pharmacogenetic analysis of glutamate system gene … · 2016-11-15 · pharmacogenetic analysis of...

191
PHARMACOGENETIC ANALYSIS OF GLUTAMATE SYSTEM GENE VARIANTS AND CLINICAL RESPONSE TO CLOZAPINE by Danielle L. Taylor A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Science University of Toronto © Copyright by Danielle L. Taylor (2015)

Upload: others

Post on 18-Apr-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

PHARMACOGENETIC ANALYSIS OF GLUTAMATE SYSTEM GENE VARIANTS AND CLINICAL RESPONSE

TO CLOZAPINE

by

Danielle L. Taylor

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

Institute of Medical Science University of Toronto

© Copyright by Danielle L. Taylor (2015)

Taylor DL

ii"""

Pharmacogenetic Analysis of Glutamate System Gene Variants

and Clinical Response to Clozapine

Danielle L. Taylor

Master of Science

Institute of Medical Science University of Toronto

2015

ABSTRACT

Altered glutamatergic neurotransmission is implicated in schizophrenia etiology and

response to the atypical antipsychotic clozapine. Response to clozapine is highly variable and

partly depends on genetic factors as indicated by twin studies. The association of 17

glutamate system gene variants and clinical response to clozapine was investigated in a

sample of European schizophrenia/schizoaffective patients deemed resistant/intolerant to

previous pharmacotherapy. Categorical and continuous response measures were assessed

following six months of clozapine treatment using change in Brief Psychiatric Rating Scale

Scores, and were analyzed using Pearson’s chi-squared test or analysis of covariance,

respectively. No significant associations were observed for all analyses after correction for

multiple testing. Prior to correction, several nominally significant associations were

observed, the most notable being between the SLC6A9 rs16831558 variant and change in

positive symptoms (puncorrected=0.008, pcorrected=0.08). This finding warrants further

investigation in larger well-characterized samples to clarify the role of SLC6A9 in clinical

response to clozapine.

Taylor DL

iii""

ACKNOWLEDGEMENTS

First and foremost, I would like to thank my thesis supervisor Dr. James Kennedy for

providing me with the opportunity to undertake a Master’s degree in his lab. His continued

support, guidance and witty sense of humor have made these last few years in graduate

school an altogether enriching experience. I would also like to thank the members of my

Program Advisory Committee, Dr. Joanne Knight, Dr. Daniel Müller and Dr. Gary

Remington for their patience and constructive feedback. I extend many thanks to my fellow

colleagues in the Neurogenetics Department for providing guidance on protocols and

statistical analyses, and for helping to troubleshoot obstacles that popped up along the way.

To the Pharmacogenetics crew, thank you all for the many laughs, good food and good times.

Much recognition goes out to my friends on the volleyball team for providing a carefree

environment where I could forget about the worries of the work week. Thank you to my

family for continuing to support me on my long scholarly journey. Finally, I would like to

acknowledge the lab technicians, trainees, collaborators, faculty mentors and funding sources

for their contribution to this project.

Taylor DL

iv"""

CONTRIBUTIONS

Dr. James Kennedy, Dr. Daniel Müller, Dr. Joanne Knight and Dr. Gary Remington helped

design this thesis project. Patient blood samples were provided by Dr. Herbert Meltzer, Dr.

Jeffrey Lieberman and Dr. Steven Potkin. Dr. Arun Tiwari provided guidance regarding

statistical analyses of genotype data and Dr. Vanessa Gonçalves provided guidance regarding

use of the Applied Biosystems OpenArray® genotyping platform. A very special thank you

is extended to the patients for their willingness to participate.

Taylor DL

v"""

TABLE OF CONTENTS

CHAPTER SECTION PAGE

ABSTRACT ii

ACKNOWLEDGEMENTS iii

CONTRIBUTIONS iv

TABLE OF CONTENTS v

LIST OF ABBREVIATIONS viii

LIST OF FIGURES xix

LIST OF TABLES xx

1 INTRODUCTION 1 1.1 Schizophrenia Overview 1

1.1.1 Epidemiology and Time Course 2 1.1.2 Deficits in Quality of Life 3 1.1.3 Burden to Society, Patients and Caregivers 4

1.2 Etiology and Pathophysiology 5 1.2.1 Environmental Risk Factors 5 1.2.2 Genetic Predisposition 5

1.2.2.1 Chromosome 6: 6p24-p22 7 1.2.2.2 Chromosome 8: 8p22-p21 8 1.2.2.3 Chromosome 11: 11q22-q21 8 1.2.2.4 Chromosome 13: 13q14-q32 8 1.2.2.5 Polygenic Inheritance 9

1.2.3 Brain Structural Abnormalities 11 1.2.4 Neurochemical Abnormalities 11

1.2.4.1 The Dopamine Hypothesis 11 1.2.4.2 The Glutamate Hypothesis 13

1.3 Glutamatergic Dysfunction in Schizophrenia 14 1.3.1 The Glutamate System 14

1.3.1.1 Ionotropic Glutamate Receptors 16 1.3.1.2 Metabotropic Glutamate Receptors 20 1.3.1.3 Amino Acid Transporters 21 1.3.1.4 Glutamate System Enzymes 23

1.3.2 Glutamate System Abnormalities in Schizophrenia 24 1.4 Schizophrenia Pharmacotherapy 26

1.4.1 Antipsychotic Medication Overview 26

Taylor DL

vi"""

1.4.2 The Mechanism of Action of Antipsychotic Drugs 27 1.4.3 Treatment Response Measures 28 1.4.4 Clozapine for Treatment-Resistant Schizophrenia 29 1.4.5 Variability in Clozapine Response 31

1.5 Pharmacogenetics of Clozapine Response 33 1.5.1 Pharmacogenetics Overview 33 1.5.2 Clozapine and Glutamatergic Neurotransmission 34 1.5.3 Polymorphisms in Glutamate System and Related Genes 38 1.5.4 Identifying Functional Gene Variants Using ENCODE 40

1.6 Study Rationale 44 1.6.1 Summary and Hypothesis 44

2 GENETIC ASSOCIATION ANALYSIS OF THE NMDA 45

RECEPTOR SUBUNIT GENE GRIN2B WITH RESPONSE TO CLOZAPINE (Submitted to Human Psychopharmacology: Clinical & Experimental) 2.1 Abstract 46 2.2 Introduction 47 2.3 Methods 50 2.4 Results 55 2.5 Discussion 56

3 PHARMACOGENETIC ANALYSIS OF GLUTAMATE 67

SYSTEM GENE VARIANTS AND CLINICAL RESPONSE TO CLOZAPINE (Submitted to Molecular Neuropsychiatry) 3.1 Abstract 68 3.2 Introduction 69 3.3 Methods 72 3.4 Results 76 3.5 Discussion 79

4 DISCUSSION 95

4.0 Overview 95 4.1 Summary of Findings 95

4.1.1 Association of GRIN2B with Clozapine Response 95 4.1.2 Association of Glutamate System Variants with Clozapine 96

Response 4.2 Implications of Findings 97

4.2.1 Glycine in Schizophrenia Etiology and Treatment 98 4.2.2 Interaction of Clozapine with the Glycine System 99

Taylor DL

vii""

4.2.3 Promoter Variant Confers Differential Response to 100 Clozapine

4.3 Limitations & Considerations 102 4.3.1 Study Heterogeneity 102 4.3.2 Inter-Study Variability 104 4.3.3 Statistical Power 106 4.3.4 Multiple Testing 108 4.3.5 Deviation from Hardy-Weinberg Equilibrium 110 4.3.6 Incomplete Outcome Data 111

4.4 Future Directions 112 4.4.1 Characterization of Glycine Transporter 1 Promoter 112

Variant 4.4.2 Gene-Environment Interactions 115 4.4.3 Gene-Gene Interaction s 117 4.4.4 Next Generation Sequencing 120 4.4.5 Personalized Medicine in Psychiatry 123

4.5 Concluding Remarks 126

5 REFERENCES 128

Taylor DL

viii""

LIST OF ABBREVIATIONS

Δ change

α alpha; probability associated with a type I error, statistical significance criteria

α-keto acid α-ketoglutaric acid

α-KG alpha-ketoglutarate

β probability associated with making a type II error

γ gamma

oC" degrees Celsius

µL microlitre

χ2 Pearson’s chi-squared goodness of fit test

5-HT1A 5-hydroxytryptamine (serotonin) type 1A receptor

5-HT2A 5-hydroxytryptamine (serotonin) type 2A receptor

5-HTTLPR serotonin-transporter-linked polymorphic region

A adenine nucleotide base

ABI Applied Biosystems Incorporated

Ac-CoA acetyl coenzyme A

AD antidepressant

ADHD attention deficit hyperactivity disorder

ADRA1A adrenergic receptor α-1°

ADRA2A adrenergic receptor α-2°

AFR African American sample

AIWG antipsychotic-induced weight gain

AMPA α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor

AMR ad mixed American sample

Taylor DL

ix""

ANCOVA analysis of covariance

ANOVA analysis of variance

AP antipsychotic

APA American Psychiatric Association

ASD autism spectrum disorder

ASN Asian sample

BDNF brain derived neurotrophic factor gene

BDNF brain derived neurotrophic factor protein

Bill S-201 Genetic Non-discrimination Act (Canada)

BNEG Brief Psychiatric Rating Scale negative symptom subscale

bp nucleotide basepairs

BP bipolar disorder

BPOS Brief Psychiatric Rating Scale positive symptom subscale

BPRS Brief Psychiatric Rating Scale

C cytosine nucleotide base

Ca2+ calcium ion

CAMH Centre for Addiction and Mental Health

CaMKII calcium/calmodulin dependent kinase II

cAMP cyclic adenosine monophosphate

CATIE Clinical Antipsychotic Trials of Intervention Effectiveness Project

CDN Canadian dollars

cDNA complimentary deoxyribonucleic acid

CGI Clinical Global Impressions Scale

ChIP-seq chromatin immunoprecipitation sequencing

CHOR Clozapine, Haloperidol, Olanzapine, Risperidone Study

Taylor DL

x"""

CHRM1 cholinergic receptor, muscarinic 1

chrN chromosome number

CI confidence interval

CIA clozapine-induced agranulocytosis

CI-OCD clozapine-induced obsessive compulsive disorder

CLZ clozapine

CNS central nervous system

Cons evolutionarily conserved variant

COS childhood onset schizophrenia

CPZ chlorpromazine

CREB cyclic AMP response element-binding protein

CRESTAR clozapine pharmacogenetics study

CSF cerebrospinal fluid

CVD cardiovascular disease

CYP1A2 cytochrome-P450 isoenzyme family 1, subfamily A, polypeptide 2 gene

CYP2D6 cytochrome-P450 isoenzyme family 2, subfamily D, polypeptide 6 gene

D’ Normalized linkage disequilibrium constant between two loci

DAAO d-amino acid oxidase gene

DAAO d-amino acid oxidase enzyme; DAO

DAOA d-amino acid oxidase activator gene

DAOA d-amino acid oxidase activator protein; G72

dbSNP National Centre for Biotechnology Information short genetic information database

DISC1 disrupted in schizophrenia 1 gene

Taylor DL

xi""

DLPFC dorsolateral prefrontal cortex

DNA deoxyribonucleic acid

DNAse-seq DNAse I hypersensitivity site sequencing

DRD1 dopamine D1 receptor gene

DRD2/D2 dopamine D2 receptor

DRD2 dopamine D2 receptor gene

DRD3 dopamine D3 receptor gene

D-ser D-serine

DSM-III-R Diagnostic and Statistical Manual, 3rd Edition Revised

DSM-IV Diagnostic and Statistical Manual, 4th Edition

DSM-5 Diagnostic and Statistical Manual, 5th Edition

DTNBP1 dystrobrevin binding protein 1 (dysbindin) gene

DTNBP1 dystrobrevin binding protein 1

DZ dizygotic

EAAT1 excitatory amino acid transporter 1

EAAT2/GLT1 excitatory amino acid transporter 2; glutamate transporter 1

EHM enhancer associated histone mark

emPCR emulsion polymerase chain reaction

ENCODE Encyclopedia of DNA Elements Project

EPS extrapyramidal symptoms

EPSCs excitatory postsynaptic currents

EPSPs excitatory postsynaptic potentials

eQTL expression quantitative trait loci

ESE/ESS exonic splicing enhancer/exonic splicing silencer

EUR European sample

Taylor DL

xii""

FDR false discovery rate

FuncPred National Institute for Environmental Health Sciences SNP Functional Prediction database

FWER family-wise error rate

G guanine nucleotide base

GABA γ-aminobutyric acid

GAD1 glutamate decarboxylase 1 gene

GAD1 glutamate decarboxylase 1 protein

GAIN Genetics Association Information Network

GATA1 GATA-binding factor 1; globin transcription factor 1; erythroid transcription factor 1

GATA4 GATA-binding factor 4; transcription factor GATA-4

GCS glycine cleavage system

GDH glutamate dehydrogenase

GERP genomic evolutionary rate profiling

GINA Genetic Information Non-discrimination Act (US)

Gln glutamine

GLS glutaminase; L-glutamine aminohydrolase

Glu glutamate

GLUR1 α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptor subunit 1 protein; GluA1

Gly glycine

GlyT1 glycine transporter 1

GRIA1 α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptor subunit 1 gene

GRIN1 N-methyl-D-aspartate receptor subunit 1 gene

GRIN2A N-methyl-D-aspartate receptor subunit 2A gene

Taylor DL

xiii""

GRIN2B N-methyl-D-aspartate receptor subunit 2B gene

GRM2 metabotropic glutamate receptor 2 gene

GRM3 metabotropic glutamate receptor 3 gene

GS glutamine synthetase

GWAS genome wide association study

H2O2 hydrogen peroxide

Haploreg Broad Institute Genomic Functional Annotation database

HRH1 histamine receptor H1 protein

HRH2 histamine receptor H2 gene

HTR1A 5-hydroxytryptamine (serotonin) receptor type 1A gene

HTR2A 5-hydroxytryptamine (serotonin) receptor type 2A gene

HTR2C 5-hydroxytryptamine (serotonin) receptor type 2C gene

HWE Hardy-Weinberg equilibrium

IP3 inositol 1,4,5-triphosphate

k independent number of tests conducted in a statistical analyses

K+ potassium ion

kb nucleotide kilobases

kDa kilodalton

kg kilogram

LD linkage disequilibrium

LGC Laboratory of the Government Chemist

LOD log of the likelihood odds ratio; confidence in D’

L-ser L-serine

LTP long term potentiation

Taylor DL

xiv""

LY2140023 pomaglumetad methionil; metabotropic glutamate receptor 2/3 agonist

MAF minor allele frequency

MAPK mitogen activated protein kinase

MC motif change

MC4R melanocortin receptor 4 gene

MDD major depressive disorder

MDR multifactor dimensionality reduction

METH-IP methamphetamine-induced psychosis

mg milligrams

Mg+ magnesium ion

mg/d milligrams per day

mGluR2 metabotropic glutamate receptor 2

mGluR3 metabotropic glutamate receptor 3

mGluR5 metabotropic glutamate receptor 5

MIM Mendelian inheritance in man gene code

min(s) minute(s)

miRNA micro ribonucleic acid

MK-801 (+)-D-aspartate 5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-iminedizocilpine (dizocilpine)

mL millilitres

Mn/Mj major/minor allele

mPFC medial prefrontal cortex

mRNA messenger ribonucleic acid

MZ monozygotic

n number; study sample size

Taylor DL

xv""

NA not available

Na+ sodium ion

Nacc nucleus accumbens

NB neuroblastoma cell line

NCBI US National Centre for Biotechnology Information

ng nanograms

ng/mL nanograms per millilitre

NGS next generation sequencing

NH3 ammonia

NHGRI US National Human Genome Research Institute

NIEHS National Institute of Environmental Health Sciences

NMDA N-methyl-D-aspartate

NMDAR N-methyl-D-aspartate receptor

NR1 N-methyl-D-aspartate receptor subunit 1

NR2A N-methyl-D-aspartate receptor subunit 2A

NR2B N-methyl-D-aspartate receptor subunit 2B

NRG1 neuregulin 1 gene

NRG1 neuregulin 1 protein

NXRN1 neurexin 1 gene

NXRN1 neurexin 1 protein

OCD obsessive compulsive disorder

OLZ olanzapine

OR odds ratio

PANSS Positive and Negative Syndrome Scale

PB protein bound

Taylor DL

xvi""

PCA principle components analysis

PCP phencyclidine

PCR polymerase chain reaction

PFC prefrontal cortex

PGC Psychiatric Genomics Consortium

PGM personal genome machine

PGx pharmacogenetics

PHM promoter-associated histone mark

PKA protein kinase A

PKC protein kinase C

pos position

PPI pre-pulse inhibition

PSD postsynaptic density

PSD-95 postsynaptic density protein 95

PubMed United States National Library of Medicine database

p-value probability value

QC quality control

QLS quality of life score

q-value false discovery rate adjusted p-value

r2 correlation coefficient between loci

RegulomeDB Stanford University Centre for Genomics and Personalized Medicine SNP annotation database

RG1678 bitopertin; glycine transporter 1 inhibitor

RNA ribonucleic acid

R/NR responder/non-responder

Taylor DL

xvii""

RR relative risk; risk ratio

rs# reference single nucleotide polymorphism identification number

s seconds

SA schizoaffective

SCZ schizophrenia

SD standard deviation

SLC1A2 solute carrier family 1, member 2 gene; excitatory amino acid transporter 2 gene; glutamate transporter 1 gene

SLC6A4 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 gene; serotonin transporter 1 gene

SLC6A9 solute carrier family 6, member 9 gene; glycine transporter 1 gene

SMR standardized mortality ratio

SNARE soluble N-ethylmaleimide sensitive factor (NSF) attachment protein receptor

SNP single nucleotide polymorphism

SPSS Statistical Package for the Social Sciences

SRR serine racemase

Suc succinate

T thymine nucleotide base

TBX18 T-box transcription factor 18 protein

TCA tricarboxylic acid cycle ; Krebs cycle ; citric acid cycle

TD tardive dyskinesia

TF transcription factor

TFBS transcription factor binding site

TRD treatment-resistant depression

TRS treatment-resistant schizophrenia

Taylor DL

xviii""

UCSC University of California, Santa Cruz

URS ultra treatment-resistant schizophrenia

UTR untranslated region

VGLUT/vGluT vesicular glutamate transporter

VKORC1 vitamin K epoxide reductase complex subunit 1 gene

VTA ventral tegmental area

Taylor DL

xix""

LIST OF FIGURES

Figure 1.1 Schematic Representation of a Glutamatergic Synapse p.15

Figure 1.2 The Ionotropic N-methyl-D-aspartate Glutamate p.19 Receptor

Figure 1.3 Clozapine is Superior to Chlorpromazine for p.30 Treatment-Resistant Schizophrenia

Figure 1.4 Identifying Functional Promoter Variants in SLC6A9 p.41 Using ENCODE Data

Figure 2.1 Human GRIN2B Gene Diagram p.60

Figure 2.2 GRIN2B Linkage Disequilibrium Map p.62

Figure 2.3 Genotype Analysis of GRIN2B Variant rs1072388 and p.65 Clozapine Response

Figure 3.1 Glutamate System Gene Diagrams p.83

Figure 3.2 Haploreg Results for Functional Glutamate System p.85 Gene Variants

Figure 3.3 GRM2, SLC6A9 and SLC1A2 Linkage Disequilibrium p.86 Maps

Figure 3.4 Genotype Analysis of SLC6A9 rs16831558 with p.92 Clozapine Response

Taylor DL

xx"""

LIST OF TABLES

Table 1.1 Select Candidate Genes and Chromosomal Regions p.10 Associated with Schizophrenia

Table 1.2 Evidence Supporting Clozapine Interacts with the p.35 Glutamate System

Table 1.3 Genetic Associations of Glutamate System Genes and p.39 Clozapine Response

Table 2.1 GRIN2B Gene Variants Genotyped in the Sample p.61

Table 2.2 Study Sample Demographics and Clinical Information p.63

Table 2.3 Association Analysis of GRIN2B Variants and p.64 Response to Clozapine

Table 2.4 Haplotype Analysis of GRIN2B Variants and Response p.66 to Clozapine

Table 3.1 Glutamate System Gene Variants Genotyped in the p.87 Study Sample

Table 3.2 Study Sample Demographics and Clinical Information p.89

Table 3.3 Associations Among Glutamate System Gene Variants p.90 and Clozapine Response

Table 3.4 Haplotype Analysis of GRM2 Variants and Response p.93 to Clozapine

Table 3.5 Haplotype Analysis of SLC6A9 Variants and Response p.94 to Clozapine

"

"

"

"

"

"

Taylor DL I. INTRODUCTION !

1!

!

Chapter 1 !

INTRODUCTION

1.1 Schizophrenia Overview

The disease concept of schizophrenia (SCZ) first emerged in the middle of the 19th century as

psychiatrists within Europe began noting disorders of mental dysfunction that arose in early

adulthood and often progressed to chronic deterioration. Several early terms were used to

refer to patients presenting with these symptoms, including “démence précoce” meaning

precocious onset of cognitive decline in France (Morel, 1860), and “adolescent insanity” in

Scotland (Clouston, 1904). Borrowing from Morel’s term, German neurologist Emil

Kraepelin (1856-1926) termed this condition ‘dementia praecox’ in 1887 and characterized

the disorder as nine different “clinical forms” (Kraepelin, 1899). Approximately two decades

later, the Swiss psychiatrist Eugen Bleuer (1857-1939) coined the term ‘schizophrenia’ on

the 24th of April, 1908 (Bleuer, 1911). The term was derived from two Greek words, ‘schizo’

meaning to tear or to split, and ‘phrenia’ meaning the mind.

In the present day, SCZ is characterized as a chronic, lifelong neuropsychiatric disorder

accompanied by positive, negative and cognitive symptoms that are diagnosed according to

the Diagnostic and Statistical Manual of Mental Health Disorders 5th Edition (DSM-5)

(American Psychiatric Association, 2013). According to this recent version of the DSM, a

patient presenting with positive symptoms, or psychosis, exhibits psychotic behaviours that

are not present in healthy individuals. The hallmark positive symptoms of SCZ include

auditory hallucinations (hearing voices), delusions (fixed false beliefs), disorganized speech,

and grossly disorganized or catatonic behaviour. In contrast, the negative and cognitive

2!

!

!

!

symptoms of SCZ manifest as deficits in normal functioning and include characteristics such

as affective flattening, poverty of speech (alogia), lack of motivation (avolition) and the

inability to experience pleasure (anhedonia). Cognitive symptoms include deficits in

attention, working memory, reasoning and processing speed. Affective symptoms and social

interaction deficits are also considered appropriate symptom categories of SCZ. Together,

these symptoms collectively contribute to the debilitating social and occupational

dysfunction commonly observed in patients suffering from this disorder.

1.1.1 Epidemiology and Time Course

SCZ affects approximately 15 out of every 100,000 people each year (median incidence,

central 80% range: 7.7-43) and almost 1% of the world’s population will suffer from SCZ

sometime in their lifetime (median lifetime morbid risk: 7.2 per 1000 persons) (Jablensky et

al., 1992; McGrath et al., 2008). The onset of SCZ usually occurs during adolescence

between the ages of 18 and 35 years old (Hafner et al., 1994). Interestingly, the disease

manifests two to three years later in females than in males (Reicher et al., 1990) and may be

caused, in part, by the anti-dopaminergic effects of estrogen (Seeman & Lang, 1990).

The time course of SCZ typically follows three distinct phases: the prodrome, the acute or

active phase and the residual phase. SCZ prodrome is the period leading up to psychosis and

is marked by abnormal behaviours and psychological states relating to cognition, emotion,

perception, communication, motivation and sleep (Klosterkotter et al., 2001). Nonspecific

clinical symptoms such as depression, anxiety, social isolation and school and/or

occupational dysfunction may also present during this time (Larson et al., 2010). The acute

phase of SCZ, otherwise known as the ‘first episode’ of psychosis, occurs once the classic

positive symptoms of SCZ manifest. This period is often accompanied by cognitive and

3!

!

!

!

social decline (Hafner et al., 1999). Following the first episode, patients are commonly

hospitalized, given a psychological assessment and diagnosed according to their symptoms.

Patients often begin a trial with antipsychotic (AP) medication, which is the mainstay in SCZ

treatment. Once positive symptoms are brought under control through the use of APs, the

residual phase begins. Unfortunately, this phase is often associated with a number of adverse

drug side effects and the presence of persistent negative and cognitive symptoms.

1.1.2 Deficits in Quality of Life

Individuals suffering from SCZ live shorter, lower quality lives in comparison to members of

the general population (standardized mortality ratio (SMR): 2.6, central 80% range: 1.2-5.8)

(McGrath et al., 2008). In addition, SCZ patients are often at higher risk of suicide (SMR:

12.9) (Saha et al., 2007; Zai et al., 2012). Indeed, approximately 5% of patients commit

suicide in their lifetime, usually near disease onset (Palmer et al., 2005). Cardiovascular

disease (CVD) is also more common in patients with SCZ and is primarily attributable to

unhealthy diet, lack of exercise, excessive smoking and co-morbid substance abuse (Laursen,

Munk-Olsen & Vestergaard, 2012). Compared to the general population, SCZ patients are

35% more likely to smoke cigarettes and 20% more likely to abuse alcohol and other

substances. A subset of patients who are diagnosed with treatment-resistant schizophrenia

(TRS) exhibit even higher rates of these health risk behaviours. In addition, patients often

suffer from motor side effects collectively called extrapyramidal symptoms (EPS), which

include tardive dyskinesia (TD) (30%), akathisia (36%) and parkinsonism (37%) (reviewed

by Kennedy et al., 2014).

Health utility scales are useful for assessing the effect of disease status on a patient’s quality

of life. On a scale from 0 to 1, zero representing death and one representing perfect health,

4!

!

!

!

severe, moderate and mild forms of SCZ have been assigned health utility scores of 0.56

(standard deviation (SD): 0.11), 0.62 (SD: 0.22) and 0.79 (SD: 0.09), respectively (Centre for

the Evaluation of Value and Risk in Health, 2011). TRS is also associated with poorer

quality of life that is approximately 20% lower than patients with SCZ who achieve

remission (0.61 vs. 0.75) (Kennedy et al., 2014). Alleviation of SCZ symptomatology

through successful therapeutic intervention usually increases a patient’s quality of life and

stresses the need for optimizing treatment outcomes (Bobes et al., 2007).

1.1.3 Burden to Society, Patients and Caregivers

The total financial burden of SCZ in Canada in 2004 was estimated to be $6.85 billion CDN,

accounting for direct healthcare and non-healthcare costs, as well as productivity lost due to

unemployment (Goeree et al., 2005; Kennedy et al., 2014). Furthermore, TRS patients report

annual healthcare costs ranging 3-to-11 times higher than schizophrenia patients who

respond to treatment ($66,360-$163,795 vs. $15,500-$22,300 per year). In addition, SCZ is

ranked as one of the top 10 causes of disability among people in developed countries

(Murray & Lopez, 1996; Barbato, 1998). Areas that are most prone to disability include

social functioning in relation to self-care, occupational performance, family relationships and

relationships in society as a whole (Janca et al., 1996). Stigma against individuals with

mental health disorders is lower than in the past, however, individuals with SCZ still

experience high degrees of rejection, discrimination and social isolation that ultimately

affects their sense of self and well-being (Goffman, 1963). Schizophrenia also represents a

substantial burden to caregivers (Schene et al., 1998; Gutierrez-Maldonado et al., 2005;

Vasudeva et al., 2013), with approximately 25% of patients living with a relative after

diagnosis (Torrey, 2013).

5!

!

!

!

1.2 Etiology and Pathophysiology

The cause of SCZ is not known, however, various hypotheses have been posited to explain

why this disorder manifests in particular individuals. These hypotheses propose SCZ

emerges due to a combination of risk factors that include environmental exposures and

genetic predisposition that ultimately lead to abnormalities in brain structure and function. A

brief overview of the etiology and pathophysiology of SCZ is presented below.

1.2.1 Environmental Risk Factors

Several environmental risk factors are thought to increase the likelihood of developing SCZ.

These factors include: male sex (relative risk (RR): 1.42, 95% Confidence Interval (CI):

1.30-1.56) (Aleman et al., 2003), first- and second-generation migrant status (RR: 2.7, 95%

CI: 2.3-3.2 and 4.5, 95% CI: 1.5-13.1, respectively) (McGrath et al., 2004; Cantor-Graae &

Selten, 2005), urban residence (RR: 2.4, 95% CI: 2.13-2.70) (Marcelis et al., 1998;

Mortensen et al., 1999), advanced paternal age (Malaspina et al., 2001; El-Saadi et al., 2004;

Sipos et al., 2004) and cannabis use (Boydell et al., 2007; De Sousa et al., 2013). Pre- and

postnatal exposure to stress and infection (Buka et al., 2001; Brown et al., 2004; Mortensen

et al., 2007), as well as prenatal nutritional deprivation (Susser & Lin, 1992; St Clair et al.,

2005) may also increase one’s risk for developing SCZ.

1.2.2 Genetic Predisposition

SCZ is also caused in part by genetic factors as evidenced by twin, family and adoption

studies. For instance, studies analyzing the incidence of SCZ within families indicate that

children are at an increased risk for developing SCZ if other family members are affected.

This risk increases as the degree of relation to the affected family member increases. As was

6!

