking in humans. Pharmacol Biochem Behav, 40(3), 691-694.

Pertwee, R. G. (1997). Pharmacology of cannabinoid CB1 and CB2 receptors. Pharmacol Ther, 74(2), 129-180.

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

Pesaran, B., Pezaris, J. S., Sahani, M., Mitra, P. P., & Andersen, R. A. (2002). Temporal structure in neuronal activity during working memory in macaque parietal cortex. Nat Neurosci, 5(8), 805-811.

Piomelli, D. (2003). The molecular logic of endocannabinoid signalling. Nat Rev Neurosci, 4(11), 873-884.

Piomelli, D., Beltramo, M., Giuffrida, A., & Stella, N. (1998). Endogenous cannabinoid signaling. Neurobiol Dis, 5(6 Pt B), 462-473.

Pistis, M., Ferraro, L., Pira, L., Flore, G., Tanganelli, S., Gessa, G. L., & Devoto, P. (2002). Delta(9)-tetrahydrocannabinol decreases extracellular GABA and increases extracellular glutamate and dopamine levels in the rat prefrontal cortex: an in vivo microdialysis study. Brain Res, 948(1-2), 155-158.

Pistis, M., Porcu, G., Melis, M., Diana, M., & Gessa, G. L. (2001). Effects of cannabinoids on prefrontal neuronal responses to ventral tegmental area stimulation. Eur J Neurosci, 14(1), 96-102.

Pope, H. G., Jr., Gruber, A. J., Hudson, J. I., Huestis, M. A., & Yurgelun-Todd, D. (2001). Neuropsychological performance in long-term cannabis users. Arch Gen Psychiatry, 58(10), 909-915.

Postle, B. R. (2006). Working memory as an emergent property of the mind and brain. Neuroscience, 139(1), 23-38.

Potter, A., Downey, L., & Stough, C. (2013). Cognitive function in ecstasy naive abstinent drug dependants and MDMA users. Curr Drug Abuse Rev, 6(1), 71-76.

Potter, D. J., Clark, P., & Brown, M. B. (2008). Potency of delta 9-THC and other cannabinoids in cannabis in England in 2005: implications for psychoactivity and pharmacology. J Forensic Sci, 53(1), 90-94.

Page 131: Evaluating The Effects of Cannabis Dependence and ...

118  

Premoli, I., Castellanos, N., Rivolta, D., Belardinelli, P., Bajo, R., Zipser, C., . . . Ziemann, U. (2014). TMS-EEG signatures of GABAergic neurotransmission in the human cortex. J Neurosci, 34(16), 5603-5612.

Prilipko, O., Huynh, N., Schwartz, S., Tantrakul, V., Kim, J. H., Peralta, A. R., . . . Guilleminault, C. (2011). Task positive and default mode networks during a parametric working memory task in obstructive sleep apnea patients and healthy controls. Sleep, 34(3), 293-301A.

Puri, B. K., Davey, N. J., Ellaway, P. H., & Lewis, S. W. (1996). An investigation of motor function in schizophrenia using transcranial magnetic stimulation of the motor cortex. Br J Psychiatry, 169(6), 690-695.

Rabin, R. A., Goodman, M.S., George, T.P., Barr, M.S. (2014). Neurobiology of Comorbid Substance Use Disorders in Mental Illness: A Closer Look at the Underlying Commonalities between Cannabis and Schizophrenia. Current Addiction Reports.

Rabin, R. A., Zakzanis, K. K., & George, T. P. (2011). The effects of cannabis use on neurocognition in schizophrenia: a meta-analysis. Schizophr Res, 128(1-3), 111-116.

Radhakrishnan, R., Wilkinson, S. T., & D'Souza, D. C. (2014). Gone to Pot - A Review of the Association between Cannabis and Psychosis. Front Psychiatry, 5, 54.

Radhu, N., de Jesus, D. R., Ravindran, L. N., Zanjani, A., Fitzgerald, P. B., & Daskalakis, Z. J. (2013). A meta-analysis of cortical inhibition and excitability using transcranial magnetic stimulation in psychiatric disorders. Clin Neurophysiol, 124(7), 1309-1320.

Ramaekers, J. G., Kauert, G., van Ruitenbeek, P., Theunissen, E. L., Schneider, E., & Moeller, M. R. (2006). High-potency marijuana impairs executive function and inhibitory motor control. Neuropsychopharmacology, 31(10), 2296-2303.

Rao, S. G., Williams, G. V., & Goldman-Rakic, P. S. (2000). Destruction and creation of spatial tuning by disinhibition: GABA(A) blockade of prefrontal cortical neurons engaged by working memory. J Neurosci, 20(1), 485-494.

Ringen, P. A., Vaskinn, A., Sundet, K., Engh, J. A., Jonsdottir, H., Simonsen, C., . . . Andreassen, O. A. (2010). Opposite relationships between cannabis use and neurocognitive functioning in bipolar disorder and schizophrenia. Psychol Med, 40(8), 1337-1347.

Robbins, T. W., & Arnsten, A. F. (2009). The neuropsychopharmacology of fronto-executive function: monoaminergic modulation. Annu Rev Neurosci, 32, 267-287.