!

!

!

mentioned earlier in Section 1.1, the risk of developing SCZ in the general population is

approximately 1%, however this risk jumps to 6% for a child born to an afflicted parent and

even higher to 9% for a person who has an afflicted sibling (Gottesman, 1991).

Twin studies have been particularly useful in understanding the contribution of genes to SCZ

etiology because twins share varying degrees of genetic similarity (reviewed by Cardno &

Gottesman, 2000). More specifically, dizygotic twins share exactly half their genes while

monozygotic twins, or identical twins, have genotypes that are exact duplicates. Twin studies

in SCZ research have yielded proband-wise concordance rates of 41-65% in monozygotic

twins and 0-28% in dizygotic pairs (reviewed by Gottesman, 1991). The amount of

concordance observed between twins as well as observed and expected resemblance between

relatives is useful for estimating trait heritability (Visscher et al., 2008). Researchers

involved in twin and family studies estimate the heritability of SCZ to be approximately 80-

85% (reviewed by Kendler, 1983). Thus, genetic factors are the largest known risk factor for

SCZ.

Adoption studies are another important tool for distinguishing the influence of heredity and

environment on human traits. Generally, traits shared between biological parents and a child

that is given up for adoption is explained by genetic factors, while traits shared

between adoptive or ‘environmental’ parents and an adopted child is attributed to

environmental influence (Plomen et al., 1997). The first adoption study in SCZ was proposed

in 1959 (Kety, 1959) and investigated whether adopted away children of parents (usually

mothers) with SCZ were at increased risk for developing the disorder. Adoption studies that

followed found that children who are raised in foster homes or institutions and whose

biological parent had SCZ developed SCZ more often than children of controls (Heston,

7!

!

!

!

1966). Similar studies have replicated these findings and provide sufficient evidence to

support a role of genetics in SCZ (Higgins, 1966; Rosenthal et al., 1968; Higgins, 1976;

Tienari & Wynne, 1994).

Molecular genetic studies aim to identify genes involved in disease etiology. Genetic linkage

studies follow the segregation of marker alleles from an affected lineage to offspring in

families (Riley & McGuffin, 2000), while genetic association studies assess the contribution

of loci to disease presentation in populations. Results from linkage studies in SCZ have been

mixed, with limited replication of findings. Positive results are generally accepted as

significant if they replicate in several additional studies. To date, SCZ has been linked to

numerous chromosomal regions including chromosome 1, 2, 4, 5-10, 13, 15, 18, 22 and the

X chromosome. The breadth of these findings exceeds the scope of this paper and have been

reviewed elsewhere (Riley & McGuffin, 2000; Riley & Kendler, 2006). Select candidate

genes and chromosomal regions are summarized in Table 1.1.

1.2.2.1 Chromosome 6: 6p24-p22

Linkage findings for chromosome 6 first emerged from studies involving Irish families with

a high density of broad-spectrum psychiatric disorders (Straub et al., 2002). One particular

gene within this region of chromosome 6, the dystrobrevin binding protein 1/dysbindin gene

(DTNBP1) has been, in general, positively associated with SCZ (Funke et al., 2004;

Numakawa et al., 2004; Williams et al., 2004). Altered expression of DTNBP1 in certain

brain regions has also been reported in patients with SCZ (Weickert et al., 2004). Although

the exact function of DTNBP1 in the brain is unknown, this protein appears to have a role in

glutamatergic neurotransmission (Talbot et al., 2004; Shao et al., 2011).

8!

!

!

!

1.2.2.2 Chromosome 8: 8p22-p21

Findings from the Maryland family sample, as well as data from numerous pedigrees of

individuals with differing ethnic background have provided linkage support for chromosome

8 (Pulver et al., 1995). Association findings have confirmed that a candidate gene located

within this region called neuregulin 1 (NRG1) is associated with increased risk for SCZ

(Stefansson et al., 2003; Williams et al., 2003; Corvin et al., 2004). The neuregulin 1 protein

(NRG1) is a CNS-expressed molecule involved in the expression and activation of

neurotransmitter receptors (reviewed by Harrison & Law, 2006). Of interest to the

glutamatergic neurotransmitter system, NRG1 regulates N-methyl-D-aspartate glutamate

receptor (NMDAR) function via a postsynaptic density protein (PSD-95) (Gu et al., 2005).

1.2.2.3 Chromosome 11: 11q22-q21

The chromosome 11 region was first implicated in SCZ etiology in a linkage study involving

two Japanese pedigrees (Nanko et al., 1992), and was later confirmed in a separate study

involving a large Canadian pedigree (Maziade et al., 1995; Gurling et al., 2001). One

particular gene of interest lying within chromosome region 11q22 is the dopamine receptor

D2 gene (DRD2). This gene is of particular interest because all antipsychotic medications on

the market for the treatment of SCZ symptoms have blockade of the dopamine D2 receptor

(DRD2) as a common mechanism of action (Seeman & Lee, 1975). A large meta-analysis of

variants within DRD2 indicates this gene is associated with SCZ (Glatt et al., 2003).

1.2.2.4 Chromosome 13: 13q14-q32

Data from a mixed sample of UK and Japanese families initially suggested linkage to

chromosome 13 (Lin et al., 1995). This region is of particular interest because approximately

120 known genes of interest are located here including the serotonin receptor type 2A gene

9!

!

!

!

(HTR2A), D-amino acid oxidase activator gene (DAOA) and the d-amino acid oxidase gene

(DAAO). Association studies involving HTR2A variants in SCZ risk have produced mixed

results (reviewed by Serretti et al., 2007), while studies assessing association with response

to atypical APs have yielded more consistently positive findings (Arranz et al., 1995; Burnet

& Harrison, 1995; Masellis et al., 1995; Nothen et al., 1995). Based on meta-analysis, DAOA

has been implicated in SCZ risk (Detera-Wadleigh & McMahon, 2006), as well as DAAO

(Chumakov et al., 2002; Schumacher et al., 2004; Yang et al., 2013). Functionally, the d-

amino acid oxidase activator protein (DAOA) activates d-amino acid oxidase enzyme

(DAAO/DAO) that is responsible for the oxidation of D-amino acids such as D-serine in the

brain. Because D-serine acts as a co-agonist at the NMDAR, variable activity of DAOA

and/or DAAO may therefore contribute to SCZ pathophysiology by affecting glutamatergic

neurotransmission.

1.2.2.5 Polygenic Inheritance

Recently, a very large genome wide association study (GWAS) consisting of up to 36,989

cases and 113,075 controls from the psychiatric genomics consortium (PGC) identified 128

independent associations spanning 108 conservatively defined loci with SCZ (Schizophrenia

Working Group of the Psychiatric Genomics Consortium, 2014). The top 128 loci included

DRD2 and many other genes related to glutamatergic function such as the metabotropic

glutamate receptor 3 gene (GRM3), the NMDAR subunit 2A gene (GRIN2A) and the α-

amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptor (AMPAR) subunit 1 gene

(GRIA1). As implicated by this recent GWAS, SCZ is considered a complex trait that almost

certainly follows a polygenic mode of inheritance (Gottesman & Shields, 1967; McClellan et

al., 2007; International Schizophrenia Consortium et al., 2009). As opposed to a Mendelian

10!

!

!

!

mode of inheritance where a single gene is responsible for disease presentation, multiple

genes and the interaction among these genes likely contribute to the development of SCZ.

Table&1.1&Select&Candidate&Genes&&&Chromosomal&Regions&Associated&with&Schizophrenia&

Gene!Region! Linkage!&!Candidate!Gene!Association!Findings! References!

1q32A42! Balanced! 1:11! translocation! identified! in! a! large! Scottish!

pedigree!with!a!chromosomal!breakpoint!at!1q42.1!

Disrupted& in& schizophrenia& 1& gene& (DISC1):!Plays!a! role! in!cytoskeletal! regulation!and!may!affect!neuronal!migration,!

neurite!outgrowth!and!intracellular!transport!

(St!Clair!et!al.,!1990;!Shaw!

et!al.,!1998;!Hovatta!et!al.,!

1999;!Brzustowicz!et!al.,!

2000;!Miyoshi!et!al.,!2003;!

Ozeki!et!al.,!2003;!

Sumiyoshi!et!al.,!2004)!

6p24Ap22! Linkage! findings! identified! in! Irish! families! with! a! high!

density!of!broad!spectrum!psychiatric!disorders!!

Dystrobrevin&binding&protein&1/dysbindin&gene&(DTNBP1):!Altered!expression! in!SCZ!brains;!appears!to!have!a!role! in!

glutamatergic!neurotransmission!!

(Straub!et!al.,!2002;!Funke!

et!al.,!2004;!Numakawa!et!

al.,!2004;!Talbot!et!al.,!

2004;!Williams!et!al.,!

2004;!Shao!et!al.,!2011)!

8p22Ap21! Genetic!data! from!the!Maryland! sample!and! several!other!

pedigrees! of! different! ethnic! origin! provide! evidence! to!

support!linkage!of!this!region!to!SCZ!

Neuregulin& 1& gene& (NRG1):! Associated!with! increased! risk!for! SCZ;! regulates! NMDAR! function! through! postsynaptic!

density!protein!

(Pulver!et!al.,!1995;!

Stefansson!et!al.,!2003;!

Williams!et!al.,!2003;!

Corvin!et!al.,!2004;!Gu!et!

al.,!2005)!

11q22Aq21! This!region!was!first! implicated!in!a! linkage!study!involving!

two!Japanese!pedigrees!and!was!later!confirmed!in!a!large!

Canadian!pedigree!

Dopamine&Receptor&D2&gene&(DRD2):!Primary!target!of!AP!

medications;!metaAanalyses! indicate! this! gene! is! positively!

associated!with!SCZ!

(Nanko!et!al.,!1992;!

Maziade!et!al.,!1995;!

Gurling!et!al.,!2001;!Glatt!

et!al.,!2003;!Schizophrenia!

Working!Group!of!the!

Psychiatric!Genomics!

Consortium,!2014)!

13q14Aq32! Data! from! a! mixed! sample! of! UK! and! Japanese! families!

initially!suggested!linkage!to!this!region!

Serotonin& receptor&2A&gene& (HTR2A):!Mixed!results! in!SCZ!

etiology;!positive!results!for!association!with!AP!response!

DFamino&acid&oxidase&activator&gene& (DAOA):!Activates!DAamino!acid!oxidase!protein;!positively!associated!with!SCZ!!

DFamino&acid&oxidase&gene&(DAAO/DAO):!Oxidizes!DAamino!

acids! such! as! NMDAR! coAagonist! DAserine;! positively!

associated!with!SCZ!

(Arranz!et!al.,!1995;!

Burnet!&!Harrison,!1995;!

Lin!et!al.,!1995;!Masellis!et!

al.,!1995;!Nothen!et!al.,!

1995;!Chumakov!et!al.,!

2002;!Schumacher!et!al.,!

2004;!DeteraAWadleigh!&!

McMahon,!2006;!Serretti!

et!al.,!2007;!Yang!et!al.,!

2013)!

11!

!

!

!

1.2.3 Brain Structural Abnormalities

Several brain structural abnormalities have been observed in patients with SCZ (reviewed by

Buckley, 2005). In brief, these structural abnormalities include: reduced cortical thickness

(Voineskos et al., 2013), gradual lateral and third ventricle enlargement (Johnstone et al.,

1976; Mathalon et al., 2001; Kasai et al., 2003), subtle decreases in thalamic volume

(Andreasen et al., 1994; Kemether et al., 2003), an enlarged caudate nucleus (Jernigan et al.,

1991), smaller temporal lobes (Wright et al., 2000; Csernansky et al., 2002) and reduced left-

right asymmetry (anisotropy) (Nakamura et al., 2012; Lee et al., 2013). Brain structural

abnormalities in patients with SCZ have been difficult to reproduce across studies because

they are relatively subtle and are also influenced by patient characteristics such as age,

gender, chronicity of illness, medication use and co-morbid substance abuse (McCarley et

al., 1999; Arango et al., 2003).

1.2.4 Neurochemical Abnormalities

In addition to brain structural aberrations, several neurochemical abnormalities have been

observed in patients with SCZ. The majority of these abnormalities relate primarily to the

dopamine, glutamate, serotonin and the inhibitory neurotransmitter γ-aminobutyric acid

(GABA) neurotransmitter systems. The roles of serotonin and GABA in SCZ etiology are

beyond the scope of this text and have been reviewed elsewhere (Meltzer, 1995a; Aghajanian

& Marek, 2000; Wassaf & Kochan, 2003; Nakazawa et al., 2012).

1.2.4.1 The Dopamine Hypothesis

Much of SCZ research has focused on perturbations in the dopamine neurotransmitter

system. The groundwork for the ‘dopamine hypothesis of SCZ’ was conducted by Arvid

Carlsson (1923-) in the late 1950s, who won the Nobel prize in 2000 for his contribution, and

12!

!

!

!

by Jacques van Rosum in the late 1960s (reviewed by Seeman, 1987). Furthermore, Seeman

& colleagues showed in 1975 that the efficacy of AP drugs was tightly correlated with

affinity of the drugs for DRD2 receptors (Seeman & Lee, 1975). Hence, the dopamine

hypothesis was based largely on observations that dopamine-enhancing drugs, such as

amphetamines, were capable of reproducing positive symptoms observed in patients with

SCZ (Lieberman et al., 1987), and that AP drugs which treat psychosis do so primarily by

blocking DRD2 receptors (Carlsson & Lindqvist, 1963; Seeman & Lee, 1975).

Biological data has also provided evidence to support the dopamine hypothesis of SCZ. A

large proportion of the dopaminergic neurons in the human brain originate in the ventral

tegmental area (VTA), and project to the prefrontal cortex (mesocortical pathway) and to

subcortical limbic brain regions (mesolimbic pathway). Dopamine activity in prefrontal

regions appears to play an inhibitory role on dopamine release in subcortical areas (Pycock,

Kerwin & Carter, 1980; Kolachana, Saunders & Weinberger, 1995; Karreman &

Moghaddam, 1996). From these findings, SCZ has been characterized as a disorder caused

by deficits in dopamine transmission (hypodopaminergia) in the prefrontal cortex,

consequently leading to over-activity of dopamine circuits (hyperdopaminergia) in the

striatum (reviewed by Davis et al., 1991). Hypofrontality in SCZ patients is evidenced by

reduced cerebral blood flow in the frontal cortex and may be related to low dopaminergic

activity in that region (Davis et al., 1991; Rubin et al., 1991; Andreasen et al., 1992). Positive

symptoms are thought to arise due to striatal hyperdopaminergia, while dopamine

hypoactivity in the prefrontal cortex is proposed to account for negative and cognitive

symptoms.

13!

!

!

!

1.2.4.2 The Glutamate Hypothesis

Despite the usefulness of the dopamine hypothesis, cortical abnormalities observed in SCZ

appear to be more complex than the originally proposed hypofrontality. Some posit that the

dopamine system of SCZ patients may in fact be normal, yet suffer from abnormal regulation

(Grace, 2012), and hints towards the involvement of several neurotransmitter systems in

SCZ’s etiology. For instance, a large amount of evidence has characterized SCZ as a neuro-

developmental disorder accompanied by perturbations in glutamatergic neurotransmission

(Weinberger, 1987; Olney & Farber, 1995; Mohn et al., 1999; Olney et al., 1999). Much of

the rationale supporting this ‘glutamate hypothesis of SCZ’ stems from observations that

administration of NMDAR antagonists, ketamine and phencyclidine (PCP), induce SCZ-like

symptoms in healthy controls and exacerbate psychotic symptoms in SCZ patients (Luby et

al., 1959; Luisada, 1978; Lahti et al., 2001). Interestingly, glutamate action, specifically at

the NMDAR, has been shown to regulate dopaminergic neurotransmission (Zweifel et al.,

2008; Parker et al., 2010).

Several theories regarding glutamate’s involvement in SCZ etiology have been proposed.

One overarching theory suggests that NMDAR glutamatergic hypofunction is responsible for

creating the positive and negative symptoms of SCZ through dysregulation of mesolimbic

and mesocortical dopamine neurons, respectively. More specifically, hypofunction of the

inhibitory glutamatergic pathway that projects from cortical pyramidal neurons to dopamine

neurons in the VTA may cause tonically uninhibited dopamine release from the mesolimbic

pathways and result in positive symptoms, while hypofunction of the stimulatory

glutamatergic pathway that acts on mesocortical dopamine neurons could cause the

14!

!

!

!

hypodopaminergia that is responsible for negative, cognitive and affective symptoms (Stahl,

2007). The glutamate hypothesis of SCZ is discussed in more detail in the following section.!!

1.3 Glutamatergic Dysfunction in Schizophrenia

A great deal of evidence has characterized SCZ as a disorder related to aberrant glutamate

signalling. The following section provides a general overview of the glutamate system with

particular emphasis placed on receptors, transporters and enzymes involved in glutamatergic

neurotransmission. This section concludes with a thorough summary of literature on

glutamate abnormalities in SCZ.

1.3.1 The Glutamate System

Glutamate is the primary excitatory neurotransmitter in the mammalian central nervous

system (CNS) (Curtis et al., 1960). Glutamate neurotransmission plays an essential role in

physiological processes such as synaptic plasticity and induction of long term potentiation

(LTP), processes that are both implicated in memory and learning (Harris et al., 1984; Morris

et al., 1986). Various receptors, transporter and enzymes are involved in the proper

functioning of glutamatergic neurotransmission. There are two main groups of excitatory

glutamate receptors, the ligand gated ion channels which include the NMDAR, AMPAR and

kainate types of receptors, and the metabotropic G-protein coupled receptors (mGluRs)

which are subdivided into group I, group II and group III (Foster & Roberts, 1981; Watkins

& Evans, 1981; Watkins & Collingridge, 1994; Pin & Duvoisin, 1995). Extracellular levels

of glutamate and other relevant neurotransmitters such as glycine are carefully regulated by

the action of transporters. Once taken up into neurons and/or surrounding microglia,

glutamate and glycine are either converted to other chemical compounds or metabolized

15!

!

!

!

through the action of several intracellular enzymes. A schematic representation of a neuronal

synapse is shown in Figure 1.1 and highlights the role of a number of key receptors,

transporters and enzymes relevant to the glutamate system. This figure also highlights

GABAergic neurons (top right corner), which are highly important in brain function due to

their general inhibitory role. It should also be mentioned that GABA and glutamate are in a

complex interplay, however this thesis largely focused on investigating the role of glutamate

system abnormalities. Therefore, please note that this diagram is an oversimplified

representation of a synapse, for example, in that the role of GABAergic neurons is not fully

depicted.

&

16!

!

!

!

Figure&1.1&Schematic&Representation&of&a&Glutamatergic&Synapse&

Glutamate!(Glu)!is!the!major!excitatory!neurotransmitter!in!the!mammalian!central!nervous!

system! with! roles! in! cognition,! memory! and! learning.! Precise! regulation! of! glutamate!

signaling! must! be! maintained! as! excitotoxicity! can! result! from! excessive! glutamatergic!

stimulation.! (1)! Glutamate! is! derived! from! glutamine! (Gln)! through! the! action! of! the!

enzyme!glutaminase! (GLS)!or! from!the!tricarboxylic!acid! (TCA)!cycle,!and! is!packaged! into!

presynaptic!vesicles!by!vesicular!glutamate!transporters!(vGluTs).!(2)!Vesicles!release!their!

contents! into! the! synaptic! cleft! through! vesicleAmembrane! fusion,! which! is! facilitated! by!

SNARE!proteins.!(3)!Presynaptic!and!postsynaptic!glutamate!binding!activates!metabotropic!

(mGlu)! and! ionotropic! (NMDA,! AMPA,! Kainate)! glutamate! receptors,! downstream! signal!

transduction!cascades!and!synaptic!plasticity.!(4)!Glutamate!is!recycled!from!the!synapse!by!

glutamate! transporters! (EAAT1/2),! which! are! expressed! predominantly! on! astroglia! (glial!

cells).! Once! inside! astroglia,! glutamate! is! converted! to! glutamine! through! glutamine!

synthetase!(GS)!and!is!then!exported!for!reAuptake!by!glutamatergic!neurons.!(5)!Glutamine!

may! also! be! taken! up! by!GABAergic! interneurons,! converted! first! into! glutamate! by!GLS,!

and!then!to!GABA!by!glutamate!decarboxylase!1!(GAD1).!Glycine!(Gly)!acts! independently!

as!an!inhibitory!neurotransmitter,!or!in!concert!with!glutamate!neurotransmission!as!a!coA

agonist! at! the! NMDAR.! (6)! Glycine! is! recycled! from! the! synapse! by! glycine! transporters!

(GlyT1)!which!are!located!primarily!on!astrocytes.!Once!taken!up!by!astrocytes,!glycine!can!

be!metabolized!to!αAketoglutaric!acid!(αAketo!acid),!ammonia!(NH3)!and!hydrogen!peroxide!

(H2O2)! through! a! series! of! intermediate! steps:! glycine! to! LAserine! (LAser)! catalyzed!by! the!

glycine! cleavage! system! (GCS),! LAserine! to! DAserine! (DAser)! catalyzed! by! serine! racemase!

(SRR),!DAserine!to!the!final!products!via!the!DAamino!acid!oxidase!enzyme,!that!is!activated!

by!DAamino!acid!oxidase!activator!protein!(G72).!Adapted!from!Pharmacology+Biochemistry+

and+ Behavior,! 100(4).!Mark! J.!Niciu,! Benjamin! Kelmendi!&!Gerard! Sanacora.!Overview!of!

Glutamatergic! Neurotransmission! in! the! Nervous! System,! 656A64.! Copyright! (2012)! with!

permission!from!Elsevier.!

!

17!

!

!

!

1.3.1.1 Ionotropic Glutamate Receptors

NMDAR and AMPAR are principally expressed on dendritic spines of postsynaptic neurons

(Kharazia et al., 1996) where they function in signaling during induction of synaptic

plasticity and long-term modulation of synaptic strength (Asztely & Gustafsson, 1996;

Shapiro, 2001). Kainate receptors mostly localize to postsynaptic membranes where they

function to modulate synaptic response to glutamate (Van den Pol, 1994; Haak, 1999).

Kainate receptors also localize to surrounding glia and presynaptic neurons where they

modulate a variety of cellular functions including glutamate reuptake and release, and

presynaptic excitability (Cartmell & Schoepp, 2000; Huettner, 2003; Grueter & Winder,

2005; Rodriguez-Moreno, 2006).

The NMDAR is a hetero-tetramer composed of two obligatory GluN1 (NR1) subunits and

either two GluN2 (NR2A-D) or two GluN3 (NR3A-B) subunits (Monyer et al., 1992)

(Figure 1.2). Subunit composition varies during development and results in various receptor

isoforms with different physiological and pharmacological properties (McBain & Mayer,

1994). The NMDAR is implicated in additional processes such as neuronal migration

(Komuro & Rakic, 1993) and differentiation (Pearce et al., 1987), response to trophic factors

(Black, 1999), dendritic spine development (Tian et al., 2007), LTP, and memory and

learning (Massey et al., 2004). Given the importance of the NMDAR in neuronal processes,

abnormal function and/or development of this receptor system could lead to the deficits in

learning and memory that are observed in patients with SCZ.

NMDAR channel activation utilizes a coincidence detection type mechanism that requires

simultaneous binding of glutamate to the NR2 subunit (Laube et al., 1997; Anson et al.,

1998), binding of an endogenous co-agonist, either glycine or D-serine, to the NR1 subunit

18!

!

!

!

(Kuryatov et al., 1994; Hirai et al., 1996) and removal of a voltage-dependent block by

external magnesium (Mg2+) (Mayer et al., 1984; Nowak et al., 1984). Channel activation

produces excitatory postsynaptic potentials (EPSPs) and increases calcium (Ca2+) and

sodium (Na+) concentration inside the cell (Schiller et al., 1998). Intracellularly, calcium acts

as a secondary messenger with downstream effects on protein kinases such as mitogen

activated protein kinase (MAPK) (Xia et al., 1996) and transcription factors such as cyclic

adenosine monophosphate (cAMP) response element binding protein (CREB) (Sala et al.,

2000). NMDAR action is also associated with effector kinases such as protein kinase C

(PKC) (Zheng et al., 1997), protein kinase A (PKA) (Tingley et al., 1997) and

calcium/calmodulin-dependent kinase II (CaMKII) (Omkumar et al., 1996).

The remaining two ionotropic glutamate receptors, AMPA and kainate, both form tetrameric

complexes consisting of GluR1-4 subunits (AMPA) or GluR5-7 and KA1-2 subunits

(kainate). Both receptors are widely expressed in the mammalian CNS and mediate fast

excitatory neurotransmission in response to the binding of glutamate (Khakh & Henderson,

2000). The function of AMPA and kainate ionotropic glutamate receptors is controlled in

part by RNA editing of subunits (Higuchi et al., 1993), as well as alternative splicing of

messenger RNA (mRNA) transcripts (Sommer et al., 1990). Ionotropic glutamate receptor

function, the AMPAR in particular, is also regulated by receptor number, localization and

facilitation at synapses ultimately controlling such processes as LTP-like strengthening of

neocortical synapses after sensory stimulation (Takahashi et al., 2003), learning in the

hippocampus (Whitlock et al., 2006) and response to stress hormones (Groc et al., 2008;

Martin et al., 2009; Yuen et al., 2011).

19!

!

!

!

Figure&1.2!The&Ionotropic&NFmethylFDFaspartate&Glutamate&Receptor!

Functional!NMDARs!are!constructed!from!assemblies!of!four!subunits!(two!NR1!and!either!

two! NR2AAD! or! NR3AAB).! Subunit! composition! varies! and! results! in! various! receptor!

isoforms! that!have!different!physiological! and!pharmacological!properties.!NR1! subunit! is!

encoded!by!eight!alternatively!spliced!variants!of!GRIN1,!and!contains!the!binding!site!for!

endogenous! coAagonists! glycine! and! DAserine.! NR2! subunits! contain! the! glutamate!

recognition! site! and! binding! sites! for! various! other! antagonists.! The! NMDAR! channel! is!

normally! blocked! at! resting! membrane! potential! by! magnesium! (Mg+)! in! a! voltageA

dependent! manner! and! is! activated! following! depolarization! of! the! postsynaptic!

membrane.! Receptor! activation! occurs! through! a! coincidence! detection! type!mechanism!

and!requires!both!glutamate!and!glycine!binding.!Receptor!activation!leads!to!calcium!(Ca2+)!

and!sodium! (Na+)! influx,!and!potassium! (K

+)!efflux,! facilitating! longAterm!potentiation!and!

synaptic! plasticity.! Antagonists! of! the! NMDAR! include:! Memantine! (treats! Alzheimer’s!

disease),! Selfotel! (discontinued! anxiolytic/anticonvulsant),!MRZ! 2/576! (research! use)! and!

Ifenprodil! (discriminates!NMDAR!subpopulations).!Adapted!from!Pharmacological+Reviews!

50.!Wojciech! Danysz! and! Chris! G.! Parsons.! Glycine! and! NAMethylADAAspartate! Receptors:!

Physiological!Significance!and!Possible!Therapeutic!Applications,!597A664.!Copyright!(1998)!

with!permission!from!American!Society!for!Pharmacology!and!Experimental!Therapeutics.!!!

20!

!

!

!

1.3.1.2 Metabotropic Glutamate Receptors

Like the kainate receptors, mGluRs modulate synaptic responses to glutamate at postsynaptic

membranes (Van den Pol, 1994; Haak, 1999) and modulate a variety of cellular functions at

presynaptic neurons (Cartmell & Schoepp, 2000; Huettner, 2003; Grueter & Winder, 2005;

Rodriguez-Moreno, 2006). The metabotropic glutamate receptors mediate signaling through

7-transmembrane region G-protein coupled receptors (Pin et al., 2003). The three subclasses

are groups I, II and III and they signal through different mechanisms. Group I (mGluR1, 5)

signals through G-protein activation that is coupled to protein kinase activation (ie PLC), and

subsequently leads to the formation of inositol 1,4,5-triphosphate (IP3) and intracellular

release of calcium (Hermans & Challiss, 2001). In contrast, group II (mGluR2, 3) and group

III (mGluR4, 6, 7, 8) receptors couple to inhibitory G-protein signaling whereby inhibition of

adenylyl cyclase activity subsequently leads to reduction in cAMP production (Conn & Pin,

1997). The mGluR receptors are tightly regulated by formation of various alternatively

spliced isoforms which control several aspects of receptor function, including surface

receptor expression, G-protein coupling, and interaction with scaffolding proteins in the

postsynaptic density (PSD) (Stowell & Craig, 1999; Tu et al., 1999; Pin et al., 2003).

Drugs targeting metabotropic glutamate receptors have been investigated for the treatment of

SCZ (reviewed by Li et al., 2015). LY404039, a specific and potent mGluR2/3 agonist

developed by Eli Lilly & Co, is one example of a drug from this class (Monn et al., 2005).

Experiments with mGluR2/3 knockout mice suggest that the antipsychotic-like behavourial

effects of LY404039 are caused by mGluR2 activation and further implicate the glutamate

system in SCZ etiology (Rorick-Kehn et al., 2007; Fell et al., 2008). Clinical studies

investigating this compound for the treatment of SCZ indicate it was well tolerated and

21!

!

!

!

lacked the adverse side effects typically associated with dopaminergic agents such as weight

gain, EPS and hyperprolactinemia (Adams et al., 2013). Unfortunately, efficacy results for

this compound were inconsistent. More specifically, the initial proof of concept study

showed LY404039 and the active control olanzapine (OLZ) significantly decreased patient

symptoms compared to placebo (Patil et al., 2007), however a Phase 2 (inpatient, dose-

ranging study) reported ‘inconclusive results’ as both LY2140023 and OLZ failed to separate

from placebo (Kinon et al., 2011). Finally, a Phase 2 (acute, fix-dosed study) showed that

LY404039 did not separate from placebo in the primary efficacy endpoint unlike the active

control risperidone (Kinon et al., 2013). Development of this drug was discontinued

following the last study. Further research into LY2140023 by Eli Lilly has indicated that

response to this compound may be more favourable in a subgroup of patients that carry a

select set of 23 single nucleotide polymorphism (SNP) gene variants (Liu et al.,

2012). Interestingly, 16 of these 23 SNPs are located in the HTR2A gene. Studying the

relationship between genetic variation and drug response is called pharmacogenetics and will

be discussed in more detail in Section 1.5.

1.3.1.3 Amino Acid Transporters

Amino acid transporters control the reuptake of amino acids, such as glutamate and glycine,

from the synapse into neurons and/or surrounding neuroglia. Two amino acid transporters are

involved in glutamatergic neurotransmission: the excitatory amino acid transporters

(EAATs) 1-5 and the glycine transporters (GlyTs) 1 and 2. The EAATs are responsible for

the rapid reuptake of synaptic glutamate into astrocytes and control extracellular

concentrations of glutamate (reviewed by O'Shea, 2002).! Strict control of extracellular

glutamate concentrations is necessary to prevent excitotoxicity and cell death (Coyle &

22!

!

!

!

Puttfarcken, 1993; Szatkowski & Attwell, 1994). Of the five EAATs, EAAT2 (otherwise

known as GLT1) is the chief glutamate transporter in the forebrain and is responsible for

more than 90% of glutamate reuptake into surrounding astrocytes (Bar-Peled et al., 1997;

Furuta et al., 1997).

The second type of transporter involved in glutamatergic neurotransmission is responsible

for controlling levels of the amino acid glycine. Within the nervous system, glycine carries

out a dual role as both an inhibitory neurotransmitter at glycine receptors and as a co-agonist

at the NR1 subunit of the NMDAR to mediate excitatory neurotransmission (Johnson &

Ascher, 1987). The primary neuronal glycine transporter, GlyT1, is expressed predominantly

on astrocytes and is responsible for tightly regulating glycine concentrations in the vicinity of

excitatory synapses through reuptake of glycine into neighbouring astroglia (Smith et al.,

1992; Zafra et al., 1995; Cubelos et al., 2005). Interestingly, changes in GlyT1 activity have

been shown to alter NMDAR-mediated neurotransmission as evidenced by increased

NMDAR excitatory postsynaptic currents (EPSCs) following GlyT1 antagonism (Bergeron

et al., 1998; Chen et al., 2003; Kinney et al., 2003) and by increased NMDAR function in

GlyT1 knockout mice (Gabernet et al., 2005).

Drugs targeting the glycine system have also been investigated for SCZ. One such drug

inhibits GlyT1 function in an attempt to ameliorate the NMDAR hypofunction associated

with SCZ etiology (reviewed by Hashimoto, 2010). One such glycine transporter inhibitor

bitopertin (RG1678), developed Roche, has been investigated as an adjunct for persistent

negative symptoms in SCZ. Five clinical trials have been conducted so far, however the

results have been non-significant (ClinicalTrails.gov identifiers: NCT01192867,

NCT01192906, NCT01192880, NCT01235520, and NCT01235559). Various clinical studies

23!

!

!

!

have also investigated the efficacy of adjunct glycine, D-serine or D-cycloserine (NMDAR

co-agonists) for the treatment of SCZ (reviewed by Javitt, 2004). On a more positive note,

the majority of these studies have reported large effect-size improvements in negative and

cognitive symptoms (Potkin et al., 1992; Heresco-Levy et al., 1996; Tsai et al., 1998; Goff et

al., 1999; Javitt et al., 2001).

1.3.1.4 Glutamate System Enzymes

There are several key enzymes involved in regulating glutamatergic neurotransmission.

These enzymes include glutaminase (GLS) and glutamine synthetase (GS), which are

responsible for interconverting glutamate and glutamine, glutamate decarboxylase 1

(GAD1), which converts glutamate to GABA, and various other enzymes involved in glycine

metabolism.

Glutamine synthetase is an astrocytic enzyme responsible for converting glutamate to

glutamine (Meister, 1974; Martinez-Hernandez et al., 1977). Glutamine is released

extracellularly as a neuroprotective form of glutamate where it can be taken up by neurons

and deaminated back into glutamate by the enzyme glutaminase (Goldstein, 1967; Curthoys

& Watford, 1995). This pattern of compartmentalization and interconversion constitutes what

is known as the ‘glutamate-glutamine cycle’ (Shank & Aprison, 1981). This cycle ensures

three events: 1) glutamate is rapidly removed from the synapse to prevent neurotoxicity; 2)

glutamate is converted to the ‘neuroprotective carrier’ glutamine; and 3) glutamine is made

available in neuronal compartments and allows for the regeneration of glutamate for

excitatory signaling (Daikhin & Yudkoff, 2000).

24!

!

!

!

Besides reuptake into glutamatergic neurons, glutamine can also be taken up by GABAergic

interneurons and metabolized to GABA by GAD1 (Erlander et al., 1991). GABA is an

inhibitory neurotransmitter that controls fundamental processes such as neurogenesis

(Fagiolini et al., 2004; Ge et al., 2006), movement, and tissue development (Nakatsu et al.,

1993). A mutation in the glutamate decarboxylase 1 gene (GAD1) in humans results in a

disorder known as spastic cerebral palsy (Lynex et al., 2004), while GAD1 knockout mice

have dramatically reduced GABA levels and die at birth due to a severe cleft palate (Asada et

al., 1997).

Lastly, a series of enzymes are responsible for the metabolism of glycine to α-ketoglutaric

acid (α-keto acid), ammonia (NH3) and hydrogen peroxide (H2O2). Upon entry into glial cells

through the action of GlyT1, glycine is first converted to L-serine (L-ser) by the glycine

cleavage system (GCS) (Kikuchi, 1973). L-serine is then converted to D-serine (D-ser) by

the enzyme serine racemase (SRR) (Wolosker et al., 1999), and then converted to the final

products α-keto acid, NH3 and H2O2 via D-amino acid oxidase (Dixon & Kleppe, 1965). Like

glycine, D-serine is an endogenous agonist at the NR1 subunit of the NMDAR and plays a

role in regulating excitatory neurotransmission (Matsui et al., 1995).

1.3.2 Glutamate System Abnormalities in Schizophrenia

Numerous glutamate system abnormalities have been reported in patients with SCZ giving

further credence to the glutamate hypofunction hypothesis. For instance, postmortem studies

show abnormalities in NMDAR subunit expression in patients with SCZ, including increased

NR2B transcript in the thalamus (Clinton & Meador-Woodruff, 2004)) and temporal lobes

(Grimwood et al., 1999), as well as higher NR1 expression in the dorsolateral prefrontal

cortex (DLPFC) (Dracheva et al., 2001), substantia nigra (Mueller et al., 2004) and superior

25!

!

!

!

temporal cortex (Nudmamud-Thanoi & Reynolds, 2004). In addition, more recent data has

indicated that unmedicated patients with SCZ have increased hippocampal glutamate in

comparison to healthy controls (Kraguljac et al., 2013).

Preclinical models also point to glutamate system abnormalities in modeling SCZ-related

behaviours. Rats administered PCP and ketamine exhibit hyperlocomotion in an open field

taken to emulate positive symptoms (Sturgeon et al., 1979; Nabeshima et al., 1987), deficits

in social behaviour (Sams-Dodd, 1995, 1998) and immobility in the forced swim test (Noda

et al., 1995, 2000) as behavioural models for negative symptoms, and impairments in various

learning paradigms to imitate the cognitive deficits seen in patients with SCZ (reviewed by

Mouri et al., 2007). Another behavioural model for SCZ involves a phenomenon called pre-

pulse inhibition (PPI) wherein, in healthy individuals, the application of a small stimulus

(typically auditory) before a strong stimulus reduces or inhibits the response to the stronger

stimulus. This PPI function is impaired in most patients with SCZ and is linked to

dysfunctions in sensorimotor gating and protection from sensory overload that can be

modeled in animals (Braff et al., 1992; Lipska et al., 1995). Administration of PCP or (+)-D-

aspartate 5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-iminedizocilpine (MK-

801) to postnatal rats during key neurodevelopmental periods induces PPI deficits in

adulthood, linking glutamate to this important model of SCZ (Harris et al., 2003; Takahashi

et al., 2006).

26!

!

!

!

1.4 Schizophrenia Pharmacotherapy

Antipsychotic medications are the mainstay in the pharmacological treatment of SCZ

(reviewed by Tandon, 2011). This section begins with an overview on the history and

mechanism of action of antipsychotic drugs in psychiatry, with a particular focus on

clozapine. Next, a brief review on the use of symptom rating scales in measuring response

will be presented. This section concludes with a discussion on CLZ’s unique efficacy for the

treatment of TRS and factors that contribute to variability in CLZ response.

1.4.1 Antipsychotic Medication Overview

Prior to the advent of antipsychotic therapy, patients suffering from SCZ were chronically ill,

confined to mental health hospitals and subjected to alternative treatment options such as

electroconvulsive shock therapy (ECT) and lobotomies, in which a portion of the patient’s

brain was removed (Moniz, 1937). The pioneer typical (first-generation) antipsychotic drug,

chlorpromazine (CLZ), was serendipitously discovered in the early 1950s following attempts

to develop a synthetic anti-malaria drug during WWII (Gilman et al., 1944). Rather than use

as an anti-malaria agent, CPZ was found to have anaesthetic-like properties with added

benefits: patients administered CPZ required lower doses of aneasthetic and were able to

better withstand the stress of surgical trauma (Laborit & Huguenard, 1951). CPZ was

initially supplied to two hospitals in Paris – the Central Military Hospital called Val-de-

Grâce and the psychiatric clinic of Sainte-Anne Hospital, to treat psychiatric disorders

(Delay et al., 1952). The first patient administered CPZ was a 57-year-old laborer admitted to

hospital due to erratic and uncontrollable behaviour. Following 3-weeks of CPZ treatment,

the patient appeared normal and was discharged (Hamon et al., 1952). In the decades

27!

!

!

!

following, other typical APs were developed including: haloperidol, fluphenazine,

thioridazine, loxapine, perphenazine, trifluoperazine and thiothixene.

Very shortly after the introduction of CPZ and its derivatives, extrapyramidal symptoms

including parkinsonism and akathisia became recognized as side effects associated with the

use of APs (Steck, 1954; Sigwald et al., 1959). From efforts to develop an AP that was free

of EPS came the birth of the second generation (atypical) APs, the first of which was

clozapine (CLZ). First manufactured by G. Stille at Wander Pharmaceuticals in Bern,

Switzerland in the early 1960s, CLZ was associated with little to no EPS (Hippius, 1996).

After two open studies in 1966 (Gross & Langner, 1966; Bente et al., 1966) and one double-

blind study in 1971 (Angst et al., 1971), CLZ was briefly marketed yet quickly withdrawn

due to reports of a life-threatening side effect in Finland populations (reviewed by Idanpaan-

Heikkila et al., 1997). This side effect is now recognized as clozapine-induced

agranulocytosis (CIA), which occurs in ~1% of CLZ-treated patients. CLZ was reintroduced

into the US market in 1990 following a landmark clinical trial that proved CLZ’s superior

efficacy for TRS patients (Kane et al., 1988). CLZ is currently a 3rd line treatment for

patients with treatment-resistant or intolerant forms of SCZ and is prescribed in combination

with mandatory blood monitoring (Moore et al., 2007). CLZ also has the propensity to cause

weight gain and metabolic syndrome. CLZ’s success quickly led to the development of

additional drugs in the atypical class including: risperidone, olanzapine, quetiapine,

ziprasidone, aripiprazole, paliperidone, iloperidone, asenapine and lurasidone.

1.4.2 The Mechanism of Action of Antipsychotic Drugs

All AP medications share a common primary mechanism of action, which involves blockade

of DRD2 (Carlsson & Lindqvist, 1963; Seeman & Lee, 1975; Creese et al., 1976). For this

28!

!

!

!

reason, DRD2 has been an important access point to the circuitry underlying the symptoms

of SCZ, however, may not be the primary mechanism through which CLZ achieves

therapeutic effect. All AP medications have variable binding profiles and affinities for

different combinations of receptors. CLZ’s multi-receptor profile likely accounts for its

ability to improve some domains of cognition and social function for patients with TRS

(Hagger et al., 1993; McGurk, 1999). Besides having a weaker affinity for DRD2, CLZ has a

high affinity for 5-HT2A, histamine H1 (HRH1/H1), muscarinic M1 (CHRM1/M1) and

adrenergic α-1A receptor (ADRA1A) (Meltzer et al., 1989a), and a moderate affinity for the

serotonin type 1A receptor (5-HT1A) and the adrenergic receptor α-2A receptor (ADRA2A)

(Schotte et al., 1996). This binding profile, more specifically the weak affinity for 5-HT2A

compared to DRD2, is thought to account for CLZ’s lower propensity to cause EPS (Meltzer

et al., 1989b).

1.4.3 Treatment Response Measures

Response to AP medication is commonly assessed using symptom rating scales (reviewed by

Mortimer, 2007). Several scales assessing symptom severity in SCZ have been used for this

purpose, the most common of which are the Brief Psychiatric Rating Scale (BPRS) (Overall

& Gorham, 1962), the Positive and Negative Syndrome Scale (PANSS) (Kay et al., 1987)

and the Clinical Global Impression Scale (CGI) (Guy, 1976). Symptom severity is assessed

at baseline prior to treatment and then again following a trial of AP medication. Change in

symptom scores from baseline can be used as either a categorical measure of response in

which ‘responders’ are those who exhibit a decrease in scores beyond a certain threshold, or

a continuous response measure whereby percent score reduction is calculated using the

following formula: % score change = [(6 month score - baseline score)/(baseline score)] x

29!

!

!

!

100%. The threshold used to define categorical response in this thesis was a 20-point or more

decrease in BPRS scores. This measure of response is commonly used, however, is

somewhat arbitrary and fails to reflect a patient’s personal, social and cognitive functioning –

factors that hold great weight as to how a patient will function in society (Mortimer, 2007).

Despite this limitation, the use of rating scales to assess response has proven useful in

research studies.

1.4.4 Clozapine for Treatment-Resistant Schizophrenia

As mentioned in Section 1.1.3, TRS is particularly burdensome to the healthcare system and

drastically decreases the likelihood of optimal disease outcome. The most commonly used

definition of TRS involves: 1) failure to respond to at least two periods of treatment in the

preceding five years with neuroleptic agents from at least two different chemical classes

(doses equivalent ≥1000 mg/day of CPZ for six weeks) without significant symptomatic

relief; and 2) no period of good functioning within the preceding five years (Kane et al.,

1988). This definition of TRS first emerged from the landmark clinical trial, which proved

CLZ was superior to CPZ for treatment resistant forms of SCZ (Figure 1.3). More

specifically, 268 patients who met criteria for TRS were entered into a double-blind

comparison study that lasted 6 weeks. Patients were randomly assigned to either one of two

groups: CLZ (up to 900mg/day) or CPZ (up to 1,800mg/day). Response was defined as: 1) a

20% decrease or more in BPRS total score; 2) a CGI score less than or equal to 3; and 3) a

BPRS total score equal to or less than 35, using 0-6 ratings. Results showed that a

significantly greater number of treatment-resistant patients responded to CLZ than to the

prototypical AP CPZ (30% vs 4%, p=0.001). The results of this trial were responsible for

30!

!

!

!

restoring CLZ to a position of utility in psychiatry for the treatment of patients who have

failed at least two previous drug trials or for those who develop intolerable side effects.

&

Figure&1.3!Clozapine&is&Superior&to&Chlorpromazine&for&TreatmentFResistant&Schizophrenia&

Results! from! a! landmark! clinical! trial! illustrated! clozapine’s! unique! efficacy! over! the!

prototypical! antipsychotic! chlorpromazine! (CPZ)! for! the! treatment! of! persistent! positive!

symptoms!in!patients!deemed!resistant!to!conventional!pharmacotherapy.!Mean!change!in!

total!Brief!Psychiatric!Rating!Scale! (BPRS)!scores!were!plotted!each!week!of! the!study! for!

patients! treated! with! clozapine! (solid! line,! n=126)! or! chlorpromazine! with! adjunct!

benztropine!mesylate! therapy! (broken! line,! n=139)! (p<0.001).! Reprinted! from!Archives+ of+

General+ Psychiatry! 45(9).! John! Kane,! Gilbert! Honigfield,! Jack! Singer,! Herbert!Meltzer! and!

Clozaril!Collaborative!Study!Group.!Clozapine!for!the!TreatmentAResistant!Schizophrenic:!A!

DoubleAblind!Comparison!with!Chlorpromazine,! 789A96.! Copyright! (1988)!with!permission!

from!Elsevier.

31!

!

!

!

Several additional trials have been conducted assessing CLZ’s unique efficacy for TRS. For

instance, one such study comparing clozapine, haloperidol, olanzapine and risperidone

(CHOR) in TRS found that CLZ and OLZ had similar antipsychotic efficacy, however, CLZ

was the most effective for treating negative symptoms (Volavka et al., 2002). Additional

studies indicate CLZ shows greater benefits for TRS as compared to OLZ (Conley et al.,

1999; Meltzer et al., 2008). Results from the Clinical Antipsychotic Trials of Intervention

Effectiveness Project (CATIE) also suggest that CLZ is superior to other AP drugs for

treatment resistance (McEvoy et al., 2006). Briefly, CATIE was a double-blind randomized

clinical trial funded by the National Institute of Mental Health (NIMH) that compared the

effectiveness of first- and second-generation APs used to treat SCZ. Patients who

discontinued treatment in the first phase with one of four newer atypical APs (olanzapine,

quetiapine, risperidone or ziprasidone) because of inadequate efficacy (n=99) were randomly

assigned to either open-label CLZ (n=49) or blinded treatment to an alternative atypical in

the second phase. Discontinuation due to lack of efficacy was significantly less in the CLZ

group than in the other four groups (11% vs ~40%). It should be noted that this study

disallows for firm conclusions regarding the advantages of CLZ because of the unblinded

nature with which CLZ was used. However, given the results from other studies, CLZ does

appear to be superior for TRS.

1.4.5 Variability in Clozapine Response

Response to CLZ is highly variable and likely influenced by a combination of clinical,

demographic, environmental and genetic factors (reviewed by Arranz & Munro, 2011). Non-

genetic factors that alter clinical response include: treatment adherence (Dunbar-Jacob &

Mortimer-Stephens, 2001), previous drug exposure and duration of illness (Perkins et al.,

32!

!

!

!

2005), diet, smoking and concomitant medications (Meyer, 2001; de Leon, 2004), as well as

gender, ethnicity and age (Palego et al., 2002). Response to APs is also overshadowed by the

occurrence of adverse side effects that result in less than optimal treatment outcomes.

Regarding genetic factors, twin and family studies of CLZ response indicate concordance for

response (Vojvoda et al., 1996). Like SCZ, response to CLZ is thought to be a complex trait

likely depending on the multiplicative effect of several genes that are involved in drug action

(International Schizophrenia Consortium et al., 2009; Sadee, 2013). However, drug response

phenotypes likely depend on fewer genes than those involved in disease etiology due to the

relatively narrow pathway a drug follows through the body to mediate therapeutic effect

(Johnson et al., 2011; Ma & Lu, 2011). Identifying genetic variation with the ability to

improve treatment outcome remains an active area of research and is discussed in the

following section.

33!

!

!

!

1.5 Pharmacogenetics of Clozapine Response

Pharmacogenetics (PGx) is the study of gene variation as it relates to drug response and the

development of adverse side effects. This section will touch on the history of PGx,

summarize findings from PGx studies on CLZ response with a particular focus on glutamate

system related genes, and will conclude by exhibiting how ENCODE data can be used to

select functional candidate SNPs for PGx studies.

1.5.1 Pharmacogenetics Overview

The roots of pharmacogenetics travel as far back as 1866 with the work of Gregor Johann

Mendel (1822-1884) on the garden pea (Pisum sativum L.) and the establishment of the rules

of heredity (Mendel, 1866). Since Mendel’s time, there have been several noteworthy

individuals who have contributed to our present day understanding of the relationship

between genetics and drug response. Noteworthy contributions include: 1) twin studies

indicating the influence of multiple genes on the pharmacokinetics of various drugs between

the years of 1957-1970 (Vesell, 1978); 2) development of the term ‘pharmacogenetics’ by

Friedrich Vogel in 1959 (Vogel, 1959); and 3) revolutionary works by a man named Werner

Kalow (1917-2008), deemed the ‘grandfather of pharmacogenetics’, who published the first

monograph on PGx entitled ‘Phamacogenetics – Heredity and the Response to Drugs’ in

1962 (Kalow, 1962). The advent of polymerase chain reaction (PCR) in 1985 was also a

significant contribution to PGx as it paved the way for gene cloning experiments and allowed

for further characterization of genes involved in drug response (Mullis et al., 1986). Shortly

thereafter in 1987, a very important family of liver enzymes called the cytochrome P450

enzymes (CYPs), such as CYP2D6 and CYP1A2, were cloned and polymorphisms within

these genes were linked to drug response (Distlerath et al., 1985). CYP enzymes are

34!

!

!

!

particularly relevant to the field of psychiatry because they are responsible for metabolizing a

large proportion of the currently prescribed antipsychotics and antidepressants (reviewed by

Guengerich, 2008).

Present day pharmacogenetics studies in psychiatry hope to elucidate gene variants that alter

response to psychotropic medications. The current prescribing method for AP medications

utilizes a ‘one size fits all’ approach and a suitable drug is identified through trial and error.

During this trial and error period, a subset of patients is plagued by lack of efficacy and side

effects due to toxicity. Generalized treatment plans are designed to satisfy the needs of the

majority of patients and do not account for patients that lie at the extremes of the distribution.

Treatment plans also fail to consider the biological patient-specific differences that cause

certain individuals sharing similar symptoms to react differently to the same medication.

Pharmacogenetics studies seek to identify the portion of patient-specific differences that lie

within the genome. Once predictive genetic markers are identified, they can be used to guide

therapeutic dosing and to warn against increase risk for developing adverse drug reactions

(Phillips et al., 2001). To illustrate current advancements in the field of psychiatry thus far,

positive results involving the use of genetic testing in depression have been reported (Hall-

Flavin et al., 2013; Winner et al., 2013; Altar et al., 2015). In addition, our Neurogenetics

section at the Centre for Addiction and Mental Health (CAMH) in Toronto is currently

leading a randomized control trial of an enhanced version of the GeneSight Psychotropic

Test for treatment response in SCZ.

1.5.2 Clozapine and Glutamatergic Neurotransmission

An important aspect of PGx studies is variant selection. Variants are often selected from

genes involved in known pharmacodynamic and pharmacokinetic pathways that relate to

35!

!

!

!

drug action, and often include variants that are located within drug metabolizing enzymes,

membrane transporters, receptors and target proteins (Sadee, 2013). A thorough

understanding of drug action can help guide selection of genetic variants that may play a role

in response. As mentioned above, CLZ has a unique binding profile and may achieve

therapeutic effect in part by modulating glutamatergic neurotransmission. A great deal of

evidence supports this theory and is summarized in Table 1.2. The information presented in

this table was used to guide gene selection for the work presented herein.

&

Table&1.2&Evidence&Supporting&Clozapine&Interacts&with&the&Glutamate&System&

Main!Findings! References!

CLZ!Attenuates!NMDAR!AntagonistAinduced!Behaviours!in!Preclinical!Models!

PPI!Impairments!

Clozapine!(5.0mg/kg)!antagonized!the!ability!of!PCP!(1.0mg/kg)!as!well!as!MKA801!

(0.5mg/kg)!to!repeatedly!and!robustly!decrease!PPI!in!a!rat!model,!without!

interfering!with!PPI!itself!

(Bakshi!et!al.,!1994)!!

Clozapine!(7.5kg/mg),!but!not!haloperidol!(0.1mg/kg),!significantly!restored!PPI!in!

ketamine!(1,!3,!or!6mg/kg)!treated!rats!

(Swerdlow!et!al.,!1998)!

Clozapine!(2.5mg/kg)!but!not!haloperidol!(0.035mg/kg)!successfully!reversed!PCPA

induced!PPI!deficits!in!a!primate!model!(Cebus!apella)!!

(Linn!et!al.,!2003)!

Clozapine!(5!or!10!mg/kg),!but!not!risperidone!(0.1!and!1mg/kg)!restored!deficits!

in!PPI!induced!by!MKA801!(0.3mg/kg)!in!a!rat!model!

(Bubenikova!et!al.,!2005)!!

Clozapine!(1.25!or!2.5mg/kg)!significantly!attenuated!PPIAimpairments!induced!by!

MKA801!(0.05mg/kg)!in!a!rat!model!

(Levin!et!al.,!2007)!

Hyperlocomotion!!!

Clozapine!and!haloperidol!(variable!doses)!were!moderately!effective!in!blocking!

PCPAinduced!(5.0mg/kg)!locomotor!stimulation!in!rats!

(Freed!et!al.,!1984)!

Clozapine!(2.5!or!5mg/day)!and!olanzapine!(0.25!or!0.5mg/kg)!potently!

antagonized!MKA801Ainduced!(0.05!or!1.1mg/kg)!locomotion!and!falling!assay!

(MKA801ALF)!in!rats!!

(Corbett!et!al.,!1995)!

Clozapine!(5A20mg/kg)!and!haloperidol!(0.01A0.1mg/kg)!progressively!potentiated!

hyperlocomotion!induced!by!PCP!administration!(3.2mg/kg)!in!rats!

!

(Sun,!Hu!&!Li,!2009)!

36!

!

!

!

Cognitive!and!Social!Deficits!

Acute!administration!of!clozapine!(5mg/kg)!but!not!haloperidol!(0.05mg/kg)!or!

chlorpromazine!(2mg/kg)!to!rats!significantly!reversed!PCPAinduced!cognitive!

impairments!using!an!operant!reversalAlearning!paradigm!following!subAchronic!

PCP!administration!(2mg/kg,!twice!daily!for!7!days)!

(AbdulAMonim!et!al.,!

2006)!

Clozapine!(2.5!or!5mg/day)!and!olanzapine!(0.25!or!0.5mg/kg)!significantly!

reversed!social!withdrawal!induced!by!PCP!(2mg/kg)!in!rats!!

(Corbett!et!al.,!1995)!

CLZ!Alters!Glutamate!Receptor!Expression,!Density!and!Binding!

NMDAR!

In+situ!hybridization!of!riboprobes!for!NMDAR!subunits!NR2A,!NR2B!and!NR1!

splice!variants!indicated!increased!NR1!and!NR2B!subunit!expression!in!rat!brains!

following!subchronic!administration!of!clozapine!(30mg/kg)!!

(Meshul!et!al.,!1996)!

Chronic!clozapine!administration!(30mg/kg/day!for!21!days)!resulted!in!a!20%!

increase!of!NMDA!receptors!localized!to!the!dentate!gyrus!of!the!hippocampus!in!

rat!brains!

(Giardino!et!al.,!1997)!

Chronic!administration!of!clozapine!(30mg/kg/day!for!30!days)!increased!NMDAR!

binding!in!insular!and!parietal!cortices!in!rat!brain!using!radioAligand!binding!and!

quantitative!autoradiography!with![3H]CGP!39653!

(Ossowska!et!al.,!1999)!

Rats!chronically!administered!clozapine!(45mg/kg/day!for!6!months)!showed!upA

regulated!NMDAR!binding!in!the!nucleus!accumbens,!and!downAregulated!

expression!of!NR2A!in!the!hippocampus!and!PFC!and!NR1!in!the!dorsolateral!PFC!

as!indicated!by!quantitative!receptor!autoradiography!with![3H]MKA801!!!

(Schmitt!et!al.,!2003)!

In+vivo+single!photon!emission!tomography!(SPET)!using!the!NMDAR!tracer!

[123I]CNSA1261!indicated!significant!reductions!in!relative!NMDAR!binding!in!the!

left!hippocampus!of!medicationAfree,!but!not!clozapineAtreated!SCZ!patients,!as!

compared!to!healthy!subjects!

(Pilowsky!et!al.,!2006)!

AMPAR!

Immunoblotting!procedures!in!rat!brains!showed!clozapine!administration!(35A

40mg/kg)!significantly!increased!AMPAR!subunit!GluR1!expression!in!the!medial!

prefrontal!cortex!(mPFC)!

(Fitzgerald!et!al.,!1995)!

Clozapine!administration!significantly!increased!AMPAR!density!in!the!frontal!and!

anterior!cingulated!cortices!compared!with!normal!controls!in!a!rat!model!

(Spurney!et!al.,!1999)!

CLZ!Increases!Glutamate!Concentrations!

Acute!administration!of!clozapine!(25mg/kg)!but!not!haloperidol!(0.5!or!1.0mg/kg)!

significantly!increased!extracellular!glutamate!levels!over!time!in!the!mPFC!of!

freeAmoving!rats!as!indicated!by!in+vivo!intracerebral!microdialysis!!