Roberts, E., & Frankel, S. (1950). gamma-Aminobutyric acid in brain: its formation from glutamic acid. J Biol Chem, 187(1), 55-63.

Rodriguez-Sanchez, J. M., Ayesa-Arriola, R., Mata, I., Moreno-Calle, T., Perez-Iglesias, R., Gonzalez-Blanch, C., . . . Crespo-Facorro, B. (2010). Cannabis use and cognitive functioning in first-episode schizophrenia patients. Schizophr Res, 124(1-3), 142-151.

Rose, E. J., & Ebmeier, K. P. (2006). Pattern of impaired working memory during major depression. J Affect Disord, 90(2-3), 149-161.

Rossler, W., Salize, H. J., van Os, J., & Riecher-Rossler, A. (2005). Size of burden of schizophrenia and psychotic disorders. Eur Neuropsychopharmacol, 15(4), 399-409.

Rothwell, J. C., & Rosenkranz, K. (2005). Role of afferent input in motor organization in health and disease. IEEE Eng Med Biol Mag, 24(1), 40-44.

Sacco, K. A., Termine, A., Seyal, A., Dudas, M. M., Vessicchio, J. C., Krishnan-Sarin, S., . . . George, T. P. (2005). Effects of cigarette smoking on spatial working memory and attentional deficits in schizophrenia: involvement of nicotinic receptor mechanisms. Arch Gen Psychiatry, 62(6), 649-659. .

Sanchez-Carrion, R., Gomez, P. V., Junque, C., Fernandez-Espejo, D., Falcon, C., Bargallo, N., . . . Bernabeu, M. (2008). Frontal hypoactivation on functional magnetic resonance imaging in working memory after severe diffuse traumatic brain injury. J Neurotrauma, 25(5), 479-494.

Sanger, T. D., Garg, R. R., & Chen, R. (2001). Interactions between two different inhibitory systems in the human motor cortex. J Physiol, 530(Pt 2), 307-317.

Page 132: Evaluating The Effects of Cannabis Dependence and ...

119  

Sansone, R. A., & Sansone, L. A. (2014). Marijuana and body weight. Innov Clin Neurosci, 11(7-8), 50-54.

Saunders, J. B., Aasland, O. G., Babor, T. F., de la Fuente, J. R., & Grant, M. (1993). Development of the Alcohol Use Disorders Identification Test (AUDIT): WHO Collaborative Project on Early Detection of Persons with Harmful Alcohol Consumption--II. Addiction, 88(6), 791-804.

Sawaguchi, T., Matsumura, M., & Kubota, K. (1989). Delayed response deficits produced by local injection of bicuculline into the dorsolateral prefrontal cortex in Japanese macaque monkeys. Exp Brain Res, 75(3), 457-469.

Schnell, T., Koethe, D., Daumann, J., & Gouzoulis-Mayfrank, E. (2009). The role of cannabis in cognitive functioning of patients with schizophrenia. Psychopharmacology (Berl), 205(1), 45-52.

Schoeler, T., & Bhattacharyya, S. (2013). The effect of cannabis use on memory function: an update. Subst Abuse Rehabil, 4, 11-27.

Schweinsburg, A. D., Schweinsburg, B. C., Cheung, E. H., Brown, G. G., Brown, S. A., & Tapert, S. F. (2005). fMRI response to spatial working memory in adolescents with comorbid marijuana and alcohol use disorders. Drug Alcohol Depend, 79(2), 201-210.

Schweinsburg, A. D., Schweinsburg, B. C., Medina, K. L., McQueeny, T., Brown, S. A., & Tapert, S. F. (2010). The influence of recency of use on fMRI response during spatial working memory in adolescent marijuana users. J Psychoactive Drugs, 42(3), 401-412.

Sevy, S., Burdick, K. E., Visweswaraiah, H., Abdelmessih, S., Lukin, M., Yechiam, E., & Bechara, A. (2007). Iowa gambling task in schizophrenia: a review and new data in patients with schizophrenia and co-occurring cannabis use disorders. Schizophr Res, 92(1-3), 74-84.

Sharma, T., Hughes, C., Soni, W., & Kumari, V. (2003). Cognitive effects of olanzapine and clozapine treatment in chronic schizophrenia. Psychopharmacology (Berl), 169(3-4), 398-403.

Shusterman, V., & Troy, W. C. (2008). From baseline to epileptiform activity: a path to synchronized rhythmicity in large-scale neural networks. Phys Rev E Stat Nonlin Soft Matter Phys, 77(6 Pt 1), 061911.

Sigmon, S. C., Steingard, S., Badger, G. J., Anthony, S. L., & Higgins, S. T. (2000). Contingent reinforcement of marijuana abstinence among individuals with serious mental illness: a feasibility study. Exp Clin Psychopharmacol, 8(4), 509-517.

Silva de Melo, L. C., Cruz, A. P., Rios Valentim, S. J., Jr., Marinho, A. R., Mendonca, J. B., & Nakamura-Palacios, E. M. (2005). Delta(9)-THC administered into the medial prefrontal cortex disrupts the spatial working memory. Psychopharmacology (Berl), 183(1), 54-64.