(Daly!&!Moghaddam,!

1993)!

Chronic!clozapine!administration!(20mg/kg/day!for!21!days)!stimulated!glutamate!

release!from!the!nucleus!accumbens!in!rats!as!indicated!by!in+vivo+microdialysis!

(Yamamoto!&!

Cooperman,!1994)!

Clozapine!treatment!significantly!increased!serum!glutamate!concentrations!in!7!

patients!with!SCZ!after!switching!from!conventional!neuroleptics!

(Evins!et!al.,!1997)!

37!

!

!

!

CLZ!Activates!Excitatory!Glutamatergic!Neurotransmission!!

Clozapine!administration!(20mg/kg)!potentiated!NMDARAmediated!synaptic!

responses!in!the!dentate!gyrus!of!chronically!prepared!rabbits!!

(Kubota!et!al.,!1996,!

2000)!

Clozapine!administration!produced!a!marked!facilitation!(300A400%)!of!NMDARA

evoked!responses!and!produced!bursts!of!EPSPs!caused!by!increased!release!of!

excitatory!amino!acids!in!pyramidal!cells!of!the!mPFC!in!rat!brain!slices!as!

indicated!by!intracellular!recording!and!singleAelectrode!voltage!clamp!

(Arvanov!et!al.,!1997;!

Chen!et!al.,!2003;!Ninan!

et!al.,!2003)!

Administration!of!CLZ!reversed!PCP!effects!on!pyramidal!cell!firing!and!cortical!

synchronization!as!recorded!extracellularly!with!glass!micropipettes,!and!

prevented!PCPAinduced!increases!in!cAfos!expression!in!PFC!pyramidal!neurons!as!

indicated!by!in+situ+hybridization!

(Kargieman!et!al.,!2007)!

To summarize, CLZ is thought to interact with the glutamate system due to its ability to

block NMDAR antagonist-induced behaviours in preclinical models including: PPI

impairments induced by PCP (Bakshi et al., 1994; Linn et al., 2003), MK-801 (Bubenikova

et al., 2005; Levin et al., 2007) and ketamine (Swerdlow et al., 1998), as well as

hyperlocomotion, social deficits and impaired cognitive function induced by PCP and MK-

801 (Freed et al., 1984; Corbett et al., 1995; Abdul-Monim et al., 2006; Sun et al., 2009).

Functional studies in preclinical models also suggest that CLZ alters glutamate receptor

subunit expression, binding and density. More specifically, CLZ administration has been

shown to: increase AMPAR density in the frontal and anterior cingulate cortices; increase

GluR1 expression in the medial prefrontal cortex (mPFC) (Fitzgerald et al., 1995; Spurney et

al., 1999); increase NR1 and NR2B expression in select brain regions (Meshul et al., 1996);

decrease NR2A expression in the PFC and hippocampus (Schmitt et al., 2003); increase

NMDAR expression in the hippocampus (Giardino et al., 1997); increase NMDAR binding

in insular and parietal cortices, as well as nucleus accumbens (NAcc) (Ossowska et al.,

1999); increase mGluR2 signaling (Fribourg et al., 2011); decrease EAAT2 expression in

cerebral cortex (Melone et al., 2001); increase synaptic glycine concentrations (Javitt et al.,

38!

!

!

!

2005); and down-regulate promoter methylation of GAD1 (Dong et al., 2008). Lastly, an in

vivo single photon emission tomography (SPET) study using the NMDAR tracer [123I]CNS-

1261 showed that CLZ significantly reduced relative NMDAR binding in the left

hippocampus of medication-free, but not clozapine treated SCZ patients, as compared to

healthy subjects (Pilowsky et al., 2006).

CLZ also alters glutamate concentrations and activates excitatory glutamate

neurotransmission. More specifically, CLZ administration has been shown to: increase

extracellular glutamate levels in the mPFC and NAcc as evidenced by in vivo micro-dialysis

in free-moving rats (Daly & Moghaddam, 1993; Yamamoto & Cooperman, 1994); increase

amplitude of excitatory postsynaptic potentials (EPSPs) elicited by single electrical

stimulation in the dentate gyrus of chronically prepared rabbits (Kubota et al., 1996, 2000);

increase EPSPs in pyramidal cells of the mPFC in rat brain slices (Arvanov et al., 1997;

Chen &Yang, 2002; Ninan et al., 2003); and increase serum glutamate concentrations in SCZ

subjects switched from conventional neuroleptics to CLZ (Evins et al., 1997). Altogether,

these findings suggest that CLZ’s therapeutic benefits may in part be mediated by re-

establishing a balance in glutamatergic neurotransmission. Therefore, genetic variation

within glutamate system-related receptors, transporters and enzymes may be ideal candidates

for PGx studies in CLZ response.

1.5.3 Polymorphisms in Glutamate System and Related Genes

A comprehensive review on the pharmacogenetics of CLZ response has recently been

conducted (Kohlrausch, 2013). Studies investigating the association among glutamate system

genes and CLZ response are summarized in Table 1.3 below.

39!

!

!

!

Table&1.3&Genetic&Associations&of&Glutamate&System&Genes&and&Clozapine&Response&

Gene/polymorphism! Sample! Main!Findings! Reference!

GRIN1!!

rs11146020+

N=183!EuropeanACaucasian!&!

N=49!AfricanAAmerican!SCZ/SA!

patients!deemed!

resistant/intolerant!to!AP!therapy;!

Response!assessed!using!BPRS!

following!6!months!of!CLZ!therapy!

All!genotype,!allele!and!

haplotype!association!

analyses!were!negative!

!!

(Hwang!et!al.,!

2011)!

GRIN2A+

!GTArepeat+

N=183!EuropeanACaucasian!&!

N=49!AfricanAAmerican!SCZ/SA!

patients!deemed!

resistant/intolerant!to!AP!therapy;!

Response!assessed!using!BPRS!

following!6!months!of!CLZ!therapy!

All!genotype,!allele!and!

haplotype!association!

analyses!were!negative!

!

(Hwang!et!al.,!

2011)!

GRIN2B+

rs1806201+

N=100!Chinese!SCZ/SA!patients!

deemed!resistant/intolerant!to!AP!

therapy;!Response!assessed!using!

BPRS!after!a!minimum!of!8!weeks!

No!significant!

associations!were!

reported,!however,!CC!

carriers!had!a!marginally!

higher!mean!clozapine!

dosage!(p=0.013)!

(Hong!et!al.,!

2001)!

GRIN2B+

rs1806201+

+

N=100!Chinese!SCZ/SA!patients!

deemed!resistant/intolerant!to!AP!

therapy;!Response!assessed!using!

BPRS!after!a!minimum!of!8!weeks!

No!significant!

associations!were!

reported,!however,!mean!

clozapine!dosage!was!

higher!for!CC!carriers!

(p=0.18)!

(Chiu!et!al.,!

2003)!

GRIN2B+

!rs10193895+

+

N=183!EuropeanACaucasian!&!

N=49!AfricanAAmerican!SCZ/SA!

patients!deemed!

resistant/intolerant!to!AP!therapy;!

Response!assessed!using!BPRS!

following!6!months!of!CLZ!therapy!

All!genotype,!allele!and!

haplotype!association!

analyses!were!negative!

!

(Hwang!et!al.,!

2011)!

NRXN1!

rs1045881+

!

N=169!EuropeanACaucasian!

SCZ/SA!patients!deemed!resistant!

or!intolerant!to!AP!therapy;!

Response!assessed!using!BPRS!

following!6!months!of!CLZ!therapy!

Post+hoc+analysis!indicated!a!genotype!of!

rs1045881!was!

significantly!associated!

with!BPRS!negative!

symptom!score!

(Lett!et!al.,!

2011)!

40!

!

!

!

Briefly, the neurexin-1 gene (NXRN1) was previously associated with CLZ response in our

group (Lett et al., 2011). The rs1045881 variant was significantly associated with change in

BPRS negative symptom score following six months of CLZ monotherapy (p=0.033). The

neurexin-1 protein (NRXN1) is a neuron-specific cell-surface molecule necessary for normal

activity of the NMDAR (Kattenstroth et al., 2004). The remaining glutamate system gene

variants are polymorphisms located in the NMDAR subunit genes GRIN1, GRIN2A and

GRIN2B. Unfortunately, all variants within these genes were reported to be non-significant

with CLZ response (Hong et al., 2001; Chiu et al., 2003; Hwang et al., 2011). Two studies

did, however, report that patients carrying the CC-genotype of GRIN2B variant rs1806201

required marginally higher mean CLZ dosages than the other genotype groups (Hong et al.,

2001; Chiu et al., 2003). Given the fact relatively few pharmacogenetics studies investigating

the contribution of glutamate system gene variation to CLZ response have been conducted

thus far, additional studies are of high priority.

1.5.4 Identifying Functional Gene Variants using ENCODE

Gene variant selection can also be approached through the use of functional annotation

databases such as the University of California Santa Cruz (UCSC) Genome Browser (Kent et

al., 2002), Haploreg (Ward & Kellis, 2012), RegulomeDB (Boyle et al., 2012) and the

National Institute for Environmental Health Sciences (NIEHS) Functional SNP Prediction

Database (Xu & Taylor, 2009). These databases provide access to data generated by the

Encyclopedia of DNA Elements (ENCODE) project. In more detail, the ENCODE project is

a public consortium research effort launched by the US National Human Genome Research

Institute (NHGRI) with the aim to decipher all of the functional elements within the human

genome (ENCODE Project Consortium, 2012). Of interest to candidate SNP selection is

41!

!

!

!

mapping of functional elements such as transcription factor binding sites (TFBS), DNase I

hypersensitivity sites and regions of histone modification from data pooled from 1,640 data

sets and 147 different cell types. Originally, a large portion of non-coding DNA identified

during the Human Genome project was believed to have little importance (International

HapMap Consortium, 2003), however, ENCODE reports the majority of the human genome

(~80%) is found to be functional in some way. In fact, many non-coding variants and SNPs

identified to be associated with disease through GWAS have been found to lie within

ENCODE-annotated functional regions that lie outside of protein-coding genes. To illustrate

the use of ENCODE data in SNP selection, Figure 1.4 shows search results displaying

ENCODE functional data for the glycine transporter 1 gene (SLC6A9) variant rs16831558

genotyped within this study, as presented by the UCSC Genome Browser.

&

Figure&1.4&Identifying&Functional&Promoter&Variants&in&SLC6A94Using&ENCODE&Data&

University! of! California! Santa! Cruz! (UCSC)! Genome! Browser! track! results! for! the! glycine!

transporter!1!gene!(SLC6A9)!approximately!5kb!upstream!from!the!promoter!region.!Search!

results! highlight! the! location! of! the! reference! SNP! rs16831558! under! ‘Single! Nucleotide!

Polymorphisms’! (dbSNP! 142)! (highlighted! in! the! red! box)! relative! to! intron! 1! of! SLC6A9!

(indicated!by!the!green!arrow).!Search!settings!were!customized!using!dropAdown!options!

located! in! the!page!below.!More! specifically,! ‘Integrated!Regulation! from!ENCODE’!under!

H3K27Ac!

!DNAse1!

!Txn!Factor!

!LBNL!Enhancers!

!

42!

!

!

!

‘Regulation’!was!set!to!‘show’!and!‘common!SNPs!(142)’! located!under!‘Variation’!was!set!

to!‘pack’.!Functionally,!the!rs16831558!was!associated!with:!Layered!H3K27Ac!histone!mark!

(lysine!27!of! the!H3!histone!protein!acetylated),!with!colours! indicating!cell! type! in!which!

H3K27Ac! mark! was! identified! [Blue:! K562! (myelogenous! leukocyte),! Purple:! NHEK!

(epidermal! keratinocytes),! Pink:! NHLF! (normal! lung! fibroblast)];! DNAse! I! Hypersensitivity!

Cluster! represents! areas! of! open! chromatin! where! regulatory! regions! likely! reside,! with!

darkness! of! the! boxes! indicating! extent! of! hypersensitivity;! Txn! Factor! ChIP! indicates!

regions! of! transcription! factor! binding! as! determined! by! chromatin! immunoprecipitation!

(ChIPAseq)! DNA! binding! motif! assays;! and! Vista! HMRAConserved! NonACoding! Human!

Enhancers! from! LBNL! that! identify! distantAacting! transcriptional! enhancers! in! the! human!

genome.!This!genome!browser!screen!shot!was!constructed!using!human!genome!assembly!

version! 19! and! can! be! accessed! by! visiting:! http://genome.ucsc.edu/cgiA

bin/hgTracks?db=hg19&position=chr1%3A44494763A44508289!(Kent!et!al.,!2002)!!

!

Several functional elements reported in Figure 1.4 deserve further explanation. For instance,

K27Ac is a histone mark (modified histone protein) in which lysine 27 of the H3 histone

protein has been acetylated as determined by a ChIP-seq assay. This histone mark is thought

to increase transcription possibly by blocking the spread of the repressive histone mark

H3K27Me3 (tri-methylation of lysine 27 on H3) (Bernstein et al., 2005). Another functional

element is the DNAse I Hypersensitivity sites that locate regions of open chromatin where

regulatory elements such as promoters, enhancers and silencers likely reside (Song &

Crawford, 2010). Transcription factor chromatin immunoprecipitation (Txn Factor ChIP) is

also used and identifies regions of DNA where transcription factors (TF) are likely to bind.

Transcription factors are proteins that bind to DNA and regulate gene expression by

interacting with RNA polymerase (Wang et al., 2012). Lastly, the Vista human, mouse, rat

(HMR)-Conserved Non-Coding Human Enhancers from Lawrence Berkeley National

43!

!

!

!

Laboratory (LBNL) identifies distant-acting transcriptional enhancers in the human genome

by coupling evolutionary conserved non-coding sequences with mouse enhancer assays

(Pennacchio et al., 2006).

Pulling from the data presented in the previous sections, a total of 17 gene variants from 6

glutamate-system related genes with evidence of CLZ-interaction were selected for study

inclusion (summarized in Table 2.1 & Table 3.1). Variants were selected based on previous

investigation in neuropsychiatric phenotypes as cited in the literature, and/or potential

functionality from information gathered from functional annotation websites. The first data

chapter of this thesis investigated the association among eight gene variants in GRIN2B with

CLZ response. This gene encodes for the NR2B subunit of the NMDAR, contains 13 exons

and is mapped to region 12p12 (Mandich et al., 1994; Endele et al., 2010). The second data

chapter investigated the association among ten additional glutamate-system gene variants and

response to CLZ. More specifically, the remaining ten variants included two variants in the

mGluR2 gene, GRM2, which maps to 3p21.1 (Joo et al., 2001), one variant in the AMPAR

subunit GluR1 gene, GRIA1, which maps to 5q31.1 (Puckett et al., 1991), three variants in

EAAT2 encoded by SLC1A2 mapped to region 11p13-p12 (Li & Francke, 1995), three

variants in GlyT1 encoded by SLC6A9 mapped to 1p33 (Jones et al., 1995), and lastly, one

variants in the GAD1 enzyme gene, encoded by GAD1 that is mapped to region 2q.31 (Bu et

al., 1992). The 17 glutamate system gene variants investigated in this study include: GRIN2B

(rs7301328, rs1072388, rs12826365, rs2284411, rs1806201, rs1806191, rs3764030, rs890);

GRM2 (rs4067, rs2518461); SLC1A2 (rs4354668, rs4534557, rs2901534); SLC6A9

(rs12037805, rs1978195, rs16831558); GRIA1 (rs2195450); and GAD1 (rs3749034).

44!

!

!

!

1.6 Study Rationale

1.6.1 Summary and Hypothesis

Treatment-resistant schizophrenia is a debilitating, costly and burdensome disorder that

affects approximately 30-40% of patients diagnosed with SCZ. Disease prognosis is highly

dependent upon successful early intervention strategies and stresses the utility of having the

ability to predict likelihood of response before a drug is prescribed. Pharmacogenetics

research aims to unravel the genetic underpinnings of drug response and to develop

personalized treatment plans that are capable of guiding adjusted therapeutic doses,

decreasing adverse drug reactions, improving treatment adherence and providing patients

with a better quality of life.

This study was designed to test for associations among glutamate system gene variants and

clinical response to CLZ in SCZ or schizoaffective (SA) disorder patients deemed resistant

or intolerant to previous AP therapy. We hypothesize that putatively functional variants with

the potential to alter glutamatergic neurotransmission may in some way interact with CLZ’s

ability to achieve therapeutic effect, and may lead to variable response among study

participants. In total, 17 glutamate system gene variants were selected for study inclusion,

many of which were reported to be functional using ENCODE data.

By investing time, effort and resources in studying the relationship between putatively

functional glutamate system gene variants and response to CLZ, we ultimately hope to

contribute to the ever-evolving field of pharmacogenetics, and help make personalized

medicine a better-established approach in psychiatric care.

Taylor DL II. ORIGINAL RESEARCH ARTICLE

45

Chapter 2

GENETIC ASSOCIATION ANALYSIS OF THE NMDA RECEPTOR

SUBUNIT GENE GRIN2B WITH RESPONSE TO CLOZAPINE

Danielle L. Taylor1,2, Arun K. Tiwari1, Jeffrey A. Lieberman3, Steven G. Potkin4, Herbert Y.

Meltzer5, Joanne Knight1,2,6, Gary Remington6,7, Daniel J. Müller1,2,6, James L. Kennedy1,2,6

1Neuroscience Research Department, Campbell Family Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada

2Institute of Medical Science, University of Toronto, ON, Canada

3Department of Psychiatry, College of Physicians and Surgeons, Columbia University and the New York State Psychiatric Institute, New York City, NY, USA

4Department of Psychiatry, University of California, Irvine, CA, USA

5Northwestern University Feinberg School of Medicine, Chicago, IL, USA

6Department of Psychiatry, University of Toronto, Toronto, ON, Canada

7Schizophrenia Program, Centre for Addiction and Mental Health, Toronto, ON, Canada

This chapter was modified from the following:

Taylor DL, Tiwari AK, Lieberman JA, Potkin SG, Meltzer HY, Knight J, Remington G, Müller DJ, Kennedy JL. Genetic association analysis of the NMDA receptor subunit gene (GRIN2B) with response to clozapine. Submitted to Hum Psychopharmacol Clin Exp.

Key Words: GRIN2B, pharmacogenetics, clozapine response, glutamate, schizophrenia

46

2.1 ABSTRACT

Approximately 30% of patients with schizophrenia fail to respond to antipsychotic therapy

and are classified as having treatment-resistant schizophrenia. Clozapine is the most

efficacious drug for treatment-resistance and may deliver its superior therapeutic effects

partly by modulating glutamatergic neurotransmission. Response to clozapine is highly

variable and may depend on genetic factors as indicated by twin studies. This study

investigated eight single nucleotide polymorphisms in the N-methyl-D-aspartate glutamate

receptor subunit gene GRIN2B with response to clozapine. Standard TaqMan procedures

were used to type rs7301328, rs1072388, rs12826365, rs2284411, rs1806201, rs1806191,

rs3764030 and rs890 in 175 European schizophrenia/schizoaffective patients deemed

resistant or intolerant to previous antipsychotic therapy. Response was assessed using change

in Brief Psychiatric Rating Scale (BPRS) scores following six months of clozapine therapy.

Categorical and continuous response was assessed using Pearson’s chi-squared test and

analysis of covariance, respectively. No associations were observed between the variants and

response to clozapine. A-allele carriers (AA and AG) of rs1072388 responded marginally

better to clozapine therapy than GG-homozygotes, however the difference was not

statistically significant (puncorrected=0.067, pcorrected=0.440, assuming 6.56 independent tests).

Our findings do not support a role for these GRIN2B variants in altering response to

clozapine in our sample.

47

2.2 INTRODUCTION

Treatment-resistant schizophrenia occurs in approximately 30% (20-40%) of patients treated

with antipsychotic drugs (Borgio et al., 2007; Elkis & Meltzer, 2007; Solanki et al., 2009).

TRS patients experience suboptimal response characterized by persistent positive symptoms

that often result in long periods of hospitalization (McGlashan, 1988) and chronic severe

disability. Treatment resistance remains an immense burden to both patients and their

families and is associated with annual healthcare costs ranging 3-to-11 times higher than

schizophrenia patients who respond to treatment ($66,360-$163,795 vs. $15,500-$22,300 per

year). TRS is also associated with poorer quality of life that is approximately 20% lower than

patients with SCZ who achieve remission (0.61 vs. 0.75) (Kennedy et al., 2014).

The atypical AP drug clozapine is particularly effective for treating the persistent symptoms

experienced by patients with TRS. A landmark clinical trial conducted by Kane et al. showed

that approximately 30% of TRS patients responded to CLZ versus only 4% of patients given

the prototypical AP CPZ (p<0.001) (Kane et al., 1988). As an atypical AP, CLZ differs from

typical APs in particular dimensions, such as decreased DRD2 receptor affinity (Altar et al.,

1986) and lowered risk for adverse effects such as extrapyramidal symptoms and tardive

dyskinesia (Pickar et al., 1992; Tandon and Fleischhacker, 2005). CLZ also has the ability to

alter other neurotransmitter systems such as serotonin and glutamate (reviewed by Miyamoto

et al., 2005).

A great deal of evidence suggests that CLZ augments glutamatergic neurotransmission

(reviewed by Heresco-Levy, 2003). One pillar supporting this theory stems from

observations that CLZ is capable of blunting the psychotomimetic effects of glutamate

48

antagonists in SCZ patients (Malhotra et al., 1997). These antagonists, such as phencyclidine

and ketamine, bind to the NMDAR and elicit schizophrenia-like symptoms in healthy

controls and exacerbate psychotic symptoms in SCZ patients (Luby et al., 1959; Luisada,

1978; Javitt & Zukin, 1991; Lahti et al., 2001) – observations that form the basis of the

‘glutamate hypofunction hypothesis’ of SCZ. Collectively, these findings suggest that

blockade and underactivity of glutamate signaling at the NMDAR contributes to the clinical

presentation of SCZ. In turn, CLZ is theorized to offer therapeutic relief in part by

augmenting glutamatergic signals.

Findings from preclinical models and functional studies support this theory. CLZ

administration reverses PCP-induced psychotic-like behaviours such as hyper-locomotion

(Freed et al., 1984; Sun et al., 2009; Zhao et al., 2012), social deficits (Corbett et al., 1995),

enhanced immobility (Noda et al., 1995) and PPI deficits (Linn et al., 2003). Functional

studies in both preclinical models and SCZ patients also indicate that CLZ alters glutamate

receptor subunit expression (Fitzgerald et al., 1995; Meshul et al., 1996), binding (Giardino

et al., 1997; Pilowsky et al., 2006), and density (Ossowska et al., 1999; Schmitt et al., 2003).

In addition, CLZ appears to alter glutamate concentrations and activity of excitatory

glutamate neurotransmission in different brain regions as exhibited by: micro-dialysis studies

in rodents (Daly & Moghaddam, 1993; Yamamoto & Cooperman, 1994), increased

amplitude of EPSPs in neuronal cell cultures (Banerjee et al., 1995; Arvanov et al., 1997;

Kubota et al., 2000; Ninan et al., 2003; Kargieman et al., 2007), and increased serum

glutamate levels in patients switched to CLZ (Evins et al., 1997). Taken together, these

results provide evidence for CLZ’s ability to restore glutamate signaling – a factor that may

distinguish CLZ as the most efficacious drug for treatment-resistant patients.

49

Clinical response to CLZ is highly variable (Bleehen, 1993; Davis et al., 2003) and an

estimated 30-60% of TRS patients fail to respond to CLZ therapy (Kane et al., 1988;

Lieberman et al., 1994). These patients are diagnosed with ultra treatment-resistant SCZ

(URS) and various augmentation strategies for managing URS have proven inconclusive

(Meltzer et al., 1989b; Lieberman et al., 1994; Remington et al., 2005). Because early

intervention strategies with AP drugs attenuates disease prognosis, the ability to predict

response prior to drug administration would have important clinical utility (Wyatt & Henter,

2001; Gunduz-Bruce et al., 2005; Perkins et al., 2005; Emsley et al., 2007). Given that a

portion of the inter-individual variability in CLZ response is thought to be caused by genetic

variation (Vojvoda et al., 1996; Horacek et al., 2001; Mata et al., 2001; Theisen et al., 2005;

Hoyer et al., 2010), a number of pharmacogenetics studies investigating genetic variability in

CLZ response have been conducted (reviewed by Kohlrausch, 2013). However, no predictive

test for AP response utilizing this genetic information is currently in clinical use. The most

notable gene variants investigated for association with CLZ response include those related to

the dopamine, serotonin and to a lesser degree, glutamate neurotransmitter systems.

Glutamate is becoming increasingly popular in recent years due to novel therapies that target

this neurotransmitter system (Heresco-Levy et al., 1996; Patil et al., 2007; Pinard et al., 2010;

Hopkins, 2011). Additional advancements in SCZ genetics have also yielded promising

genome-wide hits for glutamate genes in risk (GAIN Collaborative Research Group, 2007;

Jia et al., 2012; Schizophrenia Working Group of the Psychiatric Genomics Consortium,

2014). More specifically, genome-wide significant associations with schizophrenia have

been observed in glutamate system genes such as GRM3, GRIN2A, GRIA1 and GRIN2B. The

GRIN2B gene (138252 [MIM]) codes for subunit 2B of the NMDAR and is of particular

50

interest. This gene contains 13 exons and is mapped to 12p12 (Mandich et al., 1994; Endele

et al., 2010). Single nucleotide polymorphisms in GRIN2B have been investigated for

associations with several neuropsychiatric disorders, including drug response phenotypes

(Table 2.1). A small number of GRIN2B variants have also been studied in CLZ response –

while no associations between rs1806201 (2664C/T) (Hong et al., 2001; Chiu et al., 2003) or

rs1019385 (-200T/G) (Hwang et al., 2011) have been observed, the rs1805502 variant

(T5988C) significantly predicted negative symptom change during CLZ therapy (Martucci &

Kennedy, 2010). Associations between additional GRIN2B polymorphisms and CLZ

response have not yet been investigated. This study examined for associations among eight

GRIN2B variants and clinical response to CLZ monotherapy in a sample of treatment-

resistant/intolerant patients.

2.3 METHODS

2.3.1 Subjects

One hundred and seventy-five patients meeting Diagnostic and Statistical Manual of Mental

Disorders III-R or IV (DSM-III-R/IV) criteria for schizophrenia or schizoaffective disorder

were included in this study (American Psychiatric Association, 1987, 1994). Patients were

recruited from three clinical sites: Case Western Reserve University in Cleveland, Ohio (HY

Meltzer, n=74), Hillside Hospital in Glen Oaks, New York (JA Lieberman, n=73), and the

University of California at Irvine (SG Potkin, n=28). All patients were considered European

based on self-reported ancestry. Informed consent was obtained from each study participant

prior to enrollment in accordance with the Ethical Principles for Medical Research Involving

51

Human Subjects at the Centre for Addiction and Mental Health and with the Helsinki

Declaration of 1975, as revised in 1989 (World Medical Association, 2013).

TRS was defined as failure to respond to two or more AP drug trials in the previous five

years (involving drugs from two different chemical classes, with doses ≥1000mg

chlorpromazine equivalents for four to six weeks), accompanied by no period of good

functioning within the preceding five years (Kane et al., 1988). Less than 15% of study

participants met criteria for treatment intolerance, which was defined as the presence of

moderate to severe TD and/or extreme EPS making treatment with therapeutic dosages

intolerable (Lieberman et al., 1994). Before beginning CLZ treatment, patients underwent a

two to four week wash-out period which involved no administration of pharmacotherapy

unless clinically necessary. Following the washout period, all patients were treated with CLZ

monotherapy, with mean dosages of 453 mg/d, for a period of six months or longer.

Benzodiazepines were administered intermittently during the titration period. Throughout

treatment, CLZ serum levels were monitored to ascertain adherence to the medication.

2.3.2 Response Measures

Baseline Brief Psychiatric Rating Scale (Overall & Gorham, 1962) scores were obtained at

time of study enrollment. Following CLZ administration, response was evaluated after six

months using two BPRS scoring methods. The first was a categorical responder/non-

responder response measure that classified responders as individuals who experienced a

≥20% decrease in BPRS total score. The second scoring method was a quantitative response

measure that divided the BPRS items into three subcategories [positive (BPOS), negative

(BNEG), and general (BPRS)]. Percent score reduction for each of these subscales was

52

calculated using the following equation: [(six month score - baseline score)/(baseline score)]

x 100%.

2.3.3 SNP Selection

A literature search was conducted on PubMed for the following terms: “GRIN2B”,

“NMDAR”, “N-methyl-D-aspartate receptor”, “clozapine” and “pharmacogenetics.”

Literature pertaining to genetic association studies in psychiatric disorders and psychotropic

drug response was reviewed to identify GRIN2B SNPs of interest. Variants previously

investigated for an association with CLZ response or other psychiatric phenotypes were

considered for inclusion. Altogether, eight GRIN2B polymorphisms were selected:

rs1072388, rs12826365, rs1806191, rs2284411, rs3764030, rs7301328 (366G>C), rs1806201

(2664C>T), and rs890 (Figure 2.1 & Table 2.1). These eight variants were assessed for

potential functionality using the National Institute of Environmental Health Sciences

Functional SNP Prediction (FuncPred) database (Xu & Taylor, 2009). Variants were deemed

functional if alternate alleles were thought to have differential effects on gene transcription,

translation, or splicing.