Sim-Selley, L. J. (2003). Regulation of cannabinoid CB1 receptors in the central nervous system by chronic cannabinoids. Crit Rev Neurobiol, 15(2), 91-119.

Simpson, G. M., & Angus, J. W. (1970). A rating scale for extrapyramidal side effects. Acta Psychiatr Scand Suppl, 212, 11-19.

Sinha, R., Easton, C., Renee-Aubin, L., & Carroll, K. M. (2003). Engaging young probation-referred marijuana-abusing individuals in treatment: a pilot trial. Am J Addict, 12(4), 314-323.

Smith, E. E., Jonides, J., Marshuetz, C., & Koeppe, R. A. (1998). Components of verbal working memory: evidence from neuroimaging. Proc Natl Acad Sci U S A, 95(3), 876-882.

Sobell, L. C., Sobell, M. B., Leo, G. I., & Cancilla, A. (1988). Reliability of a timeline method: assessing normal drinkers' reports of recent drinking and a comparative evaluation across several populations. Br J Addict, 83(4), 393-402.

Sokolov, B. P. (1998). Expression of NMDAR1, GluR1, GluR7, and KA1 glutamate receptor mRNAs is decreased in frontal cortex of "neuroleptic-free" schizophrenics: evidence on reversible up-regulation by typical neuroleptics. J Neurochem, 71(6), 2454-2464.

Solowij, N., Jones, K. A., Rozman, M. E., Davis, S. M., Ciarrochi, J., Heaven, P. C., . . . Yucel, M. (2011). Verbal learning and memory in adolescent cannabis users, alcohol users and non-users. Psychopharmacology (Berl), 216(1), 131-144.

Page 133: Evaluating The Effects of Cannabis Dependence and ...

120  

Solowij, N., & Michie, P. T. (2007). Cannabis and cognitive dysfunction: parallels with endophenotypes of schizophrenia? J Psychiatry Neurosci, 32(1), 30-52.

Soubasi, E., Chroni, E., Gourzis, P., Zisis, A., Beratis, S., & Papathanasopoulos, P. (2010). Cortical motor neurophysiology of patients with schizophrenia: a study using transcranial magnetic stimulation. Psychiatry Res, 176(2-3), 132-136.

Stephens, R. S., Roffman, R. A., & Curtin, L. (2000). Comparison of extended versus brief treatments for marijuana use. J Consult Clin Psychol, 68(5), 898-908.

Steriade, M., Amzica, F., Neckelmann, D., & Timofeev, I. (1998). Spike-wave complexes and fast components of cortically generated seizures. II. Extra- and intracellular patterns. J Neurophysiol, 80(3), 1456-1479.

Stirling, J., Lewis, S., Hopkins, R., & White, C. (2005). Cannabis use prior to first onset psychosis predicts spared neurocognition at 10-year follow-up. Schizophr Res, 75(1), 135-137.

Strube, W., Wobrock, T., Bunse, T., Palm, U., Padberg, F., Malchow, B., . . . Hasan, A. (2014). Impairments in motor-cortical inhibitory networks across recent-onset and chronic schizophrenia: a cross-sectional TMS Study. Behav Brain Res, 264, 17-25.

Sun, Y., Farzan, F., Barr, M. S., Kirihara, K., Fitzgerald, P. B., Light, G. A., & Daskalakis, Z. J. (2011). gamma oscillations in schizophrenia: mechanisms and clinical significance. Brain Res, 1413, 98-114.

Szabo, B., & Schlicker, E. (2005). Effects of cannabinoids on neurotransmission. Handb Exp Pharmacol(168), 327-365.

Takahashi, S., Ukai, S., Kose, A., Hashimoto, T., Iwatani, J., Okumura, M., . . . Shinosaki, K. (2013). Reduction of cortical GABAergic inhibition correlates with working memory impairment in recent onset schizophrenia. Schizophr Res, 146(1-3), 238-243.

Tallon-Baudry, C., Bertrand, O., Peronnet, F., & Pernier, J. (1998). Induced gamma-band activity during the delay of a visual short-term memory task in humans. J Neurosci, 18(11), 4244-4254.

Tallon-Baudry, C., Kreiter, A., & Bertrand, O. (1999). Sustained and transient oscillatory responses in the gamma and beta bands in a visual short-term memory task in humans. Vis Neurosci, 16(3), 449-459.

Tandon, R., Keshavan, M. S., & Nasrallah, H. A. (2008). Schizophrenia, "just the facts" what we know in 2008. 2. Epidemiology and etiology. Schizophr Res, 102(1-3), 1-18.

Tart, C. T. (1970). Marijuana intoxication common experiences. Nature, 226(5247), 701-704. Thomas, A., Baillie, G. L., Phillips, A. M., Razdan, R. K., Ross, R. A., & Pertwee, R. G. (2007).

Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol, 150(5), 613-623.

Tiitinen, H., Sinkkonen, J., Reinikainen, K., Alho, K., Lavikainen, J., & Naatanen, R. (1993). Selective attention enhances the auditory 40-Hz transient response in humans. Nature, 364(6432), 59-60.

Tse, M. T., Piantadosi, P. T., & Floresco, S. B. (2014). Prefrontal Cortical Gamma-Aminobutyric Acid Transmission and Cognitive Function: Drawing Links to Schizophrenia from Preclinical Research. Biol Psychiatry.

Tsou, K., Brown, S., Sanudo-Pena, M. C., Mackie, K., & Walker, J. M. (1998). Immunohistochemical distribution of cannabinoid CB1 receptors in the rat central nervous system. Neuroscience, 83(2), 393-411.

Uhlhaas, P. J., Linden, D. E., Singer, W., Haenschel, C., Lindner, M., Maurer, K., & Rodriguez, E. (2006). Dysfunctional long-range coordination of neural activity during Gestalt perception in schizophrenia. J Neurosci, 26(31), 8168-8175.

Uhlhaas, P. J., & Singer, W. (2006). Neural synchrony in brain disorders: relevance for cognitive dysfunctions and pathophysiology. Neuron, 52(1), 155-168.

Uhlhaas, P. J., & Singer, W. (2010). Abnormal neural oscillations and synchrony in schizophrenia. Nat Rev Neurosci, 11(2), 100-113.

Page 134: Evaluating The Effects of Cannabis Dependence and ...

121  

United Nations Office on Drugs and Crime. (2009). World Drug Report Vienna: United Nations Vadhan, N. P., Hart, C. L., van Gorp, W. G., Gunderson, E. W., Haney, M., & Foltin, R. W. (2007).

Acute effects of smoked marijuana on decision making, as assessed by a modified gambling task, in experienced marijuana users. J Clin Exp Neuropsychol, 29(4), 357-364.

Van Hoozen, B. E., & Cross, C. E. (1997). Marijuana. Respiratory tract effects. Clin Rev Allergy Immunol, 15(3), 243-269.

Van Snellenberg, J. X., Torres, I. J., & Thornton, A. E. (2006). Functional neuroimaging of working memory in schizophrenia: task performance as a moderating variable. Neuropsychology, 20(5), 497-510.

Verdejo-Garcia, A., Bechara, A., Recknor, E. C., & Perez-Garcia, M. (2006). Executive dysfunction in substance dependent individuals during drug use and abstinence: an examination of the behavioral, cognitive and emotional correlates of addiction. J Int Neuropsychol Soc, 12(3), 405-415.

Verdejo-Garcia, A., Fagundo, A. B., Cuenca, A., Rodriguez, J., Cuyas, E., Langohr, K., . . . de la Torre, R. (2013). COMT val158met and 5-HTTLPR genetic polymorphisms moderate executive control in cannabis users. Neuropsychopharmacology, 38(8), 1598-1606.

Verdejo-Garcia, A., Rivas-Perez, C., Lopez-Torrecillas, F., & Perez-Garcia, M. (2006). Differential impact of severity of drug use on frontal behavioral symptoms. Addict Behav, 31(8), 1373-1382.

Wang, X. J., & Buzsaki, G. (1996). Gamma oscillation by synaptic inhibition in a hippocampal interneuronal network model. J Neurosci, 16(20), 6402-6413.

Wechsler, D. (2001). Wechsler Test of Adult Reading. . San Antonio: Psychological Corporation. Weinberger, A. H., Reutenauer, E. L., Allen, T. M., Termine, A., Vessicchio, J. C., Sacco, K. A., . . .

George, T. P. (2007). Reliability of the Fagerstrom Test for Nicotine Dependence, Minnesota Nicotine Withdrawal Scale, and Tiffany Questionnaire for Smoking Urges in smokers with and without schizophrenia. Drug Alcohol Depend, 86(2-3), 278-282.

Whiteford, H. A., Degenhardt, L., Rehm, J., Baxter, A. J., Ferrari, A. J., Erskine, H. E., . . . Vos, T. (2013). Global burden of disease attributable to mental and substance use disorders: findings from the Global Burden of Disease Study 2010. Lancet, 382(9904), 1575-1586.

Whittington, M. A., Traub, R. D., & Jefferys, J. G. (1995). Synchronized oscillations in interneuron networks driven by metabotropic glutamate receptor activation. Nature, 373(6515), 612-615.

Wilson, W., Mathew, R., Turkington, T., Hawk, T., Coleman, R. E., & Provenzale, J. (2000). Brain morphological changes and early marijuana use: a magnetic resonance and positron emission tomography study. J Addict Dis, 19(1), 1-22.

Wobrock, T., Hasan, A., Malchow, B., Wolff-Menzler, C., Guse, B., Lang, N., . . . Falkai, P. (2010). Increased cortical inhibition deficits in first-episode schizophrenia with comorbid cannabis abuse. Psychopharmacology (Berl), 208(3), 353-363.

Wobrock, T., Schneider, M., Kadovic, D., Schneider-Axmann, T., Ecker, U. K., Retz, W., . . . Falkai, P. (2008). Reduced cortical inhibition in first-episode schizophrenia. Schizophr Res, 105(1-3), 252-261.

Wobrock, T., Schneider-Axmann, T., Retz, W., Rosler, M., Kadovic, D., Falkai, P., & Schneider, M. (2009). Motor circuit abnormalities in first-episode schizophrenia assessed with transcranial magnetic stimulation. Pharmacopsychiatry, 42(5), 194-201.