Variants rs1072388 and rs12826365 were the most significant p-value SNPs from the

Genetics Association Information Network (GAIN) schizophrenia GWAS from the European

and African samples, respectively (GAIN Collaborative Research Group, 2007; Ayalew et

al., 2012). Variant rs1806191 was previously investigated in SCZ risk and obsessive

compulsive disorder (OCD) (Di Maria et al., 2004; Alonso et al., 2012), while variant

rs2284411 had been investigated in attention deficit hyperactivity disorder (ADHD) (Dorval

et al., 2007; Park et al., 2013). The promoter variant rs3764030 had previously been studied

53

in autism spectrum disorder (ASD) (Yoo et al., 2012) and is proposed to affect GRIN2B

expression by altering transcription factor binding. Both rs7301328 and rs1806201 were

investigated in a number of psychiatric phenotypes, including response to lithium

(Szczepankiewicz et al., 2009) and CLZ (Hong et al., 2001). These SNPs both lie near

intron-exon borders (within two base-pairs, specifically) and are proposed to regulate mRNA

spicing. Lastly, rs890 had been investigated in SCZ risk (Di Maria et al., 2004), treatment-

resistant depression (TRD) (Zhang et al., 2014), CLZ-induced OCD (CI-OCD) (Cai et al.,

2013) and response to lithium (Szczepankiewicz et al., 2009). This variant lies in a micro

RNA (miRNA) binding site at the 3’ untranslated region (UTR) of GRIN2B and is proposed

to alter translation initiation.

2.3.4 DNA Isolation and Genotyping

Venous blood samples were collected from study participants and sent to CAMH in Toronto,

Canada where genomic DNA was isolated using the high salt method (Lahiri & Nurnberger,

1991). GRIN2B genotypes were determined using TaqMan allele specific single tube assays,

the ABI Prism®7500 Sequence Detection System and the ABI allelic discrimination

software according to the manufacturer’s instructions (Applied Biosystems, Foster City, CA,

USA). The reaction mixture consisted of 20ng of genomic DNA, 2X TaqMan® Universal

Master Mix and 40X SNP Genotyping Assay for a final reaction volume of 10 µL. The

polymerase chain reaction (PCR) protocol consisted of a denaturation step for 10 mins at

95°C, followed by 50 cycles of amplification that consisted of denaturation (92°C for 15 s)

and annealing (60°C for 1 min). Genotype calls were confirmed by two independent

researchers and 10% of the total sample was re-genotyped to ensure genotyping accuracy.

Discordant genotypes were set as missing in the statistical analysis.

54

2.3.5 Statistical Analysis

Quality-control (QC) was carried out using PLINK software v1.07 (Purcell et al., 2007). To

pass filtering, SNPs were required to have a ≥90% genotyping rate and a minor allele

frequency (MAF) of ≥0.05. Individuals with a genotype success rate less than 75% across all

markers (samples producing genotypes for five SNPs or less) were also excluded from

analysis. All analyses were carried out using the Statistical Package for the Social Sciences

(SPSS) v20 software (IBM Corp, Armonk NY, USA) and PLINK.

Descriptive statistics were obtained for each clinical site. Mean age and baseline values of

BPRS, BPOS and BNEG subscale scores across sites were compared using Student’s t-test or

analysis of variance (ANOVA), while frequency counts for gender and response rates were

compared using Pearson’s chi-square test (χ2-test). Genotype and allele frequencies in the

responder/non-responder (R/NR) groups were compared using χ2-test or Fisher’s exact test

(for cell counts less than 5). Differences in percent score reduction in BPRS, BPOS and

BNEG subscales between genotype groups were compared using analysis of covariance

(ANCOVA), co-varying for age and baseline scores.

Hardy-Weinberg equilibrium (HWE) and linkage disequilibrium (LD) among SNPs were

determined using Haploview v4.2 using the Solid Spine of LD to construct LD blocks

(Figure 2.2) (Barrett et al., 2005). Haplotype analysis was carried out in UNPHASED v3.1.5

(Dudbridge, 2008). Haplotypes with <5% frequency were removed from analysis. Power

calculations were conducted in Quanto v1.2.4 (Gauderman & Morrison, 2006). Correction

for the effective number of independent markers was performed using the method described

by Nyholt and resulted in a statistical significance threshold of p<0.0076 (Nyholt, 2004).

55

2.4 RESULTS

2.4.1 Sample Characteristics

Of the eight SNPs genotyped, seven met QC criteria and were included in subsequent

association analyses. The genotype distribution for variant rs7301328 significantly deviated

from HWE (p=1.93x10-7) and was therefore excluded. The remaining SNPs had a mean

genotyping rate of 99.1%. Regarding study participants, 18 patients were removed due to low

genotyping efficiency (<75% of markers were successfully genotyped). The remaining

sample had a mean genotyping efficiency of 98.9%.

Following QC, 157 European treatment-resistant or intolerant patients were included in the

study (Table 2.2). Our sample had over 80% power to detect an odds ratio (OR) as low as

2.00 (unmatched case-control design: n=157, non-responder frequency=48.4%,

MAF=30.5%, α/2=0.05) and down to 8.5% variance in the quantitative response variable

(continuous design: n=85). The average minor allele frequency across the seven GRIN2B

variants (excluding rs7301328) was used for power calculations. Patients between clinical

sites did not differ in gender, categorical response rates, or BPRS/BPOS/BNEG baseline

scores. The sites did differ, however, in mean age (F(2,157)=7.10, P=0.001). Post hoc

analysis showed that the average age of the HY Meltzer sample was significantly lower than

the SG Potkin sample. In order to combine samples for analysis, age and baseline

BPRS/BPOS/BNEG scores were added as covariates to account for any disparities.

Following QC, the final combined sample (n=157) consisted of 76.4% male patients and had

a total mean age of 35.06 years (SD 8.1). Approximately half (51.6%) of the sample was

56

considered CLZ ‘responders’ (n=81). Quantitative response measures were available for a

subset of patients in the BPRS (n=86), BPOS (n=84), and BNEG (n=85) subscales.

2.4.2 GRIN2B SNP and Haplotype Association Analysis

Genotype and allele frequencies for each of the seven GRIN2B variants were not

significantly different between CLZ responder and non-responder groups or as measured by

the quantitative percent score reduction in BPRS, BPOS and BNEG subscales (Table 2.3).

Variant rs1072388 A-allele carriers (AA and AG) responded marginally better to CLZ

therapy than GG-homozygotes, however this difference was not statistically significant

(puncorrected=0.067) (Figure 2.3). Haplotype association analyses confirmed allele and

genotype findings with no association reported across all haplotypes tested (Table 2.4).

2.5 DISCUSSION

We report no significant associations among the seven GRIN2B variants investigated and

response to CLZ in our sample of patients. Despite A-allele carriers of the intronic variant

rs1072388 responding marginally better to CLZ therapy than GG-homozygotes

(puncorrected=0.067, genotype; puncorrected=0.069, allele), this observation was statistically non-

significant (pcorrected=0.440, assuming 6.56 independent tests). To our knowledge, this is the

first reported study to investigate associations of the remaining seven SNPs (rs1072388,

rs3764030, rs12826365, rs2284411, rs1806191, rs890 and rs7301328) with CLZ response.

The rs1806201 marker has previously been investigated in two other CLZ response samples

(Hong et al., 2001; Chiu et al., 2003). Each aforementioned variant was selected based on

previous investigation in CLZ response or other psychiatric phenotypes. After inclusion, the

57

potential functionality of these variants was assessed using the NIEHS Functional SNP

Prediction Database (Xu & Taylor, 2009).

Our negative finding for rs1806201 (2664C/T) is in accordance with previous reports that

this variant was not associated with CLZ response (Hong et al., 2001; Chiu et al., 2003).

These studies did report a marginally higher mean CLZ dosage for patients carrying the CC

genotype of rs1806201. Due to the inter-individual variability in CLZ metabolism and

absorption, future studies may benefit from measuring serum CLZ levels as opposed to

dosage in order to predict response. An optimal threshold for CLZ serum levels has already

been established (≥450 ng/mL) and may be of clinical utility to identify non-responders

(Lindenmayer & Apergi, 1996).

As a measure of genotyping quality, we tested for HWE to ensure genotype proportions

observed in the study population coincided with those expected under equilibrium conditions

(Hardy, 1908; Weinberg, 1908). The potentially functional variant rs7301328 deviated from

HWE (p<0.05, data not shown). This deviation may have been caused by a number of

reasons including systematic genotyping errors that mistyped heterozygotes as homozygotes

or vice versa (Gomes et al., 1999; Hosking et al., 2004), stochastic variation, or due to the

study population’s biological characteristics. The biological explanation posits that patient

genotypes deviate from HWE because of the effect an allele has on a disease phenotype from

which the sample was non-randomly selected (Wittke-Thompson et al., 2005; Xu & Taylor,

2009). The cause for deviation from HWE in our sample appeared to be genotyping error: no

homozygous recessive genotypes were observed for either rs7301328 responder or non-

responder groups, even though this variant has a MAF of 23% and therefore, was expected to

58

have a homozygous recessive count of approximately eight people out of 157. Because many

genetic tests such as χ2 require HWE to be assumed, rs7301328 was removed from

subsequent analyses (Sasieni, 1997). Retyping of rs7301328 with a newly designed assay

remains a priority for future work.

This study had several limitations. The sample size is relatively small and may not have had

sufficient power to detect the effect sizes of these genes. To increase statistical power in the

future, it will be necessary to collect larger CLZ response samples. One promising sample is

being collected by the CRESTAR consortium in Europe (CRESTAR, 2011). Sample

heterogeneity may also have limited our findings. Patients were collected from three clinical

sites that may have varied in methods and populations. Population ancestry was also based

on self-report and in the future, a principle components analysis (PCA) on the study sample

may provide more accurate ancestry profiles (Paschou et al., 2008; Price et al., 2008; Liu et

al., 2011).

In addition, two environmental factors with the potential to alter CLZ metabolism were not

controlled for in our study: cigarette smoking and caffeine consumption. Cigarette smoking

induces the Cytochrome P450 hepatic enzyme CYP1A2 that is primarily responsible for CLZ

metabolism (Bertilsson et al., 1994; Ghassabian et al., 2010). As a result, smokers are

observed to have lower CLZ serum levels than non-smokers (Seppala et al., 1999; Palego et

al., 2002), and therefore require higher CLZ dosages to achieve similar plasma levels as

patient who do not smoke. In contrast, caffeine inhibits CYP1A2 and increases both CLZ

plasma concentrations and the risk of developing side effects linked to toxicity (Odom-White

& de Leon, 1996; Hagg et al., 2000). Future studies incorporating these environmental

59

factors may help identify more accurate predictors of response and develop dosage

guidelines that are tailored to the individual patient.

Inter-study variability may also have limited comparison of results across studies. We

evaluated treatment response following six months of CLZ therapy, while previous studies

evaluated response after shorter periods, e.g. two months of treatment (Hong et al., 2001;

Chiu et al., 2003). Differences in treatment length are particularly problematic because

response to CLZ is variable and may take up to five months or longer in a subset of patients

(Lieberman et al., 1994; Meltzer & Okayli, 1995b). Thus, patients deemed non-responders

after a two month treatment period have the potential to be reclassified as responders

following a longer trial of CLZ therapy. This disparity makes comparing results across

studies problematic and reaching a consensus regarding treatment duration among

researchers would make meta-analyses more efficient in the future.

To conclude, we report no associations between seven GRIN2B variants and CLZ response in

our sample. Response to CLZ therapy is complex and likely depends on multiple gene

systems and environmental factors. Therefore, investigation of additional glutamate variants

and variants within related neurotransmitter systems is warranted for future studies. Even

though conclusive predictive markers remain elusive thus far, continued research to identify

such markers remains of high interest to improve clinical response and treatment outcomes.

60

Figure'2.1!Human'GRIN2B'Gene'Diagram''

Structure,! location! and! size! of! the! GRIN2B! gene! are! noted! along! with! the! eight! single!

nucleotide! polymorphisms! genotyped! in! this! study.! The! coding! region! of! GRIN2B' is!

represented!by!darkly!shaded!boxes!and!the!5’!and!3’!untranslated!regions!are!represented!

by!lightly!shaded!boxes.!!

61

Table'2.1!GRIN2B'Gene'Variants'Genotyped'in'the'Sample!

SNP!ID! Mn>Mj!

Region,!Position!!!

NIEHS!SNP!FuncPred!

MAF*! References!

rs3764030!(R489G>A)!

T>C! Promoter!14133332!

NIEHS:!TFBS;!Transition!!substitution!!

0.246! ASD:!(Yoo!et!al.,!2012)!

rs7301328!(366C>G)!!

C>G! Exon!2!14018777!

NIEHS:!ESS/ESE;!Transversion!substitution!

0.229! SCZ:!(Ohtsuki!et!al.,!2001;!Di!Maria!et!al.,!2004;!Qin!et!al.,!2005);!TRS:!(Chiu!et!al.,!2003);!TRD:!(Zhang!et!al.,!2014);!Lithium!Response:!(Szczepankiewicz!et!al.,!2009)!

rs12826365! A>G! Intronic!13948270!

R!Transition!substitution!!

0.197! GAINRAfrican!American!best!pRvalue!SNP!(Ayalew!et!al.,!2012)!

rs1072388! A>G! Intronic!13938776!

R!Transition!substitution!

0.203! GAINREuropean!best!pRvalue!SNP!!(Ayalew!et!al.,!2012)!

rs2284411! T>C! Intronic!13866172!

R!Transition!substitution!

0.317! ADHD:!(Dorval!et!al.,!2007;!Park!et!al.,!2013)!

rs1806201!(2664C>T)!

A>G! Exon!13!13717508!

NIEHS:!ESS/ESE;!Transition!substitution!

0.288! SCZ:!(Nishiguchi!et!al.!2000;!Ohtsuki!et!al.,!2001;!Di!Maria!et!al.,!2004;!Qin!et!al.,!2005);!CLZ:!(Hong!et!al.,!2001);!TRS:!(Chiu!et!al.,!2003);!TRD:!(Zhang!et!al.,!2014)!

rs1806191!(3534C>T)!

A>G! Exon!13!13716638!

R!Transition!substitution!

0.462! SCZ:!(Di!Maria!et!al.,!2004);!!OCD:!(Alonso!et!al.,!2012)!

rs890!(409T>G)!(5072T>G)!

A>C! 3’UTR!13715308!

NIEHS:!miRNA;!!!Transversion!substitution!

0.494! SCZ:!(Di!Maria!et!al.,!2004);!Lithium!response:!(Szczepankiewicz!et!al.,!2009);!TRD:!(Zhang!et!al.,!2014);!CIROCD:(Cai!et!al.,!2013)!

NA:!not!available;!Mn>Mj:!minor>major!allele;!UTR:!untranslated!region;!MAF:!minor!allele!frequency!in!our!sample!(n=157);!NIEHS:!National!Institute!of!Environmental!Health!Sciences!(TFBS:!transcription!factor!binding!site;! ESE/S:! Exonic! splicing! enhancer/silencer;! miRNA:! microRNA! binding! site);! References:! GAIN:! Genetic!Association!Information!Network!GWAS;!ADHD:!attention!deficit!hyperactivity!disorder;!ASD:!autism!spectrum!disorder;!CIROCD:!clozapineRinduced!OCD;!CLZ:!clozapine!response;!OCD:!obsessive!compulsive!disorder;!SCZ:!schizophrenia;!TRD:!treatmentRresistant!depression;!TRS:!treatmentRresistant!SCZ.!

62

Figure'2.2!GRIN2B'Linkage'Disequilibrium'Map''

Linkage!disequilibrium!among!eight!GRIN2B!variants! included! in!this!study.!The!haplotype!

block!was! constructed!with! solid! spine!of! LD!using! the! standard! (D’/LOD)! colour! scheme.!

White:!D’<1,!LOD<2;!blue:!D’=1,!LOD<2;!shades!of!pink/red:!D’<1,!LOD≥2;!bright!red:!D’=1,!

LOD≥2.!Values!within!each!box!represent!correlation!coefficient!values!as!percentages!(r2)

(Haploview!v4.2).!

63

Table'2.2!Study'Sample'Demographics'and'Clinical'Information!

! Meltzer!!(n=64)!

Lieberman!!(n=65)!

Potkin!(n=28)!

'p)value'

Total!!(n=157)!

Age!(mean±SD)! 32.67±8.1! 35.66±7.9! 39.14±6.6! 0.001b! 35.06±8.1!

Gender! ! ! ! 0.196c! !

!!!!!Male!n!(%)! 48!(75.0)! 47!(72.3)! 25!(89.3)! ! 120!(76.4)!

!!!!!Female!n!(%)! 16!(25.0)! 18!(27.7)! 3!(10.7)! ! 37!(23.6)!

R/NRa!n!!

(%)!

31/33!!

(48.4/51.6)!

37/28!!

(56.9/43.1)!!

13/15!!

(46.4/53.6)!

0.524c! 81/76!!

(51.6/48.4)!

ΔBPRS!scores!(n=86)! R10.64±13.0! NA! R6.82±11.8! 0.192b! R9.40±12.67!

ΔBNEG!scores!(n=88)! R1.19±3.5! NA! R0.57±4.2! 0.470b! R0.99±3.70!

ΔBPOS!scores!(n=84)! R3.43±5.45! NA! R3.25±6.65! 0.896b! R3.37±5.84!

SD:! standard! deviation;! R/NR:! responder/nonRresponder;! BPRS:! Brief! Psychiatric! Rating! Scale;! BNEG/BPOS:!BPRS! negative! and! positive! subscales;! NA:! data! not! available;! aResponder! defined! as! having! at! least! 20%!reduction! in!Brief!Psychiatric!Rating!Scale! scores! from!baseline;! bpRvalues! from!Student’s! tRtest!or!oneRway!ANOVA;!c!pRvalues!from!χ2Rtest.!

64

Table'2.3!Association'Analysis'of'GRIN2B'Variants'and'Response'to'Clozapine!!

SNP!HWE!

Genotype! R/NRa! ΔBPRS±SDb! ΔBPOS±SDb! ΔBNEG±SDb! Allele! R/NRa!

'rs1072388! AA! 4/1! R8.61±11.90! R7.50±3.54! R0.50±3.54! A! 39/23!

0.5692! AG! 31/21! R10.19±15.01! R3.88±4.79! R0.60±3.56! ! !

! GG! 45/50! R13.50±6.36! R3.04±6.44! R0.96±3.80! G! 121/121!

! P' 0.067c! 0.933! 0.624! 0.898! p' 0.069!

rs12826365! AA! 3/0! R12.50±2.12! R3.50±4.95R! R0.50±6.36! A! 33/28!

0.3081! AG! 27/28! R8.15±10.80! R3.50±5.87! R0.23±2.96! ! !

! GG! 52/47! R10.08±14.01! 3.29±5.95! R1.46±4.00! G! 131/122!

! P' 0.309c! 0.886! 0.503! 0.715! p' 0.745!

rs2284411! TT! 8/8! R12.11±16.62! R5.89±7.08! R0.89±2.89! T! 48/50!

0.632! TC! 32/34! R8.30±12.98! R2.00±4.60! R1.00±3.83! ! !

! CC! 42/33! R9.92±11.48! R4.16±6.44! R1.00±3.82! C! 116/100!

! P' 0.660! 0.352! 0.092! 0.892! p' 0.437!

rs1806201! AA! 6/7! R10.00±13.61R! R6.71±6.68! R0.43±3.10! A! 45/45!

1.00! AG! 33/31! R9.21±11.81! R3.21±5.27! R1.48±3.70! ! !

! GG! 42/37! 9.41±13.26! R2.98±5.99! R0.78±3.81! G! 117/105!

! P' 0.893! 0.875! 0.244! 0.398! p' 0.665!

rs1806191! AA! 19/19! R7.82±12.67! R2.86±6.35! R0.41±3.66! A! 73/74!

0.465! AG! 35/36! R8.10±12.14! R3.10±5.89! R0.64±3.64! ! !

! GG! 28/20! R13.45±13.72! R4.43±5.30! R2.29±3.72! G! 91/76!

! P' 0.595! 0.500! 0.633! 0.092! p' 0.392!

rs3764030! TT! 8/6! R6.67±9.37! R2.17±6.68! R0.50±3.15! T! 44/33!

0.0625! TC! 28/21! R10.03±10.79! R3.31±5.94! R1.30±3.55! ! !

! CC! 44/48! R9.35±14.06! R3.35±5.79! R0.86±3.89! C! 116/117!

! P' 0.522! 0.849! 0.848! 0.540! p' 0.263!

rs890! AA! 18/20! R8.35±13.40! R4.16±5.75! R0.21±3.57! A! 74/74!

0.3527! AC! 38/34! R10.19±13.36! R3.38±6.28! R1.77±3.97! ! !

! CC! 25/21! R9.24±11.85! R2.97±5.59! R0.53±3.44! C! 88/76!

! P' 0.800! 0.941! 0.718! 0.396! p' 0.518!

! HWE:!HardyRWeinberg! equilibrium;! R/NR:! responder/nonRresponder;! BPRS:! Brief! Psychiatric! Rating! Scale;!BNEG/BPOS:!BPRS!negative!and!positive!subscales;!SD:!standard!deviation;!apRvalues!from!χ!2Rtest;!bpRvalues!from!ANCOVA!coRvaried!for!age!and!baseline!scores;! cpRvalues!from!Fisher’s!Exact!Test,!category!with! low!cell!value!(<5)!collapsed!with!heterozygous!group.!

65

Figure'2.3!Genotype'Analysis'of'GRIN2B'Variant'rs1072388'and'Clozapine'Response''

ARallele!carriers!of!GRIN2B! variant! rs1072388!responded!marginally!better! to!CLZ! therapy!

than!GGRhomozygotes! as!measured! by! categorical! responder/nonRresponder! frequencies,!

however!this!observation!was!not!statistically!significant! (puncorrected=0.067,!pcorrected=0.440,!

assuming!6.56!independent!tests).!!

GG! AG!+!AA!NonRresponders! 0.698! 0.301!

Responders! 0.555! 0.444!

0!

0.1!

0.2!

0.3!

0.4!

0.5!

0.6!

0.7!

0.8!

Respon

se'Frequ

ency'

66

Table'2.4!Haplotype'Analysis'of'GRIN2B'Variants'and'Response'to'Clozapine!

! R/NR!n!(%)! ΔBPRS!n!(%)! ΔBPOS!n!(%)! ΔBNEG!n!(%)!

! rs1072388Rrs12826365Rrs2284411*!

Global!p)valuea! p''''0.188! p''''0.843! p''''0.529! p'''0.680!

ARGRC!

!

27.2/12.3!(17.4/8.4)!

0.0248!

15.3!(9.7)!

0.352!

14.2!(9.1)!

0.116!

15.0!(9.6)!

0.660!

ARGRT! 11.8/10.7!(7.6/7.3)!

0.652!

14.7!(9.3)!

0.556!

14.8!(9.5)!

0.503!

14.0!(9.0)!

0.776!

GRARC!

!

GRGRC!

!

GRGRT!

30/26!(19.2/17.8)!

0.751!

55.8/58.7!(35.8/40.2)!

0.513!

31.2/38.3!(20.0/26.2)!

0.121!

33!(20.9)!

0.909!

59.7!(37.8)!

0.724!

35.3!(22.3)!

0.598!

30!(19.2)!

0.912!

62.8!(40.3)!

0.740!

34.2!(21.9)!

0.691!

31!(19.9)!

0.269!

60.0!(38.4)!

0.764!

36.0!(23.1)!

0.238!

! rs1806201Rrs1806191Rrs890*!

Global'pRvaluea! p''''0.454! p''''0.431! p''''0.293! p'''0.230!

ARGRA!

!

45/46!(28.1/30.3)!

0.678!

44!(26.2)!

0.803!

41!(25.0)!

0.0645!

42!(25.3)!

0.267!

GRARA! 10.8/11.6!(6.8/7.7)!

0.693!

13.88!(8.3)!

0.686!

13.95!(8.5)!

0.421!

13.6!(8.2)!

0.203!

GRARC!

!

60.2/63.4!(37.6/41.7)!

0.484!

71.12!(42.3)!

0.380!

70.05!(42.7)!

0.529!

71.4!(43.0)!

0.119!

GRGRA!

!

18.2/17.4!(11.4/11.4)!

0.962!

17.12!(10.2)!

0.974!

16.05!(9.8)!

0.207!

16.4!(9.9)!

0.457!

GRGRC!

!

25.8/13.6!(16.1/9.0)!

0.0605!

21.88!(13.0)!

0.0587!

22.95!(14.0)!

0.787!

22.6!(13.6)!

0.218!

*Haplotypes!with!frequencies!<0.05!were!excluded;!auncorrected!pRvalues!from!UNPHASED!version!3.1.5.!

Taylor DL III. ORIGINAL RESEARCH ARTICLE

67

Chapter 3

PHARMACOGENETIC ANALYSIS OF GLUTAMATE SYSTEM GENE

VARIANTS AND CLINICAL RESPONSE TO CLOZAPINE

Danielle L. Taylor1,2, Arun K. Tiwari1, Jeffrey A. Lieberman3, Steven G. Potkin4, Herbert Y.

Meltzer5, Joanne Knight1,2,6, Gary Remington6,7, Daniel J. Müller1,2,6, James L. Kennedy1,2,6

1Neuroscience Research Department, Campbell Family Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada

2Institute of Medical Science, University of Toronto, ON, Canada

3Department of Psychiatry, College of Physicians and Surgeons, Columbia University and the New York State Psychiatric Institute, New York City, NY, USA

4Department of Psychiatry, University of California, Irvine, CA, USA

5Northwestern University Feinberg School of Medicine, Chicago, IL, USA

6Department of Psychiatry, University of Toronto, Toronto, ON, Canada

7Schizophrenia Program, Centre for Addiction and Mental Health, Toronto, ON, Canada

This chapter was modified from the following:

Taylor DL, Tiwari AK, Lieberman JA, Potkin SG, Meltzer HY, Knight J, Remington G, Müller DJ, Kennedy JL. Pharmacogenetic analysis of glutamate system gene variants and clinical response to clozapine. Submitted to Mol Neuropsychiatry.

Keywords: GRM2, SLC1A2, SLC6A9, GRIA1, GAD1, pharmacogenetics, clozapine response,

glutamate

68

3.1 ABSTRACT

Altered glutamatergic neurotransmission is implicated in schizophrenia etiology as well as in

response to clozapine, the drug of choice for treatment-resistant schizophrenia. Response to

antipsychotic therapy is highly variable, although twin studies suggest a genetic component.

We investigated the association among ten glutamate system gene variants with clozapine

response in 175 European schizophrenia or schizoaffective disorder patients deemed resistant

or intolerant to previous pharmacotherapy. GRM2 (rs4067, rs2518461), SLC1A2 (rs4354668,

rs4534557, rs2901534), SLC6A9 (rs12037805, rs1978195, rs16831558), GRIA1 (rs2195450)

and GAD1 (rs3749034) were typed using standard genotyping procedures. Response was

assessed following six months of clozapine treatment using change in Brief Psychiatric

Rating Scale scores. Categorical and continuous response variables were analyzed using

Pearson’s chi-squared test and analysis of covariance, respectively. No significant

associations were observed following correction for multiple testing. Prior to correction,

several nominally significant associations were seen for SLC6A9, SLC1A2, GRM2 and

GRIA1. Most notably, CC-homozygotes of rs16831558 located in the glycine transporter 1

gene exhibited an allele dose-dependent improvement in positive symptoms compared to T-

allele carriers (puncorrected=0.008, pcorrected=0.08). To clarify the role of SLC6A9 in clinical

response to antipsychotic medication and clozapine in particular, this finding warrants further

investigation in larger well-characterized samples.

69

3.2 INTRODUCTION

The glutamate system has emerged as an area of strong interest for both schizophrenia risk

and response to antipsychotic medications (Shan et al., 2012). The glutamate hypothesis of

SCZ first emerged from observations that administration of NMDAR antagonists such as

PCP and ketamine elicit behaviours that mimic the symptoms of SCZ in healthy individuals

and dramatically worsen symptoms in SCZ patients (Luby et al., 1959; Luisada, 1978; Javitt

& Zukin, 1991; Lahti et al., 2001). Glutamate levels also appear to be altered in SCZ brains

and dysregulation of glutamate receptors has been postulated to play an integral role in

hyperdopaminergic states often thought to be the primary cause of psychosis (Moghaddam &

Javitt, 2012).

CLZ is an atypical AP drug that is particularly effective for treating patients with TRS (Kane

et al., 1988). Approximately 30% of patients fail two or more trials with AP drugs, and are

subsequently diagnosed with TRS. CLZ is the one drug that has been approved for treatment

of this particular subpopulation (Borgio et al., 2007; Elkis & Meltzer, 2007; Solanki et al.,

2009). Response to CLZ is complex and thought to depend, at least in part, on genetic factors

as indicated by twin and family studies (Vojvoda et al., 1996; Horacek et al., 2001; Mata et

al., 2001; Hoyer et al., 2010). Like other APs, high inter-individual variability is also

observed with CLZ treatment (Bleehen, 1993; Potkin et al., 1994; Davis et al., 2003), with up

to 50% of eligible patients failing to respond (Kane et al., 1988; Lieberman et al., 1994).