Wolff, K., & Johnston, A. (2014). Cannabis use: a perspective in relation to the proposed UK drug-driving legislation. Drug Test Anal, 6(1-2), 143-154.

Yucel, M., Bora, E., Lubman, D. I., Solowij, N., Brewer, W. J., Cotton, S. M., . . . Pantelis, C. (2012). The impact of cannabis use on cognitive functioning in patients with schizophrenia: a meta-analysis of existing findings and new data in a first-episode sample. Schizophr Bull, 38(2), 316-330.

Page 135: Evaluating The Effects of Cannabis Dependence and ...

122  

Yucel, M., Zalesky, A., Takagi, M. J., Bora, E., Fornito, A., Ditchfield, M., . . . Lubman, D. I. (2010). White-matter abnormalities in adolescents with long-term inhalant and cannabis use: a diffusion magnetic resonance imaging study. J Psychiatry Neurosci, 35(6), 409-412.

Zanelati, T. V., Biojone, C., Moreira, F. A., Guimaraes, F. S., & Joca, S. R. (2010). Antidepressant-like effects of cannabidiol in mice: possible involvement of 5-HT1A receptors. Br J Pharmacol, 159(1), 122-128.

Zavitsanou, K., Garrick, T., & Huang, X. F. (2004). Selective antagonist [3H]SR141716A binding to cannabinoid CB1 receptors is increased in the anterior cingulate cortex in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry, 28(2), 355-360.

Ziemann, U., Rothwell, J. C., & Ridding, M. C. (1996). Interaction between intracortical inhibition and facilitation in human motor cortex. J Physiol, 496 ( Pt 3), 873-881.

Zuardi, A. W. (2006). History of cannabis as a medicine: a review. Rev Bras Psiquiatr, 28(2), 153-157.

Zuardi, A. W., Crippa, J. A., Hallak, J. E., Moreira, F. A., & Guimaraes, F. S. (2006). Cannabidiol, a Cannabis sativa constituent, as an antipsychotic drug. Braz J Med Biol Res, 39(4), 421-429.

Zuardi, A. W., Shirakawa, I., Finkelfarb, E., & Karniol, I. G. (1982). Action of cannabidiol on the anxiety and other effects produced by delta 9-THC in normal subjects. Psychopharmacology (Berl), 76(3), 245-250.

Page 136: Evaluating The Effects of Cannabis Dependence and ...

123  

Appendix A: Telephone Screen

Screen ID: Sz Screen date:

Interviewer’s Initials

Eligible Recontact

Y N

Y N

Name (first/last): Phone no H/W/C.:

Age: ________ (16 to 55) DOB (dd/m/yr) :__________________________ Gender: M (exclude females)

Referral Source: ______________

SMOKING:

Do you smoke tobacco? No (exclude), Yes.

On average, how many cigarettes do you smoke in a day/week?

PSYCHIATRIC HISTORY: (Diagnosis of schizophrenia or schizoaffective disorder, currently in TX)

Have you ever had any psychiatric treatment? No Yes, currently

Diagnosis? ____________________ Clinician: ______________________ Program: ______________

Last hospitalization for psychiatric symptoms? ________________________

Current Medications (dose and recent changes) _____________________________________________

(Must be on stable antipsychotic dose for at least one month, if not, re-contact)

Have you ever had a heady injury with LOC? No Yes details

Did you black-out? For how long? Did you need to be hospitalized?

Are you experiencing medical problems at this time? No Yes details

Do you have any metal in your body? No Yes details

Page 137: Evaluating The Effects of Cannabis Dependence and ...

124  

CURRENT SUBSTANCE USE:

Marijuana: ............................................... No (exclude) Yes, assess for current dependence:

Frequency and duration of use:________________________

• substance is taken in larger amounts or over long period than intended

• persistent desire to quit or cut down • majority of time spent involved in activities of obtaining/using/recovering • social, occupational, recreational activities given up or reduced • continued use despite physical and psychological problems • tolerance: needed for same effect/ ↓effect with same amount • withdrawal

Yes to 3 of above symptoms No (exclude), Yes

Are you currently actively trying to quite marijuana? (treatment-seeking are not eligible and should be referred elsewhere)

No Yes (exclude),

CURRENT/PAST SUBSTANCE USE:

Alcohol: ..................................................... Current:_____________ (exclude: M>14/ wk) Past: ____________

Cocaine: ..................................................... Current: Y (exclude) N Past: _____________________

Opiates (heroin, p-dope, percocets): .............. Current: Y (exclude) N Past: _____________________

Methadone: ................................................ Current: Y (exclude) N Past: _____________________

“Pills” (valium, xanax, etc): .......................... Current: Y (exclude) N Past: _____________________

Other (acid, speed, illy, etc): ......................... Current Y (exclude) N Past: _____________________

Cannabis Abstinence Study Eligibility:

No Yes Yes, possibly Availability:

Would you be willing to quite marijuana for a one-month period?

IN-PERSON appointment SCHEDULED: No Yes TBA (pending)

If Yes: Appointment date, time, RA:

Consent to contact for participation in future studies or add to Sz research registry

Y N

Y N

Y N

Y N

Y N

Y N

Y N

Page 138: Evaluating The Effects of Cannabis Dependence and ...

125  

Appendix B: Study Consent

STUDY INFORMATION AND CONSENT FORM:

Effects of Cannabis Abstinence on Neurocognition in Schizophrenia: Neurophysiology Assessment

Investigators Contact Phone Number Tony P. George, MD, FRCPC 416-535-8501 x 34544

Zafiris J. Daskalakis, MD, PhD, FRCPC 416-535- 8501 x 34319

Rachel A. Rabin, MSc 416-535-8501 x 36115

Mera S. Barr, PhD 416-535-8501 x 33095

Study Purpose:

The use of cannabis influences the brain’s ability to concentrate, pay attention, reason, and remember. Collectively these are known as cognitive functions. How cannabis affects these cognitive functions are still unclear.

Cannabis may influence brain mechanisms called cortical inhibition and plasticity, which in turn, influences cognitive function. Cortical inhibition is a brain process that is designed to filter excessive information in our environment in order to process information efficiently. Plasticity is involved in learning and memory and. It has been discovered that cortical inhibition and plasticity may impaired in the brains of people who use cannabis. Cortical inhibition and plasticity is also impaired in patients with schizophrenia who suffer from cognitive impairment as part of their illness.

Transcranial magnetic stimulation (TMS) is a method used to measure brain inhibition. TMS excites nerves over the area of the brain that is responsible for thinking and planning. When the nerves are stimulated, this causes brain activity to change, which will be recorded with electromyography (EMG) and during electroencephalography (EEG), which will be analyzed later.

Page 139: Evaluating The Effects of Cannabis Dependence and ...

126  

The purpose of this study is to measure cortical inhibition, plasticity and cognitive function in patients and control participants who are cannabis dependent. These measures will then be evaluated during a one month period of abstinence. We hope that this study will help us understand the interaction between cannabis, cognition, and schizophrenia. This is not part of any treatment plan.

Procedures: You are being asked to provide informed consent. After reading through this form, you will be given a chance to ask questions. All study procedures will be completed at Dr. George’s BACDRL lab (1st floor, 33 Russell Street, Toronto, ON, Room 1910A) and at the Temerty Centre for Therapeutic Brain Intervention (1001 Queen Street West, Unit 4-1, Room 124) at the Centre for Addiction and Mental Health. The study will take place over a 1-month period + a one-month follow-up, where you will be asked to come into the lab. These sessions will either take place on the same day as primary assessments or can be done within the same week on a separate day. The assessments will be conducted by Ms Michelle Goodman, Ms. Rachel Rabin and Dr. Mera Barr. Ms Goodman and Rabin are graduate students in the Institute of Medical Science at the University of Toronto and the Centre for Addiction and Mental Health, Schizophrenia Program, and this study is part of their graduate research program. Rachel Rabin will lead the project and be responsible for recruitment, enrolment and screening of participants, as well as assessments and cognitive testing. Ms. Michelle Goodman will be responsible for the neurophysiological assessment conducted at the Temerty Centre. Dr. Mera Barr is an Independent Scientist in the Schizophrenia Division of the Complex Mental Illness Program at CAMH. Dr. Mera Barr will oversee the assessment of cortical inhibition and plasticity with TMS and measure the memory task with electroencephalography (EEG). Drs. George and Daskalakis are research psychiatrists in the CAMH Schizophrenia Division who act as supervising and medically qualified investigators for this study.

DT-MRI

The sessions to measure cortical inhibition in your brain will be conducted at baseline prior to the initiation of cannabis abstinence, once a week for 4 weeks (during the one month abstinence), and one month post abstinence.

• The MRI will be done at the Centre for Addiction and Mental Health. The MRI scanning is a technique of taking pictures of the brain that gives information on brain structure and brain function.

• Before the scan begins, you will be asked to remove all metal that you are wearing because MRI generates images using a strong magnetic field and this can damage some electrical devices.

• You will then be asked to lie on a padded bed that will be moved into a tunnel-like

Page 140: Evaluating The Effects of Cannabis Dependence and ...

127  

machine for the MRI scan. The tunnel is not very large, and the study requires you to lie still in the scanner.

• You should try to remain as still as possible during the scan. Movements will not be dangerous to you in any way, but would blur the picture of your brain.

• You will be in the scanner for about 60 minutes. • You might hear moderately loud knocking or beeping during the scan when the MRI

machine is in operation. These sounds reflect the normal functioning of the MRI. • MRI scanning is not associated with any known risks to your health, and there is no

proof that there will be short-term or long-term side effects. • This will be done before and after 28 days of abstinence.

TMS TMS sessions will measure cortical inhibition and plasticity in your brain. At baseline and following cannabis abstinence, we will measure cortical inhibition and plasticity using TMS and EEG. Once a week during the one month abstinence (4 testing sessions) and 1 month post abstinence we will conduct the TMS session without EEG. The TMS and EEG sessions at baseline and following cannabis abstinence will take approximately 8 hours. The weekly TMS sessions and one month follow up will take approximately 30 minutes.