Studies in pharmacogenetics aim to identify gene variants with the potential to predict

dosing, response and side effects prior to starting drug treatment (Müller et al., 2003). The

implications of a genetic test incorporating such variants would have far-reaching clinical

applications; given the fact that early and effective treatment has been associated with

70

favourable outcome (Gunduz-Bruce et al., 2005; Perkins et al., 2005). To date, the majority

of PGx studies investigating CLZ response have focused on gene variants in dopamine and

serotonin neurotransmitter pathways (reviewed by Kohlrausch, 2013).

As noted, there is mounting evidence to suggest that glutamatergic neurotransmission may

also play a role in mediating response to CLZ (Potkin et al., 1999; reviewed by Heresco-

Levy, 2003). The major observation supporting this hypothesis is that if CLZ is co-

administered with NMDAR antagonists, their psychotomimetic effects are blunted (Malhotra

et al., 1997). Glutamate concentrations and excitatory glutamatergic neurotransmission are

also increased by CLZ administration, as shown by micro-dialysis studies in rodents (Daly &

Moghaddam, 1993; Yamamoto & Cooperman, 1994), EPSPs in neuronal cell cultures

(Banerjee et al., 1995; Arvanov et al., 1997; Kubota et al., 2000; Ninan et al., 2003;

Kargieman et al., 2007), and serum glutamate levels in patients switched to CLZ (Evins et

al., 1997).

CLZ is also posited to modulate various receptors, transporters and enzymes that are

involved in glutamate signaling and consequently implicates single nucleotide polymorphism

gene variants within these pathways as strong candidates for assessing susceptibility to CLZ

non-response. In regard to glutamate receptors, CLZ administration has been observed to

increase AMPAR density and GluR1 expression (Fitzgerald et al., 1995; Spurney et al.,

1999). The GluR1 protein is encoded by the GRIA1 gene and has been mapped to 5q31.1

(Puckett et al., 1991). CLZ also increases mGluR2 signaling (Fribourg et al., 2011), which is

encoded by the GRM2 gene, which maps to 3p21.1 (Joo et al., 2001). AMPA and mGluR

glutamate receptors are involved in primary depolarization of glutamate-mediated

neurotransmission and synaptic plasticity (Ichise et al., 2000; Santos et al., 2009).

71

With respect to transporter proteins, CLZ reportedly down-regulates expression of EAAT2 in

rat cerebral cortex (Melone et al., 2001), which is encoded by the SLC1A2 gene that maps to

region 11p13-p12 (Li & Francke, 1995). EAAT2 localizes to astrocytes in the mammalian

CNS and is responsible for the greatest proportion of total glutamate reuptake from the

synapse (Rothstein et al., 1996). Functional inactivation of EAAT2 raises extracellular

glutamate levels – a biological phenomenon implicated in AP non-response (Egerton et al.,

2012; de la Fuente-Sandoval et al., 2013; Demjaha et al., 2014). CLZ also increases synaptic

glycine concentrations (Javitt et al., 2005). In neuronal tissue, the glycine transporter 1

chiefly determines the availability of glycine in the brain by mediating glycine reuptake into

surrounding nerve terminals and glial cells (Kim et al., 1994). The SLC6A9 gene codes for

GlyT1 and is mapped to 1p33 (Jones et al., 1995).

Lastly, CLZ has been shown to down-regulate promoter methylation of the GAD1 gene in

mice (Dong et al., 2008). This gene is mapped to 2q.31 (Bu et al., 1992) and codes for the

GAD1 enzyme, which catalyzes the decarboxylation of glutamic acid to GABA and carbon

dioxide (Erlander et al., 1991). Interestingly, abnormal GABAergic function is also reported

in patients with SCZ (Taylor & Tso, 2014). Based on this aforementioned body of evidence,

the present study set out to investigate the contribution of variants within SLC1A2, SLC6A9,

GRIA1, GRM2 and GAD1 to response to CLZ in a sample of treatment resistant or intolerant

patients.

72

3.3 METHODS

3.3.1 Study Sample

Subjects included in this study were recruited from three clinical sites: Case Western Reserve

University in Cleveland, Ohio (HY Meltzer, n=74); Hillside Hospital in Glen Oaks, New

York (JA Lieberman, n=73); and University of California, Irvine (SG Potkin, n=28) (Total

sample, n=175). All patients had a diagnosis of either SCZ or SA disorder according to the

DSM-III-R/IV (American Psychiatric Association, 1987, 1994) and met criteria for either

TRS or intolerance to standard pharmacotherapy. Treatment-resistant schizophrenia was

defined as failure to respond to two or more antipsychotic trials with drugs from at least two

different chemical classes at doses of ≥1000mg/d chlorpromazine equivalents for four to six

weeks, accompanied by no period of good functioning in the preceding five years (Kane et

al., 1988). A smaller portion of patients (<15%) met criteria for treatment intolerance defined

as the presence of moderate to severe tardive dyskinesia or extreme sensitivity to

extrapyramidal symptoms (Lieberman et al., 1994).

For this genetic study, written informed consent was obtained from all participants in

accordance with the Ethical Principles for Medical Research Involving Human Subjects at

the Centre for Addiction and Mental Health and with the Declaration of Helsinki, as revised

in 1989 (World Medical Association, 2013). Participants underwent a two to four week

washout period during which time no medication was administered unless clinically

necessary. Patients were then placed on CLZ therapy for six months or longer with mean

doses in the range of 450 mg/day. For the duration of the study, patients were seen weekly

for blood draws to monitor white blood cell count as a precaution against clozapine-induced

agranulocytosis.

73

3.3.2 Response Measures

CLZ response was assessed using both categorical and continuous response measures.

Categorical response was assessed using change in Brief Psychiatric Rating Scale scores

(Overall & Gorham, 1962) from baseline and following six months of CLZ therapy.

Participants were considered responders if they experienced a 20% or more decrease in their

BPRS scores. Continuous response was measured using percent score reduction for total

BPRS, as well as for the BPOS and BNEG subscales, using the following calculation: [(six

month score – baseline score)/baseline score] x 100%. Using this formula, scores below zero

indicated symptom improvement.

3.3.3 SNP Selection

In total, ten glutamate system gene variants were included in this study (Figure 3.1 & Table

3.1). Four variants were selected based on previous investigation in other neuropsychiatric

phenotypes as cited in the literature, and the remaining six SNPs were selected from several

functional annotation websites based on their potential to alter gene expression.

3.3.3.1 Identification of SNPs from the Literature

A literature search was conducted on PubMed using the following search terms: “GRM2”,

“Metabotropic Glutamate Receptor 2”, “SLC1A2”, “Glutamate Transporter 1”, “GLT1”,

“Excitatory Amino Acid Transporter 2”, “EAAT2”, “SLC6A9”, “Glycine Transporter 1”,

“GlyT1”, “AMPA”, “GRIA1”, “GluA1”, “GLUR1”, “GAD1” and “Glutamate Decarboxylase

1.” Articles investigating genetic associations (genome-wide and candidate SNP studies)

with neuropsychiatric phenotypes were reviewed. As well, articles investigating the

functional significance (positron emission tomography ligand binding assays, electrophoretic

74

mobility shift assays, luciferase promoter assays, etc.) of variants within SLC1A2, SLC6A9,

GRIA1, GRM2 and GAD1 were reviewed to identify glutamate system SNPs of interest. Four

variants were selected based on previous investigations in other phenotypes: 1) SLC1A2

(rs4354668) in bipolar disorder (BP), response to lithium salts (Dallaspezia et al., 2012),

EAAT2 expression in SCZ subjects (Ohnuma et al., 1998) and plasma glutamate

concentrations (Mallolas et al., 2006); 2) SLC1A2 (rs4534557) in SCZ risk (Deng et al.,

2004; Nagai et al., 2009); 3) GRM2 (rs4067) in major depressive disorder (MDD), BP,

response to fluvoxamine, methamphetamine-induced psychosis and SCZ (Tsunoka et al.,

2009, 2010); and 4) GAD1 (rs3749034) in childhood onset SCZ (COS), cortical gray matter

volume (Addington et al., 2005) and GAD1 mRNA expression (Straub et al., 2007).

3.3.3.2 Identification of SNPs with Potential Functionality

The Broad Institute’s HaploReg database was used to identify glutamate system variants with

potentially functional effects on gene expression (Ward & Kellis, 2012). The gene coding

regions plus an additional 10kb upstream and 2kb downstream were submitted to the ‘Build

Query’ search field on HaploReg. Gene regions (chrN:start-end) were obtained from the

UCSC Genome Browser using Human Genome assembly version 19: GRM2 (chr3:

51,731,081-51,754,625), SLC1A2 (chr11: 35,270,752-35,461,610), SLC6A9 (chr1:

44,455,172-44,507,164) and GRIA1 (chr5: 152,849,175-153,088,732).

Seven criteria to measure functionality were provided for each SNP: 1) sequence

conservation across mammals, 2) promoter histone marks, 3) enhancer histone marks, 4)

DNAse hypersensitivity sites, 5) bound proteins, 6) expression quantitative trait loci (eQTL)

data and 7) effect on regulatory motifs. SNPs with a minor allele frequency of at least 5% in

the European (EUR) population that satisfied at least 3 of these 7 functionality criteria were

75

considered for study inclusion. Six variants satisfying these criteria were selected: SLC1A2

(rs2901534), SLC6A9 (rs12037805, rs1978195, rs16831558), GRIA1 (rs2195450) and GRM2

(rs2518461) (Figure 3.2). RegulomeDB (Boyle et al., 2012) and NIEHS Functional SNP

Prediction (FuncPred) database scores (Xu & Taylor, 2009) were also obtained to further

assess functionality.

3.3.4 DNA Isolation and Genotyping

Venous blood samples were collected from study participants and sent to our Centre

(CAMH, Toronto, Canada) where genomic DNA was extracted using the high salt method

(Lahiri & Nurnberger, 1991). Genotyping was performed using the QuantStudio 12K Flex

Real-Time PCR System (Applied Biosystems, Foster City, CA, USA). Final PCR reaction

mixtures consisted of 20ng of genomic DNA and 2µL of 2X TaqMan® OpenArray®

Genotyping Master Mix. Samples were loaded onto a QuantStudio Digital PCR Plate using

QuantStudio 12K Flex AccuFill System and run on the QuantStudio 12K Flex Instrument as

per the manufacturer’s instructions. Genotype calls were visualized using the DigitalSuite

Software and were validated by two independent researchers blind to genotyping conditions.

To ensure genotyping accuracy, 10% of the samples were re-genotyped and conflicting

genotypes were set as missing for analysis.

3.3.5 Statistical Analysis

Quality-control of genotyping data was carried out using PLINK v1.07 (Purcell et al., 2007).

In order to pass QC, SNPs were required to have ≥90% genotyping rate and a MAF of ≥0.05,

and to satisfy HWE (p>0.005). In addition, individuals with genotyping efficiency less than

76

80% across all markers (samples producing genotypes for seven SNPs or less) were excluded

from analysis.

Descriptive statistics were carried out using SPSS v20 (IBM Corp, Armonk NY, USA).

Continuous variables were analyzed using Student’s t-test or ANOVA, and categorical

variables were analyzed using χ2-test. Genotype and allele frequencies in R/NR groups were

compared using χ2-test or Fisher’s Exact for cell counts less than 5. Differences in percent

score reduction in BPRS, BPOS and BNEG subscales among genotypic groups were

compared using ANCOVA, with age and baseline scores added as covariates.

HWE and LD among SNPs were determined using Haploview v4.2 and the Solid spine of

LD to construct LD blocks (Figure 3.3) (Barrett et al., 2005). Haplotype analysis was carried

out using UNPHASED v3.1.5 and p-values were corrected using permutation (10,000 tests)

(Dudbridge, 2008). Haplotypes with a frequency of <0.05 were removed from subsequent

analyses. Power calculations were performed using Quanto v1.2.4 (Gauderman & Morrison,

2006). Multiple testing correction for genotype and allele tests was performed using the

Nyholt method and resulted in a revised statistical significance threshold of p<0.005 (Nyholt,

2004).

3.4 RESULTS

3.4.1 Sample Characteristics

Following QC procedures, twelve individuals were removed due to low genotyping

efficiency (<80% of markers were successfully genotyped). The remaining sample had a

mean genotyping efficiency of 98.9%. All SNPs met QC criteria and had a mean genotyping

rate of 99.3%. In total, 163 European treatment-resistant/intolerant patients were included in

77

the statistical analysis (Table 3.2). Our sample had over 80% power to detect an OR as low

as 2.1 (unmatched case-control design, n=163, non-responder frequency=47.2, MAF=26.5%,

α/2=0.05) and down to 8.5% of the variance in the quantitative response variable (continuous

design, n=91). The average minor allele frequency across all ten glutamate system variants

was used for power calculations.

For the three clinical sites, no statistically significant differences were observed for gender,

response rates, or percent score reduction in BPRS, BPOS and BNEG scales. A statistically

significant difference was observed, however, for age of study participants across clinical

sites (p=0.001). Post hoc analysis revealed that patients originating from the SG Potkin

sample were significantly older than patients from the HY Meltzer sample. Baseline BPRS,

BPOS and BNEG values were also correlated with their respective percent score reductions.

In order to combine the three samples for analysis, both age and baseline scores were

included as covariates in the ANCOVA analysis of continuous response. The combined

sample (n=163) was 74.8% male and had a mean age of 35.1 years (SD 8.1). Following six

months of CLZ therapy, patients experienced a mean percent score reduction of -9.1 points

(SD 12.4), -1.1 (SD 3.6) and -3.2 (SD 5.7) from baseline for the BPRS, BNEG and BPOS

scales, respectively. In addition, 52.8% of patients experienced a ≥20% decrease in BPRS

scores and were subsequently classified as responders.

3.4.2 Genotype and Allele Association Analysis

Following correction for multiple testing, no significant differences between responder and

non-responder groups or change in percent scores using BPRS, BPOS and BNEG scales

were observed for genotype and allele frequencies for each of the ten glutamate system gene

78

variants (Table 3.3). A number of nominally significant associations were observed;

however, none of these findings were significant following correction for multiple testing.

Prior to correction, SLC6A9 rs16831558 C-allele carriers exhibited an allele dose-dependent

reduction (improvement) in BPOS subscale scores following six months of CLZ therapy

(puncorrected=0.008, pcorrected=0.08) (Figure 3.4). CC-homozygotes of rs16831558 also

exhibited greater reduction in BPRS total scores (puncorrected=0.045, pcorrected=0.421). In

addition, greater reduction in BNEG subscale scores were observed for five variants:

SLC1A2 rs4534557 homozygotes (GG and CC) (puncorrected=0.015, pcorrected=0.140), GRIA1

rs2195450 A-allele carriers (puncorrected=0.017, pcorrected=0.162), SLC1A2 rs4354668 TT-

homozygotes (puncorrected=0.026, pcorrected=0.243), GRM2 rs2518461 GG-homozygotes

(puncorrected=0.037, pcorrected=0.346), and SLC1A2 rs2901534 CC-homozygotes

(puncorrected=0.047, pcorrected=0.440).

3.4.3 GRM2 and SLC6A9 Haplotype Analysis

No haplotype blocks within GRM2 rs4067-rs2518461 were significantly different between

responder/non-responder groups, or regarding continuous response measures (Table 3.4).

Initially, the haplotype block rs16831558-rs12037805-rs1978195 T-T-A within SLC6A9

appeared significantly associated with change in BPOS scores following CLZ treatment

(puncorrected=0.001, pcorrected=0.006, permutation corrected, individual haplotype effect).

However, further investigation revealed that the positive association was driven solely by

rs16831558. When the SLC6A9 rs12037805-rs1978195 haplotype was analyzed

independently with the rs16831558 variant added as a conditioning marker, this two-marker

haplotype was no longer significant (Table 3.5).

79

3.5 DISCUSSION

This investigation explored the association between ten glutamate system gene variants and

clinical response to CLZ. No significant associations were observed following correction for

multiple testing in our sample of patients. To our knowledge, this is the first reported study to

investigate the role of SLC1A2 (rs4354668, rs4534557, rs2901534), SLC6A9 (rs12037805,

rs1978195, rs16831558), GRIA1 (rs2195450), GRM2 (rs4067, rs251) and GAD1 (rs3749034)

in clinical response to CLZ. Four of these variants were previously investigated in other

neuropsychiatric phenotypes and the remaining six SNPs were previously unstudied and

were selected using HaploReg.

Several nominally significant associations were observed for SLC6A9, SLC1A2, GRM2 and

GRIA1 with CLZ response before correction for the number of independent tests. Of interest

was the association between C-allele carriers of SLC6A9 rs16831558 and percent score

reduction in the BPOS subscale (puncorrected=0.008). Individuals carrying two copies of the C-

allele showed a 4.25% decrease in the severity of positive symptoms, those carrying one

copy experienced no symptom change, and individuals with two copies of the T-allele

experienced a worsening of their positive symptoms as indicated by BPOS scores that were

approximately 3.00% higher than at baseline. This finding became non-significant following

correction (pcorrected=0.08), but given our relatively small sample size, this nominally

significant finding for SLC6A9 rs16831558 deserves further investigation in larger CLZ

response samples.

The SLC6A9 locus is particularly complex, with 16 different mRNA transcripts and 14

different splice variants (Thierry-Mieg & Thierry-Mieg, 2006). The rs16831558 variant

80

reported herein is located approximately 5kb upstream of the SLC6A9 start-site. This variant

has a RegulomeDB functional score of 2b (‘likely to affect binding’) and is predicted to lie

within an enhancer histone mark, a DNAse I hypersensitivity site, and to affect regulatory

motifs. Therefore, characterization of this SNP’s potential functionality using mobility shift

and luciferase promoter assays remains a priority for future work.

As was mentioned in the introduction, GlyT1 has recently been the focus of several novel

therapies for SCZ (Harvey & Yee, 2013). Five phase III clinical trials investigating

bitopertin, a synthetic GlyT1 inhibitor, as an adjunct to conventional AP therapy for

persistent negative symptoms or partial responders have been conducted, although the results

have been non-significant thus far (ClinicalTrails.gov identifiers: NCT01192867,

NCT01192906, NCT01192880, NCT01235520, and NCT01235559). Because GlyT1

regulates extracellular glycine concentrations in the vicinity of NMDARs (Smith et al.,

1992), GlyT1 is able to regulate NMDAR function (Kuryatov et al., 1994; Laube et al.,

1997). By blocking glycine reuptake from the synapse, GlyT1 inhibitors cause an increase in

NR1 glycine site occupancy and NMDAR activity, ultimately reversing the NMDAR

hypofunction that is thought to contribute to SCZ etiology.

Following the same rationale, various NR1 glycine site agonists including glycine, D-serine

and D-cycloserine have been investigated as adjuncts to conventional AP therapy for the

treatment of persistent negative and cognitive symptoms in SCZ (reviewed by Javitt, 2004).

Even though glycine agonist drugs are not designed to improve the positive symptoms seen

in TRS patients, observations of their use in clinical studies to improve negative symptoms

have shed light on a possible action of CLZ on the glycine system: NR1 glycine site agonists

are generally well received when paired with most APs, however, provide little therapeutic

81

benefit and may in fact worsen symptoms when administered to patients taking CLZ (Potkin

et al., 1999; Singh & Singh, 2011). This incompatibility between NR1 glycine site agonists

and CLZ may be due to CLZ’s pre-existing ability to potentiate NMDAR-mediated

neurotransmission through an as-of-yet-unknown mechanism (Lane et al., 2006). Several

mechanisms have been posited, for instance, CLZ may already increase synaptic glycine

levels (Javitt et al., 2005) or may already act as a partial agonist at the NR1 glycine site of

the NMDAR (Schwieler et al., 2008). Alternatively, CLZ’s ability to alter dopamine activity

and achieve therapeutic effect may depend in part on availability of the NMDAR glycine site

(Schwieler & Erhardt, 2003). Future studies clarifying the role of glycine transporter gene

variants in the mechanism of CLZ’s action are necessary.

There are some limitations inherent in our study that must be mentioned. For instance, our

small sample size may not have had sufficient statistical power to detect the effect sizes of

these gene variants, and going forward the collection of larger samples will help determine

the contribution of gene variants to CLZ response. One such sample is currently being

collected by the CRESTAR consortium in Europe (CRESTAR, 2011) with the goal of

identifying markers involved in CLZ response and side effects. Heterogeneity within our

study sample may also have confounded association findings. Participants were collected

from three clinical sites that may have differed in population substructure. In addition,

patients with a diagnosis of either SCZ or SA disorder were eligible for study inclusion.

Incomplete outcome data may also have limited our findings. Individuals who ceased

treatment prior to study completion invariably have an affect on response rates and may have

confounded our ability to identify genetic variants linked to treatment response.

Unfortunately, rates for treatment discontinuation in AP drug trials are estimated to be as

82

high as 40%, with common reasons for discontinuation being noncompliance, lack of

efficacy and the occurrence of adverse side effects (Lieberman et al., 1994; Meltzer &

Okayli, 1995b; Kinon et al., 2006). In certain patients, response to CLZ may take up to five

months or longer and suggests that a portion of participants who discontinue treatment

because of lack of efficacy most likely do so prematurely. The high rates of discontinuation

during AP drug trials stresses the need for understanding inter-individual variability in drug

response phenotypes and emphasizes the utility associated with identifying clinically relevant

predictive markers.

To conclude, we report no significant associations among ten glutamate system gene variants

and response to CLZ in our sample following correction for multiple testing. A nominally

significant association within the glycine transporter 1 gene variant rs16831558 was

observed prior to correction, and deserves further examination in larger well-characterized

CLZ response samples.

83

!

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

9.7kp

rs 29 01 53 4*

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

C

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

Kobil

ka BK,

Frielle

T,

Collin

s S,

Yang-

Feng

T,

Kobil

ka TS,

Franc

ke U,

Lefko

witz

RJ,

Caron

MG, 1

987.

An

intron

less

gene

encod

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

3

5’

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

SLC1A2 (11p13-p12, 168.86kb)

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

C

o

l

l

i

n

s

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

C

o

l

l

i

n

s

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

C

o

l

l

i

n

s

K

o

b

il

k

a

B

K

,

F

ri

e

ll

e

T

,

C

o

ll

i

n

s

S

,

Y

a

n

g

-

Ko

bil

ka

BK

,

Fri

ell

e

T,

Co

lli

ns

S,

Ya

ng

-

Fe

ng

T,

Ko

bil

ka

TS

,

Fr

an

ck

e

U,

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

C

o

l

l

i

n

s

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

C

o

l

l

i

n

s

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

C

o

l

l

i

n

s

3

5’

GRM2 (3p21.1, 11.57kb)

800b

p 2.18kb

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

e

T

,

C

o

l

l

i

n

s

S

,

200b

p

78bp

SLC6A9 (1p33, 35.02kb)

5’ 3

rs 16 83 15 58*

rs 19 78 19 5*rs 12 03 78 05*

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

Ko

bil

ka

BK

,

Fri

ell

e

T,

Co

lli

ns

S,

Ya

ng

-

Fe

ng

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

2.1kb 2.9kb K

o

b

i

l

k

a

B

K

,

F

r

i

e

l

l

GAD1 67 kDa (2q31, 44.46kb)

rs 37 49 03 4*

3’

5’

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

K

o

b

i

l

Kob

ilka

BK,

Frie

lle

K

o

b

i

l

K

o

b

i

l

GRIA1 (5q31.1, 323.3kb)

*

5’

K

o

b

i

l

k

a

B

K

,

K

o

b

i

l

k

a

B

K

Kobilka BK,

Frielle T,

Collins S,

Yang-Feng

T, Kobilka

TS, Francke

U,

Lefkowitz

RJ, Caron

MG, 1987.

An

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

K

o

b

i

l

k

a

B

K

3’

84

Figure!3.1*Glutamate!System!Gene!Diagrams!!

Schematic* representation* of* GRM2,* SLC1A2,* SLC6A9,* GRIA1* and* GAD1* genes* indicating*

chromosomal* location* and* gene* size.* Single* nucleotide* polymorphisms* genotyped* in* this*

study* are* shown.*Dark*boxes* represent* coding*exons* and* light* boxes* represent* 5’* and*3’*

untranslated* regions* (UTRs).* Gene* diagrams* were* constructed* using* NCBI* Reference*

Sequence*from*Homo1sapiens,*transcript*variant*1.*Alternatively*spliced*variants*exist*for*all*

five*genes*and*can*be*viewed*on*NCBI*AceView*(ThierryVMieg*&*ThierryVMieg,*2006).

85

Figure!3.2*HaploReg!Results!for!Functional!Glutamate!System!Gene!Variants**

To* be* considered* for* study* inclusion,* variants* were* required* to* have* a* minor* allele*

frequency* of* at* least* 5%* and* to* satisfy* at* least* 3* of* 7* functionality* criteria.* Chr:*

chromosome;* pos:* position;* LD:* linkage* disequilibrium;* r2:* correlation* coefficient;* D’:*

Hedrick’s* multiVallelic* D’;* Ref:* reference* allele;* Alt:* alternate* allele;* AFR/AMR/ASN/EUR*

freq:*African/ad*Mixed*American/Asian/European*minor*allele*frequency;*GERP/SiPhy*cons:*

genomic* evolutionary* rate* profiling/SiPhy* mammalian* conservation* algorithms;* DNAse:*

DNAse* I* hypersensitivity* site;* eQTL:* expression* quantitative* trait* loci;* dbSNP* fun* annot:*

dbSNP*functional*annotation*(Ward*&*Kellis,*2012).***

86

Figure!3.3*GRM2,&SLC6A9&and&SLC1A2*Linkage!Disequilibrium!Maps!*

Haplotype* blocks*were* constructed* using* Solid* spine* of* LD* and* standard* colour* scheme.*

Bright*red*represents*Hedrick’s*multiVallelic*D’=1*and*logarithm*of*the*odds*(LOD)*≥2;*blue*

represents* D’=1* and* LOD<2;* shades* of* pink/red* represents* D’<1* and* LOD≥2;* white*

represents*D’<1*and*LOD<2.*Correlation*coefficient*values*as*percentages*(r2)*are*displayed*

within*each*box*(Haploview*v4.2).