Cortical Inhibition

• We will attach soft foam electrodes to the skin surface over your hand muscles, and then connect these to a recorder that will record the activity of your hand muscles (for the first part, then TMS will be applied to the front of your head)

• A magnetic coil will be held on the surface of your scalp • To obtain brain inhibition, different modes of TMS will be applied to the head

surface and EMG will be recorded from your hand muscle. • When the magnetic stimulation is applied you will feel a twitch or small movement in

your hand but no pain. • There are many pulses but each pulse is very short and not too close to the next.

Plasticity

• This set up will be the same as measuring cortical inhibition but with an additional electrode placed on your right wrist that will deliver low level electrical stimuli prior to the delivery of the TMS pulse

• There will be several pulses with TMS alone and TMS combined with the electrical stimulus

• This will take approximately 3-4 hours.

EEG

Page 141: Evaluating The Effects of Cannabis Dependence and ...

128  

The sessions to measure TMS cortical inhibition, working memory, and mismatch negativity will be conducted at baseline prior to the initiation of cannabis abstinence and week 4 of abstinence.

• EEG or electroencephalography is a measurement of the electrical activity of the brain. • We will use this to look at brain activity during TMS brain stimulation. • You will be seated in a comfortable chair and will have on a cap containing many

electrodes that record your brain activity. It takes approximately 30 minutes to put on the cap and to prepare for recording.

• There is gel on the inside of the cap that may be sticky. Shampoo will be provided after the testing session to wash your hair.

• You will be asked to rest comfortably while the TMS is applied and the computer is recording information. The combination of TMS and EEG will take approximately 4.5 hours.

Working Memory Task

• We will use this to look at brain activity while at rest and during a working memory task.

• You will be seated in a comfortable chair and will have on a cap containing many recorders that record your brain activity. It takes approximately 15 minutes to put on the cap and get it ready for recording.

• There is gel on the inside of the cap that may be sticky; you will be allowed to rinse it out after the test.

• The recorders are attached to a computer to record the information. • This will last approximately 1.5 hours.

Mismatch Negativity Task (MMN)

• We will use this to look at brain activity that takes place when you are not directly paying attention to auditory stimuli.

• You will be seated in a comfortable chair with the EEG cap and two foam earphones placed in the ears.

• Auditory tones will be played in both ears while you watch a silent cartoon video. • This will last approximately 45 minutes.

Are there any side effects or pain associated with TMS, EEG, or MRI?

There may be some mild side effects with transcranial magnetic stimulation. At certain positions on the head, the stimulation may cause your eyes to blink or a brief contraction of the scalp, neck, trunk or upper arm muscles. You may find these contractions annoying, but they should not be painful. Some people may experience mild headache or shoulder stiffness after testing, however, these symptoms will usually go away in time. Acetaminophen

Page 142: Evaluating The Effects of Cannabis Dependence and ...

129  

(Tylenol) is effective in treating these side effects. If you have further concerns you may contact your study doctor at any time. The magnetic stimulator makes a clicking noise when it stimulates. Since this may be distracting, you will be offered ear-plugs which mute the sound. Magnetic brain stimulation has been used on thousands of individuals in the United States, Canada and Europe over several years without any serious problems. There are no serious side effects for EEG. Some people report a headache following the procedure but this is only due to the pressure of wearing a cap and not the procedure itself. MRI is a non-invasive procedure. Certain items such as a pacemaker and having surgical staples or other ferrous metal in or near the head are dangerous with an MRI. For this reason you will be screened using the precautionary screen for these items designed by the MRI Unit at CAMH.

Risks

You should be aware that the magnetic fields generated by the stimulator may damage magnetic cards, watches and some electrical devices. Please remove any such items before testing.

Exposure to magnetic stimulation or any strong magnetic field is not permitted in people who have a pacemaker, an implanted medication pump, a metal plate in the skull, or metal objects inside the eye or skull (for example, after brain surgery or a shrapnel wound). Please inform the investigators if you might have any of these.

Magnetic brain stimulation has caused brief epileptic seizures in a few patients with stroke and epilepsy out of thousands of people tested. This has never happened with the form of TMS you will be receiving. Patients with epilepsy and patients with stroke can have an increased risk of seizures. Therefore, there is a very small possibility that magnetic stimulation may cause seizures in persons with a heightened risk. The investigators cannot promise that this will never happen in the future; however, the probability of this occurring in the experiment is low. As we have stated above, these tests have been used in thousands of healthy volunteers all over the world and there is no report of a seizure in healthy people.

Risks of TMS

Common Headache and Scalp pain

Tingling sensation and/or heat at coil location

Remote

Effects of Hearing (earplugs are worn to prevent this)

Syncope

Seizure (no reports in current literature during paired-pulse TMS)

Page 143: Evaluating The Effects of Cannabis Dependence and ...

130  

You might feel slight fatigue during the testing sessions. While a cannabis withdrawal syndrome has been reported it does not include significant physical, medical, or psychiatric problems. You may experience irritability, anger, depression, difficulty sleeping, craving, and decreased appetite. Headaches, physical tension, sweating, stomach pain, and general physical discomfort have also been observed during cannabis withdrawal, but are less common.