SLC6A9 SLC1A2 GRM2

87

Table!3.1*Glutamate!System!Gene!Variants!Genotyped!in!the!Study!Sample*

Gene* SNP*ID* Mn>*Mj*

Region,*Position***

Functional*Characterization**

MAF** References*

GRM21 rs4067* A>G* Promoter*51738256*

HaploReg:*EHM;*NIEHS:*TFBS,*miRNA;*Transition*substitution*

0.163* MDD,*BP,*fluvoxamine*response:*(Tsunoka*et*al.,*2009);*METHVIP,*SCZ:*(Tsunoka*et*al.,*2010)*

GRM2* rs2518461* A>G* Promoter151738101*

HaploReg:*EHM,*DNAse,*MC;*NIEHS:*TFBS,*miRNA;*RegulomeDB*score:*2a;*Transition*substitution**

0.071* NA*

SLC1A21 rs4354668*(V181A>C)*

G>T* 5’UTR*Promoter*35440976*

HaploReg:*Cons,*PHM,*EHM,*DNAse,*PB,*MC;*NIEHS:*TFBS;*RegulomeDB*score:*2b;*Transversion*substitution**

0.457* BP,*lithium*response:*(Dallaspezia*et*al.,*2012);*SCZ:*(Ohnuma*et*al.,*1998);*Gene*expression:*(Mallolas*et*al.,*2006)*

SLC1A2* rs4534557* G>C* Intronic*35413964*

HaploReg:*Cons,*PHM,*EHM,*DNAse,*PB,*MC;*RegulomeDB*Score:*2a;*Transversion*substitution*

0.396* SCZ:*(Nagai*et*al.,*2009)*

SLC1A2* rs2901534* C>G* Intronic*35364569*

HaploReg:*PHM,*EHM,*DNAse,*PB,*MC;*Transversion*substitution**

0.211* NA*

SLC6A9* rs12037805* C>T* Intronic**44497249*

HaploReg:*Cons,*PHM,*EHM,*DNAse,*PB,*MC;*NIEHS:*TFBS;*Transition*substitution*

0.339* NA*

SLC6A91 rs1978195* G>A* 2.1kb*upstream*44499242*

HaploReg:*PHM,*EHM,*DNAse,*PB,*MC;*NIEHS:*TFBS;*Transition*substitution*

0.407* NA*

SLC6A91 rs16831558* T>C* 5kb*upstream*44502163*

Haploreg:*EHM,*DNAse,*MC;*RegulomeDB*Score:*2b;*Transition*substitution**

0.123* NA*

GRIA11 rs2195450* A>G* Intronic*152871009*

Haploreg:*Cons,*PHM,*EHM,*MC;*NIEHS:*TFBS;*Transition*substitution*

0.239* NA**

88

GAD11 rs3749034* A>G* 5’UTR*Exon*1*171673475*

Haploreg:*Cons,*PHM,*DNAse,*PB,*MC;*NIEHS:*TFBS;*Transition*substitution*

0.253* SCZ:*(Addington*et*al.,*2005;*Lundorf*et*al.,*2005;*Straub*et*al.,*2007)*

NA:*not*available;*Mn>Mj:*minor>major*allele;*UTR:*untranslated*region;*MAF:*minor*allele*frequency*in*our*sample* (n=163);* NIEHS:* National* Institute* of* Environmental* Health* Sciences* (TFBS:* transcription* factor*binding* site;*miRNA:*microRNA*binding* site);*Haploreg*Functional*Database* (2a/2b:* likely* to*affect*binding;*Cons:* evolutionarily* conserved* variant;* P/EVHM:* promoter/enhancer* associated* histone* mark;* PB:* protein*bound;*MC:*motifs* changed);* References* (BP:* bipolar* disorder;*MDD:*major* depressive* disorder;*METHVIP:*methamphetamineVinduced*psychosis;*SCZ:*schizophrenia).*

89

Table!3.2*Study!Sample!Demographics!and!Clinical!Information*

* Meltzer**(n=66)*

Lieberman**(n=69)*

Potkin**(n=28)*

1p8value1

Total**(n=163)*

Age*(mean±SD)* 32.70±7.78* 35.70±8.27* 39.15±12.78* 0.001b* 35.07±8.10*

Gender* * * * 0.150c* *

*****Male*n*(%)* 48*(72.7)* 49*(71.0)* 25*(89.3)* * 122*(74.8)*

*****Female*n*(%)* 18*(27.3)* 20*(29.0)* 3*(10.7)* * 41*(25.2)*

R/NRa*n**

(%)*

33/33**

(50.0/50.0)*

40/29**

(58.0/42.0)**

13/15**

(46.4/53.6)*

0.496c* 86/77**

(52.8/47.2)*

ΔBPRS*scores*(N=94)* V10.05±12.67* NA* V6.82±11.78* 0.253b* V9.09±12.44*

ΔBNEG*scores*(N=93)* V1.32±3.40* NA* V0.57±4.19* 0.365b* V1.10±3.65*

ΔBPOS*scores*(N=91)* V3.13±5.30* NA* V3.25±6.65* 0.925b* V3.16±5.71*

SD:* standard* deviation;* R/NR:* responder/nonVresponder;* BPRS:* Brief* Psychiatric* Rating* Scale;* BNEG/BPOS:*BPRS*negative*and*positive*subscales;*NA:*not*available;*aResponders*defined*as*having*at*least*20%*or*more*reduction* in*Brief*Psychiatric*Rating*Scale* scores* from*baseline;* bpVvalues* from*Student’s* tVtest*or*oneVway*ANOVA;*c*pVvalues*from*χ2Vtest.*

90

Table!3.3*Associations!Among!Glutamate!System!Gene!Variants!and!Clozapine!Response**

Gene1

SNP*

HWE1

Genotype* R/NRa* ΔBPRS±SDb* ΔBPOS±SDb* ΔBNEG±SDb* Allele* R/NRa*

GRM21 AA* 1/4* V6.67±12.66* 0.67±8.14* V3.00±4.36* A* 26/30*

rs4067* AG* 23/22* V8.14±10.78* V2.57±6.72* V1.14±3.91* * *

0.78381 GG* 62/51* V9.49±13.05* V3.52±5.27* V1.00±3.58* G* 148/124*

* P1 0.497c* 0.666* 0.223* 0.688* p1 0.276*

GRM21 AA* 1/0* NA* NA* NA* A* 12/12*

rs2518461* AG* 10/12* V11.30±12.53* V6.70±3.77* 0.30±3.62* * *

0.60251 GG* 75/65* V8.86±12.55* V2.76±5.80* V1.26±3.66* G* 162/142*

* P1 0.657c* 0.535* 0.335* 0.037! p1 0.756*

SLC1A21 GG* 17/14* V11.29±11.61* V3.56±5.54* V1.25±3.30* G* 80/67*

rs4354668* GT* 44/39* V7.57±11.85* V3.02±5.17* V0.17±3.13* * *

0.76051 TT* 25/24* V10.94±13.58* V3.70±6.69* V2.19±4.35* T* 94/87*

* P1 0.944* 0.430* 0.912* 0.026! p1 0.653*

SLC1A2* GG* 17/11* V12.24±10.82* V2.60±4.43* V2.00±3.16* G* 69/57*

rs4534557** CG* 33/35* V6.63±11.26* V3.91±5.82* 0.20±2.88* * *

0.11551 CC* 35/31* V9.50±14.29* V2.51±6.14* V1.86±4.31* C* 103/97*

* P1 0.548* 0.212* 0.834* 0.015** p1 0.566*

SLC1A2* CC* 5/1* V16.00±8.88* V3.80±4.32* V4.33±3.98* C* 39/28*

rs2901534* CG* 29/26* V10.12±12.21* V3.71±5.69* V1.71±3.24* * *

0.82331 GG* 52/50* V7.70±12.88* V2.80±5.93* V0.31±3.68* G* 135/126*

* P1 0.628c* 0.343* 0.551* 0.047! p1 0.343*

SLC6A91 TT* 2/3* 5.67±4.04* 3.00±1.73* 2.67±1.53* T* 22/21*

rs16831558* TC* 18/15* V7.27±11.37* 0.00±5.66* V2.13±3.83* * *

0.14621 CC* 66/59* V10.41±12.37* V4.25±5.42* V1.07±3.63* C* 152/133*

* P1 1.00c* 0.045! 0.008! 0.104* p1 0.790*

SLC6A91 CC* 14/8* V10.77±9.55* V3.50±4.77* V1.86±3.70* C* 64/43*

rs12037805* CT* 36/27* V12.09±12.95* V4.19±5.39* V1.48±4.23* * *

0.17821 TT* 35/42* V6.52±12.79* V2.39±6.27* V0.62±3.27* T* 108/111*

* P1 0.203* 0.238* 0.663* 0.198* p1 0.0746*

91

SLC6A9* GG* 17/8* V10.27±9.27* V2.00±4.761* V1.75±4.31* G* 73/57*

rs1978195* AG* 38/41* V10.74±12.36* V5.07±5.69* V1.10±3.45* 1 *

0.75211 AA* 30/26* V6.39±14.46* V1.23±5.86* V0.71±3.81* A* 99/93*

* P1 0.213* 0.445* 0.143* 0.179* p1 0.418*

GRIA11 AA* 8/4* V19.50±18.27* V6.50±2.89* V4.00±5.48* A* 46/36*

rs2195450* AG* 30/28* V8.70±11.92* V3.45±5.81* V1.26±3.19* * *

0.40351 GG* 48/44* V8.42±11.91* V2.81±5.88* V0.65±3.66* G* 128/116*

* P1 0.874c* 0.174* 0.664* 0.017! p1 0.568*

GAD11 AA* 5/6* V6.75±9.54* V3.57±6.02* V2.00±3.93* A* 44/36*

rs3749034* AG* 34/24* V10.47±13.95* V3.33±6.16* V1.70±4.33* * *

0.68781 GG* 44/47* V8.33* V3.04±5.74* V0.26±2.86* G* 124/118*

* P1 0.428* 0.794* 0.986* 0.349* p1 0.559*

HWE:* HardyVWeinberg* equilibrium;* R/NR:* responder/nonVresponder;* BPRS:* Brief* Psychiatric* Rating* Scale;*BNEG/BPOS:*BPRS*negative*and*positive*subscale;*SD:* standard*deviation;* apVvalues* from*χ* 2Vtest;* bpVvalues*from*ANCOVA*with*baseline*scores*and*age*added*as*covariates;*cpVvalues*from*Fisher’s*Exact*Test*in*which*category*with*low*cell*value*(<5)*collapsed*with*heterozygous*group;**pVvalue*from*Welch*ANOVA*test*due*to*significant*Levine’s* test;*Bolded*pVvalues* indicate*nominally* significant* findings*obtained*prior* to*correction*(<0.05).*

92

Figure!3.4*Genotype!Analysis!of!SLC6A9&rs16831558!with!Clozapine!Response!!

CVallele* carriers* of* SLC6A9* variant* rs16831558* experienced* an* allele* doseVdependent*

reduction* in* BPOS* subscale* scores* following* six*months* of* CLZ* therapy* (puncorrected=0.008,*

pcorrected=0.08,* assuming* 9.36* independent* tests),* however,* this* finding* did* not* remain*

significant*following*correction*for*multiple*testing.*

puncorrected = 0.008 pcorrected = 0.08

93

Table!3.4*Haplotype!Analysis!of!GRM2!Variants!and!Response!to!Clozapine*

* rs4067Vrs2518461**

* R/NR*n*(%)*

p1110.711a*

ΔBPRS*n*(%)*

p1110.719a*

ΔBPOS*n*(%)*

p1110.190a*

ΔBNEG*n*(%)*

p1110.133a*

AVG*

*

GVA*

*

GVG*

27*(15.0)/30*(18.1)*

0.441*

12(6.7)/12(7.2)*

0.837*

141(78.3)/124(74.7)*

0.425*

28*(16.3)*

0.695*

10*(5.8)*

0.506*

134*(77.9)*

0.456*

27*(15.9)*

0.132*

10*(5.9)*

0.236*

133*(78.2)*

0.517*

27*(15.7)*

0.567*

10*(5.8)*

0.055*

135*(78.5)*

0.541*

*Haplotypes*with*frequency*<0.05*were*excluded;*auncorrected*pVvalues*from*UNPHASED*version*3.1.5.**

94

Table!3.5*Haplotype!Analysis!of!SLC6A9!Variants!and!Response!to!Clozapine*

* ******rs16831558Vrs12037805Vrs1978195** rs12037805Vrs1978195*†*

* R/NR*n*(%)* ΔBPRS*n*(%)* ΔBNEG*n*(%)* ΔBPOS*n*(%)* ΔBPOS*n*(%)*

* p1110.159a* p1110.020a* p1110.096a* p1110.001a* p1110.757a*

CVCVG*

*

CVTVA*

*

CVTVG*

*

TVTVA*

54/35*(34.2/24.0)*

0.051*

70/67*(44.3/45.9)*

0.781*

16/23*(10.1/15.7)*

0.143*

18/21*(11.4/14.4)*

0.436*

44*(28.6)*

0.066*

71*(46.1)*

0.404*

21*(13.6)*

0.167*

18*(11.7)*

0.016*

45*(29.2)*

0.022*

70*(45.4)*

0.530*

20*(13.0)*

0.551*

19*(12.3)*

0.143*

45*(29.6)*

0.781*

68*(44.7)*

0.079*

20*(13.2)*

0.905*

19*(12.5)*

0.001*

45*(29.6)*

0.614*

68*(44.7)*

0.483*

20*(13.2)*

0.730*

19*(12.5)*

1.000*

*Haplotypes* with* frequency* <0.05* were* excluded;* auncorrected* pVvalues* from* UNPHASED* version* 3.1.5;*†rs16831558*included*as*‘conditioning*marker’*to*twoVmarker*haplotype.*

Taylor DL V. DISCUSSION

95##

Chapter 4 #

DISCUSSION

4.0 OVERVIEW

This thesis work investigated the association of 17 glutamate system gene variants with

clinical response to CLZ. No significant associations for genotype, allele, or haplotype

analyses were observed across all genes following correction for multiple testing. Prior to

correction, several nominally significant associations were observed, the most important of

which was for the glycine transporter 1 gene variant rs16831558 and change in BPRS

positive symptom subscale scores. In this section we discuss main findings, explore their

implications, assess study limitations and conclude with future directions.

4.1 SUMMARY OF FINDINGS

4.1.1 Association of GRIN2B with Clozapine Response

The first chapter of this thesis investigated the association of eight GRIN2B variants with

response to CLZ. To our knowledge, this is the first reported study to investigate associations

among rs7301328, rs1072388, rs3764030, rs12826365, rs2284411, rs1806191 and rs890

with CLZ response, while the rs1806201 variant had previously been investigated in two

other response studies (Hong et al., 2001; Chiu et al., 2003). Variant rs7301328 significantly

deviated from Hardy-Weinberg equilibrium and was therefore excluded from analysis

(pHWE=1.93x10-7). The implications regarding deviation from HWE are discussed in more

detail in Study Limitations Section 4.3.5.

96#

#

For all GRIN2B SNPs investigated, no significant associations were observed for allele,

genotype, or haplotype analyses prior to and after correction for multiple testing. A-allele

carriers of variant rs1072388 responded marginally better to CLZ therapy than GG-

homozygotes, however this finding was not statistically significant (puncorrected=0.067,

pcorrected=0.440) (Chapter 2, Figure 2.3). Our negative association findings for GRIN2B

variant rs1806201 was consistent with previous reports that also found no association

between this variant and CLZ response in two independent samples of Chinese TRS patients

(Hong et al., 2001; Chiu et al., 2003).

4.1.2 Association of Glutamate System Variants with Clozapine Response

The second chapter investigated the association of ten glutamate system gene variants and

response to CLZ. To our knowledge, this is the first reported study to investigate the

association of GRM2 (rs4067, rs2518461), SLC1A2 (rs4354668, rs4534557, rs2905134),

SLC6A9 (rs12037805, rs1978195, rs16831558), GRIA1 (2195450) and GAD1 (rs3709054)

with clinical response to CLZ. No significant genotype, allele, or haplotype associations

were reported across all SNPs following correction for multiple testing.

Prior to correction, several nominally significant associations were observed. Briefly, a

greater reduction in BPRS negative symptom subscale scores was observed for five variants:

SLC1A2 rs4534557 homozygotes (GG and CC) (puncorrected=0.015, pcorrected=0.140); GRIA1

rs2195450 A-allele carriers (puncorrected=0.017, pcorrected=0.162), SLC1A2 rs4354668 TT-

homozygotes (puncorrected=0.026, pcorrected=0.243), GRM2 rs2518461 GG-homozygotes

(puncorrected=0.037, pcorrected=0.346), and SLC1A2 rs2901534 CC-homozygotes

(puncorrected=0.047, pcorrected=0.440).

97#

#

The most interesting association observed before correction was for SLC6A9 rs16831558: C-

allele carriers experienced an allele dose-dependent reduction in BPOS subscale scores

following six months of CLZ therapy (puncorrected=0.008, pcorrected=0.08) (Chapter 3, Figure

3.4). More specifically, individuals carrying two copies of the C-allele showed a 4.25%

decrease in severity of positive symptoms, those carrying one copy experienced no symptom

change, and individuals carrying two copies of the T-allele experienced a 3.00% increase in

positive symptoms compared to baseline. The rs16831558 SNP is a previously unstudied

variant that was identified using HaploReg functional annotation database (Benson et al.,

2013). Located in the promoter region of SLC6A9, this variant lies in an enhancer/promoter-

associated histone mark and is proposed to affect both SLC6A9 transcriptional activity and

protein expression by altering transcription factor binding. This variant’s functional potential

and biological plausibility, in combination with the nominally significant association finding

reported herein makes rs16831558 an ideal candidate to be further investigated in larger CLZ

response samples and for characterization using functional assays.

4.2 IMPLICATIONS OF FINDINGS

The main findings of this work suggest that genetic variation in the glycine transporter 1

gene may confer risk for clozapine non-response. This section begins with a discussion on

the biological significance of the glycine neurotransmitter system in relation to SCZ etiology

and treatment. CLZ’s ability to interact with the glycine system will then be noted. This

section concludes by exploring a possible mechanism through which the SLC6A9 promoter

variant rs16831558 may regulate glycine transporter expression, and lead to differential drug

effects.

98#

#

4.2.1 Glycine in Schizophrenia Etiology and Treatment

Glycine is a necessary co-agonist at the NR1 subunit of the NMDAR. Through a coincidence

detection type mechanism, the NMDAR will not fire without the simultaneous binding of

both glycine and glutamate (Johnson & Ascher, 1987; Mayer et al., 1989; Thomson, 1990).

Therefore, control of glycine levels at glutamatergic synapses is an important regulatory

component for the activation of excitatory neurotransmission. As was mentioned in the

introduction, the glycine transporter 1 protein is localized to glutamatergic neurons and

controls the reuptake of glycine from the synapse (Kleckner & Dingledine, 1988; Bergeron et

al., 1998). Changes in GlyT1 activity alter NMDAR-mediated neurotransmission as

evidenced by increased NMDAR excitatory post-synaptic currents following GlyT1

antagonism (Bergeron et al., 1998) and enhanced NMDAR function in GlyT1 knockout mice

(Gabernet et al., 2005).

Because glycine is necessary for NMDAR activation, abnormalities in the glycine system

may contribute to the hypoglutamatergic states implicated in SCZ etiology. Several

abnormalities related to glycine biology have been reported in patients with SCZ. In

comparison to healthy controls, AP naïve patients have decreased plasma levels of glycine

and low glycine levels have been shown to correlate with more severe negative symptoms

(Sumiyoshi et al., 2004; Neeman et al., 2005). Binding assays with 3H-glycine in

postmortem brain tissue also show patients experience increased NR1 glycine site binding in

sensory and motor cortices (Ishimaru et al., 1992), and may indicate postsynaptic

compensation for glutamatergic hypofunction.

Since glycine abnormalities may contribute to the hypoglutamatergic states that are

implicated in SCZ’s etiology, glycine has been targeted as a plausible mechanism for novel

99#

#

treatments. Glycine was also recognized as having antipsychotic potential upon the discovery

that pretreatment with glycine or glycine-like compounds reverse PCP-induced hyperactivity

in preclinical models (Javitt et al., 1997). The glycine-modulating compounds that exist at

present include the glycine transporter inhibitors, such as bitopertin (RG1678) developed by

Roche, as well as the use of adjunct glycine or glycine-like compounds for the treatment of

persistent negative symptoms. Glycine-modulating compounds share a common mechanism

of action to achieve therapeutic effect: glycine transporter inhibitors antagonize GlyT1

function to restore normal levels of glycine at the synapse, while glycine compounds directly

stimulate NMDAR function.

4.2.2 Interaction of Clozapine with the Glycine System

CLZ is theorized to achieve therapeutic effect, in part, by acting on the glycine system.

Scientific evidence supporting this theory is as follows: 1) CLZ treatment significantly

restores plasma glycine to levels seen in healthy controls (Neeman et al., 2005; Yamamori et

al., 2014); 2) baseline glycine levels have the ability to predict negative symptom response

during CLZ treatment (Sumiyoshi et al., 2004); and 3) glycine/glycine-like compounds have

no effect or worsen patient symptoms when combined specifically with CLZ (Goff et al.,

1999; Tsai et al., 1999; Evins et al., 2000). The mechanism through which CLZ alters glycine

levels is largely unknown, however, one study has suggested that CLZ may do so by

interfering with glycine reuptake from neuronal synapses (Javitt et al., 2005).

Alternative mechanisms have been postulated to explain how glycine compounds interfere

with CLZ’s therapeutic effect. One group of researchers propose that CLZ may directly

interact with the NMDAR by binding to the NR1 glycine site (Schwieler et al., 2004;

Schwieler et al., 2008), however, no direct evidence supporting this theory currently exists,

100#

#

for instance, from co-immunoprecipitation assays. An alternative hypothesis suggests that

CLZ may depend on NR1 glycine site availability to achieve therapeutic effect (Schwieler &

Erhardt, 2003). Despite the mechanism, these findings suggest that response to CLZ depends

in part on plasma glycine levels and/or availability of the NR1 glycine site.

4.2.3 Promoter Variant Confers Differential Response to Clozapine

We observed that individuals carrying two copies of the rs16831558 C-allele showed a

4.25% improvement in positive symptom severity, those carrying one copy experienced no

symptom change, and patients with two copies of the T-allele experienced a 3.00%

worsening of their positive symptoms from baseline. The rs16831558 promoter variant is

proposed to alter GlyT1 expression through altering transcription factor binding. Future work

entails conducting luciferase promoter assays to test if the C-allele has greater transcriptional

activity over the T-allele. Functional characterization of this SLC6A9 promoter variant is

discussed in more detail in Future Directions Section 4.4.1.

If rs16831558 is found to have an affect on GlyT1 expression, we propose that variation in

rs16831558 may lead to differential response to CLZ. This proposition stems from previous

research indicating that CLZ response is contingent upon glycine levels: baseline levels of

glycine inversely correlate with negative symptom response during CLZ treatment

(Sumiyoshi et al., 2004) and increasing levels of NR1 agonists through the addition of

exogenous glycine/glycine-like compounds during CLZ treatment leads to lack of response

or worsening of symptoms (Goff et al., 1999; Tsai et al., 1999; Evins et al., 2000). Therefore,

variants with the ability to alter the quantity or quality of glycine at neuronal synapses may

also affect CLZ efficacy. We now propose a mechanism through which the glycine

transporter 1 gene promoter polymorphism rs16831558 may regulate GlyT1 expression, and

101#

#

subsequently lead to differential response to CLZ. The mechanism is as follows: if the C-

allele is found to increase SLC6A9 transcriptional activity, individuals carrying the CC-

genotype should exhibit increased GlyT1 expression, leading to lower levels of synaptic

glycine and a more favourable response to CLZ. In contrast, T-allele carriers should exhibit

normal-to-low GlyT1 expression, resulting in normal-to-high levels of endogenous synaptic

glycine, which may either have no effect or may interfere with CLZ’s ability to achieve

therapeutic effect. Further characterization of the rs16831558 variant will be needed to give

credence to this proposed mechanism.

The clinical implications of predictive biomarkers that are capable of distinguishing

responders from non-responders prior to treatment are far reaching and would serve as an

invaluable tool for optimizing treatment outcomes for patients with SCZ. Several examples

of promoter variants conferring risk for non-response exist in other areas of medical research

and provide hope for the clinical application of predictive variants in psychiatric care. For

instance, the vitamin K epoxide reductase complex subunit 1 gene (VKORC1) promoter

variant -1639G>A significantly correlates with Warfarin dose (Carlquist et al., 2008). A

luciferase promoter assay was used to show the G-allele of this variant is associated with a

50% increase in VKORC1 transcriptional activity as compared to the A-allele (Yuan et al.,

2005). One additional example demonstrating an interaction between gene promoter

sequence variants and drug treatment response is in asthma (Drazen et al., 1999).

Recent literature has confirmed that polymorphisms in drug targets have the ability to alter

patient sensitivity to treatment and change the pharmacodynamics of treatment response.

Continued research in pharmacogenetics that aims to increase our understanding of the

complex interplay between genetic variants that control the pharmacokinetic and

102#

#

pharmacodynamic factors involved in drug effects is an important step on the road to

personalizing treatment. An overview of the future of personalized medicine in psychiatry is

discussed in more detail in Personalized Medicine in Psychiatry Section 4.4.5.

4.3 LIMITATIONS AND CONSIDERATIONS

Several limitations associated with our study must be considered before interpreting our

findings. This section explores the importance of study heterogeneity, inter-study variability,

statistical power, correction for multiple testing, deviation from Hardy-Weinberg equilibrium

and incomplete outcome data, as they relate to our findings.

4.3.1 Study Heterogeneity

Study heterogeneity refers to the variability inherent in a study and can be caused by

differences in study design (methodological diversity), and variation among participants,

interventions and outcomes (clinical diversity) (Hanson & Levin, 2013). Heterogeneity in

association studies is particularly problematic because it may lead to spurious associations or

the inability to detect true associations between marker loci and phenotypes of interest. Steps

are often taken to limit the heterogeneity in a sample so that results may be interpreted with

the least amount of confounding.

The first source of heterogeneity in our sample was caused by methodological and clinical

diversity. Participants were collected from three clinical sites that may have differed in study

methodology and populations. Combining individuals with different ancestral backgrounds

into a single group for analysis can lead to population stratification where false positive

associations arise because of population substructure rather than true associations (Cardon &

Palmer, 2003). Ad-mixed populations particularly confound drug response phenotypes

103#

#

because response tends to vary according to a patient’s ethnicity (Frackiewicz et al., 1997;

Wilson et al., 2001).

Patients included in our study were considered to be European based on self-reports. Even

though the accuracy between self-reported ancestry and actual ancestry is fairly consistent

(agreement rate of 75.1%) (Choudhry et al., 2007; Lee et al., 2010), the use of a principle

components analysis to experimentally determine ancestry profiles remains a priority for

future work. Briefly, PCA utilizes a panel of SNPs that are known to differ in allele

frequencies across populations of different geographical regions. Ancestry is then determined

by comparing genotyping results to reference panels from well-characterized datasets such as

genetic data from the HapMap Project (Paschou et al., 2008). PCA analysis is also useful for

detecting population stratification (Liu et al., 2011). Ideally, genetic data from admixed

populations should be stratified prior to analysis to maintain as much homogeneity as

possible.

Heterogeneity related to disease diagnosis and differences between SCZ and SA disorder

may also have increased variation in our sample. For instance, these disorders have different

clinical presentations: SA disorder is characterized as periods of psychosis with concurrent

symptoms meeting criteria for a Major Mood Episode, while in comparison, SCZ is

characterized by less frequent/prominent depressed or manic mood states (American

Psychiatric Association, 2013). As well, SA disorder patients tend to experience higher rates

of response and better clinical outcomes than patients with SCZ (Harrow et al., 2000). Taken

together, these differences suggest that SCZ and SA disorder may differ in their underlying

biology (Abrams et al., 2008; American Psychiatric Association, 2013; Cosgrove & Suppes,

2013,). The propensity of SA disorder patients to respond more favourably to treatment may

104#

#

have increased our CLZ response rate, and combining these two disorders into a single

sample for analysis may limit detection of causal variants that contribute to response.

Lastly, our sample consisted of patients who were placed on CLZ for different reasons. CLZ

is a 3rd line treatment for SCZ patients who are diagnosed as having TRS either because they

failed to respond to previous treatments with pharmacotherapy (treatment resistance) or

because they developed intolerable side effects during treatment (treatment intolerance)

(Moore et al., 2007). Patients diagnosed with treatment resistance experience suboptimal

response characterized by persistent psychotic symptoms, while those experiencing

intolerance cease treatment due to intolerable side effects such as TD. Clinical evidence

suggests treatment intolerant and resistant patients respond differently when treated with

CLZ: treatment intolerant patients tend to experience higher rates of response than patients

who are resistant (Claghorn et al., 1987; Owen et al., 1989; Lieberman et al., 1994). In fact,

higher baseline levels of EPS have been shown to correlate with a favourable response to

CLZ in treatment intolerant patients (Pickar et al., 1992; Lieberman et al., 1994). The

implications for combining treatment resistant and intolerant patients into a single group for

association analyses in unknown, and ideally patients should be stratified to maintain as

much homogeneity as possible.

4.3.2 Inter-Study Variability

Inter-study variability limits comparison of results across studies and also complicates the

process of combining data for meta-analysis. Meta-analyses are important because they aim

to reconcile the contribution of common variants to disease risk, when genetic associations

are unclear from individual studies (Lohmueller et al., 2003). To increase comparability of

results across studies, variables such as inclusion criteria, duration of treatment and clinical

105#

#

outcomes should be selected with consideration to readily accepted standards in the

literature.

Many definitions for TRS exist in the literature and these inconsistencies impede

interpretation of results across studies (Suzuki et al., 2011). Studies using less conservative

definitions for TRS are likely to report higher rates of response. This is because the

likelihood of response decreases after each subsequent failed AP drug trial. Herein, we

defined TRS as failure to respond to two or more adequate trials with AP drugs from at least

two different chemical classes (with doses of ≥1000mg chlorpromazine equivalents for 4-6

weeks), and no period of good functioning in the previous five years (Kane et al., 1988). This

definition of TRS is consistent with definitions existing in the literature (Canadian

Psychiatric Association, 2005; Kreyenbuhl et al., 2010; McIlwain et al., 2011; Kennedy et

al., 2014) and accurately reflects CLZ’s current prescribing policy as a 3rd line treatment

(Elkis & Meltzer, 2007; Moore et al., 2007).

The duration of treatment that constitutes an adequate trial with CLZ also varies across

studies (Rosenheck et al., 1999). High inter-individual variability in AP response has been

observed during treatment and response has been reported to occur as soon as one week

(Pickar et al., 1992; Stern et al., 1994) and as late as one year (Meltzer, 1992; Wilson, 1996)

following treatment initiation. In addition, length of CLZ treatment has been found to

correlate with treatment outcomes: approximately 30% of patients respond in the first six

weeks (Kane et al., 1988; Lieberman et al., 2008) and an additional 20% after three months

(Claghorn et al., 1987; Lindstrom, 1988). These findings suggest that studies assessing

response after only one or two months may do so prematurely for a subset of patients. Our

study assessed CLZ response after a six month trial of CLZ to ensure response rates were

106#

#

accurate and accounted for ‘late responders’. Agreement upon the length of time that

constitutes an adequate CLZ trial will help maintain consistency among studies and aid in the

interpretation of results across studies.

The last source of variability in our study stems from differences in the assessment of clinical

outcomes. Various definitions for response exist in the literature; however, most studies

assessing response use change in either the PANSS or the BPRS (reviewed by Suzuki et al.,

2012). The difference between these two scales is negligible and their scores are easily

interchangeable (Overall & Gorham, 1962; Kay et al., 1987). Herein, response was assessed

using change in BPRS scores from baseline following six months of CLZ therapy.

Categorical response was measured using responder/non-responder frequencies and CLZ

responders were classified as those individuals who experienced a ≥20% decrease in BPRS

scores, while continuous response was measured using percent change in scores controlled

for baseline (Kane et al., 1988). This assessment of clinical outcome was in line with similar

studies evaluating CLZ response from the literature (Hong et al., 2001; Chiu et al., 2003).

Despite widespread use, symptom rating scales have been criticized for their inability to

reflect a patient’s personal, social and cognitive functioning – factors that hold greater weight

in regards to how a patient will function in society. To ensure the clinical utility of response

studies, it may be worthwhile to construct a more ‘patient focused’ response assessment

method that takes factors beyond symptom change into consideration (Mortimer, 2007).