Benefits:

The information you provide will improve our knowledge about the effects of cannabis abstinence in individuals with schizophrenia, and might encourage you to want to quit using cannabis. Voluntary Participation & Subject Obligations: You are free to choose not to participate, and may withdraw from this study at any time. If you withdraw, it will not affect your ability to receive treatment at CAMH. Study staff/investigators may, at their discretion, end your participation in this study at any time. If you are an existing CAMH client, you will also be asked to give the research team permission to access your medical records for the purpose of confirming your medication and treatment status. Study Provisions: You will be paid for attending and completion of each study visit at a rate of $10/ hour for a total of 9 hours. If all visits are attended for a total of $220.

Page 144: Evaluating The Effects of Cannabis Dependence and ...

131  

Payment Schedule: $10/hour for a total of $200

Assessment

Length (UP TO; h)

Assessment

Completion (UP TO)

Baseline: PAS,TMS-EEG, Working Memory, MMN, and fMRI

9 hours $90

Wk 1 TMS 1 hour $10

Wk 2 TMS 1 hour $10

Wk 3 TMS 1 hour $10

Wk 4: PAS TMS-EEG Working Memory, MMN, and fMRI

9 hours $90

Wk 8: TMS 1 hours $10

TOTAL 22 hours $220

Confidentiality:

If you decide to take part in this research study, you will be required to answer some questions about your drug use and problems you may be having relating to drug use. Your answers to these questions, as well as other data collected will only be used by the study investigators and their designates and will remain confidential to the extent permitted by law.

• As part of continuing review of the research, your study records may be assessed on behalf of the Research Ethics Board. A person from the research ethics team may contact you to ask you questions about the research study and your consent to participate. The person assessing your file or contacting you must maintain your confidentiality to the extent permitted bylaw.

• In accordance with federal requirements, CAMH will maintain archived study records for 10 years. However, those documents that contain personal identifiers (i.e. consent forms) will be stored separately from data files.

• General results of this study might be published, but will not identify you by name.

Page 145: Evaluating The Effects of Cannabis Dependence and ...

132  

As part of the Research Services Quality Assurance Program, this study may be monitored and/or audited by a member of the Quality Assurance Team. Your research records and CAMH records may be reviewed during which confidentiality will be maintained as per CAMH policies and extent permitted by law.

Contact: If you have any further questions or desire further information about this study, you may contact Rachel Rabin at 416-535-8501 x 36115, Dr. Mera Barr at 416-535-8501 x 33095 or the Principal Investigator Dr. Tony George at 416-535-8501 x 34544. If you have any questions about your rights as a study participant, you may contact Dr. Padraig Darby, Chair of the Research Ethics Board, Centre for Addiction & Mental Health, at 416-535-8501 (x36876).

Page 146: Evaluating The Effects of Cannabis Dependence and ...

133  

AGREEMENT TO PARTICIPATE

I, ___________________________________have read (or had read to me) the information form for the study named Effects of Cannabis Abstinence on Neurophysiology in Schizophrenia: Neurophysiology Assessment. I understand that my role is that of a participant in this study. I have been given an opportunity to ask questions about this study. Any questions that I have had, have been answered to my satisfaction, so that I now understand the study procedures, the potential risks of participating, and my right to the confidential treatment of the information that is collected about me. I also understand that my participation in this study is entirely voluntary, and that I may refuse to participate or withdraw from the study at any time, without any consequences for my continuing care. By signing this consent form, I do not waive any of my legal rights nor relieve the investigators/institution from legal responsibilities. If I have any questions about my rights as a study participant, I may contact Dr. Padraig Darby, Chair of the Research Ethics Board, Centre for Addiction and Mental Health, at 416- 535 8501, extension 6876.

I have received a copy of this consent form for my own record.

Participant’s Initials:________

Participant Name: Person who conducted informed consent discussion:

Print name Print name

Date:

Signature of Participant Signature of Witness

 

 

 

Page 147: Evaluating The Effects of Cannabis Dependence and ...

134  

Appendix C: Case Report Form

Effects of Cannabis Abstinence on Neurocognition in Schizophrenia: Neurophysiology Assessment

Subject ID: ___________________________

Testing Completed by: __________________

Time Point: Week 0

Date: ________________________________

Time of Last cannabis smoked: ___________ AM PM

Testing Time Start: _____________________ AM PM

Testing Time End: ______________________ AM PM

Without Cap

Resting Motor Threshold: ________________

1 mV Peak to Peak: ________________

SICI: ___________________________________________________________________

LICI: ___________________________________________________________________

CSP: ___________________________________________________________________

With Cap

Resting Motor Threshold: ________________

1 mV Peak to Peak: ________________

Rest:

LICI Motor Single: _______________________________________________________

LICI Motor Paired: _______________________________________________________

ICF Motor Paired: ________________________________________________________

LICI DLPFC Single: ______________________________________________________

LICI DLPFC Paired: ______________________________________________________

ICF DLPFC Paired: _______________________________________________________

Page 148: Evaluating The Effects of Cannabis Dependence and ...

135  

Nback 1-back: ___________________________________________________________

3-back: ___________________________________________________________

Time Point: Week 0

Comments: ________________________________________________________________________________________________________________________________________________________________________________________________________________________________________________________________________________________________


Recommended