4.3.3 Statistical Power

The power of a statistical test is defined as the probability of correctly rejecting the null

hypothesis of ‘no association’ when the null hypothesis is false, or said in another way, the

ability to detect a true effect when it is present. Power is equal to one minus the probability

107#

#

of a type II error (1-β). Type II errors are otherwise known as false negatives and occur when

true effects are not detected even though they are present. The power of an association study

is influenced by a number of factors including sample size, statistical significance criteria (α)

and the magnitude of the desired effect size to be detected (Evans & Purcell, 2012). Power

tends to be highest for studies with large sample sizes, less stringent significance levels and

larger effect sizes.

A common limitation of genetic association studies is insufficient statistical power. Many

pharmacogenetics studies investigating response are underpowered due to limited sample

sizes. Patients placed on CLZ are particularly hard to come by because of the strict

guidelines surrounding its prescribing and the need for mandatory blood monitoring to

protect against the life-threatening adverse effects of CIA (Warnez & Alessi-Severini, 2014).

Averaged between the two manuscripts, our sample of 160 patients with categorical

(responder/non-responder) response data and 88 patients with continuous response data, had

over 80% power to detect an odds ratio (OR) as low as 2.05 and down to 8.5% of the

variance in the quantitative response measure (unmatched case control design: non-responder

frequency=47.8%, MAF=28.5%, α/2=0.05) (Gauderman, 2002; Gauderman & Morrison,

2006).

Similar to the phenotype of SCZ, AP response is a complex trait that follows a polygenic

mode of inheritance. However, unlike risk for SCZ, drugs administered to a patient for

treatment follow a relatively narrow pathway through the body to the brain where they bind

to a limited number of target receptors. AP drugs thereby actively provoke the phenotype of

response by enacting change in the symptoms of the patient. Therefore, drug response

phenotypes should exhibit larger gene effects than seen when studying SCZ diagnosis due to

108#

#

the a priori selection of genes based on the known pharmacologic mechanism of the drug. A

recent example demonstrating this phenomenon is the large effect size of the melanocortin

receptor 4 gene (MC4R) (RR=3.5 or greater, n=139) for predicting antipsychotic-induced

weight gain (Malhotra et al., 2012; Chowdhury et al., 2013). Therefore, our sample of

patients likely had sufficient statistical power to detect the effect sizes of genes contributing

to response.

For other genes with very small effects, the future collection of larger CLZ response samples

will help unravel their contribution to complex phenotypes, such as drug response. A larger

CLZ response sample is currently being collected by the CRESTAR consortium in Europe

with the aim of developing pharmacogenetic biomarkers for treatment response and side

effects (CRESTAR, 2011). To illustrate the effect of sample size on power: a sample size of

373 cases and 392 controls would have sufficient statistical power (80%) to detect an odds

ratio as low as 1.5 (unmatched case-control design, MAF=15%, α/2=0.05) (Gauderman,

2002; Gauderman, Morrison, 2006). Samples of this size, once available, will aid in the

discovery of novel genetic markers with the ability to predict response to CLZ.

4.3.4 Multiple Testing

Genetic studies often test multiple alleles for association with a desired phenotype and

invariably inflate the type I error rate (α), or likelihood of observing a false-positive finding.

To account for multiple comparisons, a more stringent threshold of significance is applied.

The Bonferroni method is a commonly used correction tool that divides the significance

criteria by the number of independent tests that were conducted (k) (Bonferroni, 1936). This

method is considered conservative for correlated tests such as when SNPs are in linkage

disequilibrium, as this method assumes that tests are independent. To account for these

109#

#

correlations, an alternative method was developed by Nyholt that computes a new value for

the number of independent tests that considers linkage among variants (Nyholt, 2004).

For the first data chapter of this thesis, the Nyholt method was used to compute a corrected

significance threshold of 0.008, based on 6.56 independent tests among seven GRIN2B

variants. The Nyholt method was also used for the second data chapter to compute a

corrected significance threshold of 0.005, based on 9.36 independent tests among the ten

glutamate system gene variants. If all tests across the two manuscripts were combined, a

study-wise corrected threshold of 0.003 based on 15.92 tests would be computed. With these

significance threshold adjustments in place, no polymorphism typed in our study would

reach statistical significance.

Our correction criteria considered the number of SNPs to equal to the number of tests being

performed, however, alternative methods for correction do exist. A less stringent method is a

gene-wise correction, in which k is considered to be equal to the number of SNPs present

within a single gene (≥2) (as used in Kalsi et al., 2010). To illustrate, a gene-wise correction

for the three SNPs located in SLC6A9 would result in a corrected significance threshold of

0.019 based on 2.70 tests and would have rendered the association between rs16831558 and

CLZ response statistically significant (puncorrected=0.008). Opinions differ on what level of

stringency should be used for correction and some researchers may consider this gene-wise

correction to be insufficient. On the other hand, a considerably more stringent approach is to

correct for the total number of intragenic SNPs in the human genome. This correction is

analogous to meeting statistical criteria for a genome wide association study, in which p

equal to 10-8 (Risch & Merikangas, 1996). Many believe this correction to be overly stringent

and argue that a genome wide correction would leave the majority of association studies

110#

#

underpowered (Neale & Sham, 2004; Patnala et al., 2013). A final method to consider

corrects for the total number of statistical tests that are performed and in our case, would

include all allele, genotype and haplotype analyses performed for each variant (Lewis &

Knight, 2012).

The Bonferroni and Nyholt methods of correction are considered family-wise error rate

(FWER) procedures that seek to reduce the probability of observing even one false

discovery. FWER correction methods are disadvantageous for studies in which many tests

are performed because the significance threshold becomes increasingly small and often result

in insufficient statistical power to detect true effects (Perneger, 1998). Because the

consequences of false discoveries in complex traits like AP response are less severe than for

single gene disorders (van den Oord & Sullivan, 2003), less conservative methods of

correction may be applied with caution. A more recently developed less conservative method

is the false discovery rate (FDR) approach. Like the Bonferroni and Nyholt methods, FDR

also determines adjusted p-values for each test (q-values), however, does so by controlling

for the number of false positive discoveries in tests for which the null hypothesis was

rejected as opposed to taking all tests into consideration. This method results in greater

statistical power to find true effects while controlling for the type 1 error rate (Benjamini &

Hochberg, 1995).

4.3.5 Deviation from Hardy-Weinberg Equilibrium

Hardy-Weinberg equilibrium is a quality-control measure used to ensure observed

genotyping frequencies in a study population coincide with those expected under equilibrium

conditions where alleles segregate randomly (Hardy, 1908; Weinberg, 1908). HWE is

calculated using χ2-test where observed genotype frequencies in the study population are

111#

#

compared to those expected under HWE (Weir, 1996). One variant in our sample, GRIN2B

rs7301328, significantly deviated from HWE. HWE threshold values were set at the

multiple-testing adjusted significance levels calculated for each study using the Nyholt

method. This SNP was removed from subsequent analyses because chi-square tests require

SNPs to be in HWE (Sasieni, 1997).

Several reasons may have accounted for why this variant deviated from HWE such as:

genotyping errors that mistype heterozygotes as homozygotes or vice versa (Gomes et al.,

1999; Hosking et al., 2004), stochastic variation, or due to the underlying biological

characteristics of the study population (Nielsen et al., 1998). Deviations may also be a

symptom of disease association (Balding, 2006).

4.3.6 Incomplete Outcome Data

Incomplete outcome data is generated when a patient discontinues treatment and is usually as

result of noncompliance, lack of efficacy, the occurrence of adverse effects, or because the

patient was lost to follow-up. Patients who leave a study prematurely contribute to inaccurate

estimations of response rates in response studies and confound the ability of researchers to

identify genetic markers that may have predictive potential. Noncompliance is particularly

common among patients with SCZ and has been linked to relapse, re-hospitalization and

poor outcome (Centorrino et al., 2001). Paranoia, cognitive dysfunction and bad judgment

are potential reasons to explain why patients may not adhere to their medication regimens

(Lacro et al., 2002). Fortunately, routine blood monitoring for patients taking CLZ allows

treating physicians to both ascertain compliance and ensure CLZ serum levels have reached

optimal threshold that is necessary to achieve response (≥450 ng/mL) (Lindenmayer &

Apergi, 1996).

112#

#

Discontinuation due to lack of efficacy and adverse reactions also similarly effect response

rates. Lack of efficacy to a medication may indicate that drug levels are too low, while

toxicity and adverse effects suggest drug levels are too high, and in both cases a dose

adjustment may be necessary. As would be expected for a phenotype that has high inter-

individual variability, optimal doses for achieving maximum therapeutic effect vary greatly

among patients. Doses within the therapeutic window for the majority of patients may be

inadequate or excessive for a subset of patients and may lead to undesirable treatment

outcomes (Ma & Lu, 2011). The high rates of discontinuation in AP drug trails stresses the

importance of understanding this inter-individual variability in drug response phenotypes and

for identifying predictive markers that may have clinical utility.

4.4 FUTURE DIRECTIONS

This section explores future research initiatives assuming time and resources were unlimited.

The first initiative would characterize the potential functionality of the glycine transporter 1

gene variant rs16831558 using a luciferase promoter assay. The second initiative would

investigate the role of gene-gene and gene-environment interactions in drug response

phenotypes. Lastly, the third initiative would investigate the contribution of novel rare

variants in drug response phenotypes using next generation sequencing (NGS) platforms.

This section concludes with a look at the emerging field of personalized medicine in

psychiatry.

4.4.1 Characterization of Glycine Transporter 1 Promoter Variant

The GlyT1 promoter variant rs16831558 emerged as the highest priority SNP from our

study. As mentioned previously, CC-homozygotes exhibited a notable improvement in their

113#

#

positive symptom scores compared to T allele carriers following six months of CLZ therapy

(puncorrected=0.008, pcorrected=0.08, assuming 9.36 independent tests). This novel variant is

located approximately 5kb upstream of the SLC6A9 gene and is proposed to be functional

according to data generated by ENCODE (ENCODE Project Consortium, 2012).

Four functional characteristics have been cited for this variant: 1) rs16831558 is localized to

a region on chromosome 1 where GATA-binding factor 1 (GATA1) binds to the DNA (Hu et

al., 2011). GATA1, otherwise known as erythroid transcription factor, has been shown to

play a role in cell growth and cancer (Ferreira et al., 2005); 2) rs16831558 is localized to a T-

box element DNA-motif where T-box transcription factor 18 (TBX18) binds to the DNA

(Matys et al., 2006; Pique-Regi et al., 2011). TBX18 has been shown to interact with

GATA4 and other transcription factors in gene promoter regions to modulate transcriptional

activity (Farin et al., 2007); 3) rs16831558 is found within a DNAse hypersensitivity site in

13 different cell lines from ENCODE, including a medulloblastoma neuron precursor cell

line (Medullo_D341) and cerebellum tissue from Caucasian donors (Cerebellum_OC)

(ENCODE Project Consortium, 2012). DNAse-sequencing methods map DNAse

hypersensitivity sites to locate regions of open chromatin where gene regulatory elements

such as promoters, enhancers and silencers are likely to reside (Song & Crawford, 2010); and

lastly, 4) rs16831558 is localized to a region of DNA designated as ‘enhancer’ in tissues

from various brain regions (Bernstein et al., 2010; Ernst & Kellis, 2012). Collectively, this

evidence provides sufficient rationale to investigate rs18631558’s potential to regulate

transcription using, for example, a luciferase promoter assay.

Luciferase promoter assays allow researchers to study changes in mammalian gene

expression in vitro by measuring the strength of a luminescent signal that is created when a

114#

#

reporter gene that encodes the luciferase protein is transcribed (Fan & Wood, 2007).

Conducting a luciferase promoter assay for rs16831558 would first involve designing two

promoter DNA fragments: one containing the C-allele and the other containing the T-allele.

These fragments would then be cloned into two different luciferase reporter vectors upstream

from the luciferase complementary DNA (cDNA), which has had its promoter elements

removed. These vectors would then be transfected into a neuronal cell-line to assess the

differential ability of each promoter variant to drive expression of the luciferase protein.

Expression levels are then measured by comparing the luminescent signal generated by each

rs16831558 promoter construct to basal levels of luminescent intensity generated by

promoter-less vector controls (Solberg & Krauss, 2013).

Certain aspects of the study methodology must be considered before designing a luciferase

promoter assay for rs16831558. For instance, the cell-line chosen for the in vitro experiment

is of particular importance: selecting a cell-line that mimics or is closest to the tissue of

interest will yield results that more accurately represent that variant’s function in vivo.

Neuroblastoma (NB) cell lines derived from neural crest cells are often used in neurobiology

studies (Shastry et al., 2001). Because luciferase promoter assays are performed in vitro, they

do not account for certain cellular and environmental factors that are present in vivo.

Furthermore, sites relatively far away from the gene of interest may in reality have the ability

to effect gene expression. This is possible due to looping and folding of the longer stretches

of DNA in its native state, which is a 3D chromatin structure. Therefore, the results obtained

from in vitro studies may only partially reflect the actual ability of the cloned promoter DNA

construct to regulate gene expression in vivo. Consequently, additional studies to further

115#

#

characterize this GlyT1 variant would be necessary. If this variant is found to be functional,

further characterization in transgenic mice, for instance, may be indicated.

4.4.2 Gene-Environment Interactions

Gene-environment interactions occur when the effect of an individual’s genotype on a

particular phenotypic outcome depends on the level of exposure to an environmental factor

(Clayton & McKeigue, 2001). The contribution of gene-environment interactions to risk

phenotypes is often overlooked for reasons such as lack of environmental data or bias in

estimating the effect of exposure (Arranz & Munro, 2011). Gene-environment interactions

also involve additional tests that invariably inflate the type I error rate (Peduzzi et al., 1995;

Peduzzi et al., 1996). Despite these limitations, the combined effect of genetic and

environmental factors such as cigarette smoking, caffeine consumption and concomitant

medication use have been shown to contribute to variation in drug response phenotypes (de

Leon, 2004; Caspi & Moffitt, 2006; Arranz & Munro, 2011). Therefore, future work

characterizing gene-environment interactions in CLZ response may be useful for

personalizing CLZ treatment and developing targeted interventions for patients at high risk

of non-response and adverse side effects.

The first gene-environment interaction worth exploring in CLZ response investigates the

joint effect of cigarette smoking and variation in the GRIN2B gene. The rationale for

investigating this interaction is as follows: 1) genetic variation in GRIN2B is associated with

early onset cigarette smoking and smoking initiation (Vink et al., 2009; Grucza et al., 2010);

2) nicotine administration in rats increases GRIN2B expression in particular brain regions

(Wang et al., 2007); and 3) by-products that are generated from smoking tobacco induce

CLZ metabolism and result in lower CLZ serum levels in patients who smoke versus those

116#

#

who do not (Seppala et al., 1999; Meyer, 2001; Palego et al., 2002). Patients who carry a

particular GRIN2B genotype that predisposes them to smoking behaviours may be more

likely to experience sub-optimal CLZ response due to sub-therapeutic serum levels and/or

alteration of NMDAR subunit expression. Identification of a gene-environment interaction

between cigarette smoking and high-risk genotypes in GRIN2B may have clinical

applications. Should an interaction be identified, for instance, clinicians may recommend that

patients with risk genotypes reduce their cigarette smoking behaviours, or clinicians may

prescribe higher CLZ dosages to patients who smoke.

Caffeine consumption may also interact with glutamate system genes and alter response to

CLZ. The following observations support investigating this interaction: 1) caffeine

consumption enhances glutamate release and excitatory neurotransmission in particular brain

regions by binding to adenosine receptors on glutamatergic neurons (Dunwiddie, 1980;

Dunwiddie et al., 1981; Solinas et al., 2002; Salmi et al., 2005); and 2) during acute

administration, caffeine has been shown to inhibit clozapine metabolism and lead to higher

CLZ serum levels in patients who consume caffeine versus patients who do not (Hagg et al.,

2000; de Leon, 2004). Therefore, higher serum CLZ levels in patients who drink coffee, in

combination with caffeine’s ability to mediate glutamate signaling may have multiplicative

effects on drug response and may increase the risk of developing adverse side effects. Akin

to the first interaction, understanding the relationship between caffeine intake and CLZ

response may have clinical applications. Following Hagg et al., clinicians may initially

prescribe lower CLZ dosages to patients who consume high amounts of caffeinated

beverages, or recommend that patients with risk genotypes either limit caffeine consumption

117#

#

or make attempts to maintain consistency in the amount of caffeine consumed per day (Hagg

et al., 2000).

4.4.3 Gene-Gene Interactions

Gene-gene interaction, or epistasis, is the dynamic interaction among different genetic

variants that occurs when one allele modifies the effect of alleles at other loci, or when two

or more loci interact to create a new phenotype (Phillips, 2008). Epistatic interactions are

thought to account for a portion of the inter-individual variability that is observed in

polygenic traits, such as AP response (Long & Langley, 1999; Sadee, 2012; Zuk et al.,

2012). Treatment outcomes likely depend on the multiplicative effect of several genes that

are involved in drug action, such as enzymes involved in metabolism, membrane

transporters, receptors and target proteins (Sadee, 2013). However, compared to the large

number of genes involved in disease phenotypes such as SCZ, CLZ response likely depends

on a lesser number of genes to determine treatment outcomes as evidenced by

pharmacogenetics studies (Johnson et al., 2011; Ma & Lu, 2011). Therefore, effective

biomarker panels requiring only a limited number of genes may be able to capture much of

the genetic variation contributing to drug response (Sadee, 2013). The clinical utility of such

predictive tests provides sufficient rationale for the investigation of three potential epistatic

interactions involving glutamate genes with response to CLZ.

The first gene-gene interaction worth exploring in CLZ response involves two genes that

were involved in this study: the NMDAR NR2B subunit gene GRIN2B, and the GlyT1 gene

SLC6A9. Three observations provide support for investigating this gene-gene interaction: 1)

Differential expression of either NR2B and GlyT1 are capable of altering NMDAR

activation (Flint et al., 1997); 2) NR2B and GlyT1 co-localize to glutamatergic synapses and

118#

#

may physically interact (Cubelos et al., 2005); and 3) NMDAR activation is thought to

account for a portion of CLZ’s therapeutic effects (Banerjee et al., 1995; Arvanov et al.,

1997; Kubota et al., 2000; Ninan et al., 2003; Kargieman et al., 2007). Collectively, these

findings suggest that differential expression of NR2B and GlyT1 proteins by potentially

functional genetic variants may interact to augment NMDAR neurotransmission, and

subsequently, response to CLZ. A proposed gene-gene interaction analysis would investigate

the putatively functional SLC6A9 variant rs16831558 that was genotyped in this study, and

the GRIN2B variant rs1805502 that was previously associated with negative symptom

change during CLZ therapy (Martucci & Kennedy, 2010).

An epistatic interaction may also exist for the mGluR2 gene GRM2 that was investigated in

this study, and the 5-HT2A gene, HTR2A. The rationale for investigating this interaction is as

follows: 1) In particular brain regions, translation of the 5-HT2A receptor is necessary for

normal mGluR2 expression (Gonzalez-Maeso et al., 2008); 2) mGluR2 has been shown to

form a special hetero-complex with 5-HT2A through specific transmembrane helix domains

(Gonzalez-Maeso et al., 2008); 3) CLZ binds to the 5-HT2A component of this complex and

up-modulates mGluR2-elicited signaling by 40% (Fribourg et al., 2011); and 4) The 5-

HT2A-dependent antipsychotic-like behavioural effects of CLZ require the expression of

mGluR2 (Fribourg et al., 2011). Taken together, these findings suggest that CLZ may act on

the mGluR2-5-HT2A complex to achieve symptom change in SCZ patients and that altered

expression of both HTR2A and GRM2 may have an additive affect on response. Of high

priority is the functional HTR2A variant rs6314 previously associated with CLZ response

(Nothen et al., 1995; Masellis et al., 1995; Burnet & Harrison, 1995; Arranz et al., 1995) and

the potentially functional GRM2 SNP rs2518461 identified in this study.

119#

#

The final interaction worth considering for future work involves GRIN2B and the 5-HT1A

gene, HTR1A. Support for this interaction is as follows: 1) CLZ is a partial agonist at the 5-

HT1A receptor (Newman-Tancredi et al., 1996); 2) CLZ facilitates formation of a synergistic

triad between 5-HT1A, the NMDAR, and CaMKII (Purkayastha et al., 2012); and 3) This

triad is necessary for CLZ-mediated increases in EPSPs in PFC pyramidal neurons

(Purkayastha et al., 2012). Genetic variation in both HTR1A and GRIN2B may have the

potential to alter triad formation and, as a result, alter CLZ’s ability to elicit excitability in

certain brain regions. The functional HTR1A promoter variant rs6295 previously implicated

in depression and antidepressant response (Lemonde et al., 2003; Lemonde et al., 2004;

Parsey et al., 2006) is of high interest and deserves investigation with GRIN2B SNPs of

interest.

There are a number of challenges associated with analysing gene-gene and gene-environment

interactions in drug response phenotypes. One challenge of modelling such relationships is

the increasing dimensionality that results from a large number of possible interactions

(Moore, 2004; Motsinger et al., 2007). As a result, the application of traditional parametric

tests to analyse interaction data has limited use and often increases the risk of type I and type

II errors, while at the same time decreasing power (Moore, 2004). For these reasons, more

advanced statistical approaches have been developed such as the multifactor dimensionality

reduction (MDR) method (Ritchie et al., 2001). MDR reduces dimensionality by dividing

high-risk and low-risk interactions into two separate groups for analysis, and then selects the

model(s) best able to accurately predict response/non-response status in independent testing

sets generated through cross-validation. Models with the highest cross-validation consistency

are used in permutation testing to identify models with statistically significant results not

120#

#

expected by chance alone. Often the most challenging aspect of interaction analysis involves

interpreting the biological plausibility of the results, and follow-up studies using cell-culture

or other in vitro experiments can be useful aids to interpretation (Ritchie & Motsinger, 2005).

4.4.4 Next Generation Sequencing

Next generation sequencing (NGS) is a high-throughput DNA sequencing technology that

has revolutionized the way scientists extract genomic data from biological systems. NGS

utilizes a similar concept to Sanger sequencing whereby bases in a small fragment of DNA

are sequentially identified from fluorescent signals emitted as each fragment is resynthesized

(Sanger et al., 1977). However, NGS carries out this process on a grander scale by

sequencing millions of DNA fragments at once and results in a quicker, less expensive

method for sequencing whole genomes (Shendure & Ji, 2008).

One particular application of NGS that is of interest to disease risk and medication response

is targeted sequencing, wherein a subset of genes is sequenced, as opposed to the whole

genome. Targeted sequencing allows researchers to achieve high sequence coverage, obtain

sequencing results for many individuals, and makes it possible to identify rare variants that

were missed using previous methods. Rare, in this case, refers to variants that occur less than

once per 100 individuals. Rare variants have given rise to what is known as the ‘common

disease-rare variant’ hypothesis, which argues that multiple different rare mutations in

overlapping genomic locations and/or biochemical pathways drive susceptibility to risk

phenotypes, and are likely to contribute to a portion of the missing heritability in complex

traits (Schork et al., 2009; Altmann et al., 2013).

121#

#

NGS technologies have already identified excess rare variants and deletions in glutamate-

related synaptic genes in SCZ and autism (Kelleher et al., 2012; Myles-Worsley et al., 2013;

Kenny et al., 2014). Regarding drug treatment, NGS has mostly been used in cancer research

to identify rare and common variants in response to chemotherapeutic agents (Ross &

Cronin, 2011; Tran et al., 2013; Ong et al., 2014). A current review of the literature does not

reveal any major discoveries using NGS technologies to identify genetic predictors of

response to AP therapy. However, the practical applications of such discoveries cannot be

understated. Discovery of rare variants and predictive markers would allow clinicians to

distinguish between responders and non-responders and identify patients who are at high risk

of failing treatment. The information derived from NGS may also be used to guide

development of novel drugs that target aberrant gene systems in the non-responder group.

Therefore, future research involving targeted sequencing of glutamate system genes is

recommended to help uncover rare variants involved in CLZ response.

To briefly illustrate the methodology behind NGS, a hypothetical workflow for sequencing a

gene of interest using the Ion Torrent Personal Genome Machine (PGM) Sequencer (Life

Technologies) in our CAMH laboratory is proposed. The four steps are as follows: 1) Library

preparation: The first step is to create a library of DNA fragments that are flanked by Ion

Torrent adapters. Adapters can be added to DNA fragments by one of two ways – ligating

the adapter to the PCR product, or by incorporating the adapter sequence automatically

during PCR using specially designed primers; 2) Template preparation: The second step

involves coating special ‘Ion Sphere’ particles with DNA fragments via their adapter

sequences, followed by clonal amplification of the library fragments by emulsion PCR

(emPCR); 3) Sequencing: The Ion Sphere particles coated with library fragments are then

122#

#

deposited onto the sequencing chip, placed into the Ion Torrent machine, and the sequencing

run is begun, 4) Data Analysis: The final step of the work flow analyses sequencing data.

Data is transferred to the appropriate servers, and then run through signal processing and

base-calling algorithms to re-construct DNA sequences. Sequencing data is then imported

into analysis programs such as the Partek® Genomics SuiteTM. One additional point to

consider during study design is the depth of coverage. Depth refers to the number of times

the region of interest is re-sequenced. Rare variants and somatic mutations that are present at

low frequencies require higher depths of coverage to ensure the mutation calls are made with

sufficient statistical confidence (Life Technologies, 2011).

Challenges associated with NGS technology must also be mentioned. Because this

technology produces vast amounts of data in a relatively short period of time, future research

initiatives will need to focus on how to organize and interpret the sheer volume of genomic

data that is produced. In addition, the ability to detect rare variants depends on the

availability of large sample sizes and stresses the need for a collaborative effort in the

research community. Ideally, a common database will be developed where researchers

investigating AP response phenotypes can upload their DNA samples and/or sequencing

results, with the shared goal of advancing medical research within this field. Ethical issues

also abound and stress the need for legislation to protect citizens from discrimination based

on their genome (Mathieu et al., 2013). Several nations have passed legislation barring

insurance companies and employers from discriminating against citizens based on their DNA

sequence. The United States of America passed the Genetic Information Nondiscrimination

Act of 2008 (GINA) (United States of America, 2008), while the United Kingdom passed the

Equality Act of 2010 (United Kingdom of Great Britain, 2010). Canada passed Bill C-445 in

123#

#

2011, which amended the Canadian Human Rights Act to include protection of Canadian

citizens from discrimination based on their ‘genetic characteristics’ (Parliament of Canada,

2011). Shortly afterwards, Bill S-201 was passed by Canadian legislature, otherwise known

as the Genetic Nondiscrimination Act (Parliament of Canada, 2013), to provide Canadian

citizens with further protection against discrimination based on their DNA. Despite these

limitations, NGS has already revolutionized the field of genetics research and is predicted to

lead to many groundbreaking discoveries in the future.

4.4.5 Personalized Medicine in Psychiatry

Personalized medicine in psychiatry is a relatively new field with the goal of tailoring drug

treatment to suit an individual’s personal genetic makeup. The realization that inter-

individual variability in drug response had a genetic underpinning was a pivotal discovery on

the path to personalized medicine (Garrod, 1909; Beutler et al., 1955; Evans et al., 1960).

Furthermore, launch of the Human Genome Project (Lander et al., 2001) and the

development of more cost-effective genome sequencing methods have made genetic

information more easily accessible to the general population. New studies in

pharmacogenetics that investigate how drugs and genetics interact are also of high

importance for the developing field of personalized medicine.

The field of personalized medicine in psychiatry is evolving and seeing the development of

genetic panels and algorithms used as aids in medication prescribing. A number of

pharmacogenetic tests are available on the market and have been reviewed elsewhere (Arranz

& Munro, 2011). One genetic test worth mentioning is the GeneSight® test developed by

AssureRX Health Inc. This test incorporates genotype data for four different CYP enzymes,

as well as the serotonin transporter 1 (SLC6A4) and HTR2A genes. Studies investigating the

124#

#

utility of the test have reported improved outcomes should the test be used in routine clinical

care (Hall-Flavin et al., 2013).

Unfortunately, a scientifically validated test for CLZ response has not yet emerged.

Approximately 15 years ago, researchers Kerwin and Arranz along with their colleagues

published findings on a genetic panel of six polymorphisms that were capable of predicting

CLZ response with a 76.6% success rate in their sample of 200 Caucasian SCZ patients

(p=0.001) (Arranz et al., 2000). The six variants identified were located in neurotransmitter-

receptor related genes including two HTR2A variants (102T/C and His452Tyr), two serotonin

2C receptor gene (HTR2C) variants (-330-GT/-244-CT and Cys23Ser), the serotonin

transporter promoter (5-HTTLPR), and the histamine H2 receptor gene (HRH2) variant (-

1018-G/A). These six polymorphisms, when combined, had a retrospective sensitivity of

95% for identifying patients with ‘satisfactory response’. This report was the first of its kind

attempting to predict CLZ response using genetic markers. Unfortunately, other authors were

unable to replicate these findings (Arranz et al., 2000; Schumacher et al., 2000). A

commercialized test was developed based on Kerwin and Arranz’s findings by a UK

company known as LGC (formerly known as the Laboratory of the Government Chemist that

was privatized in 1996). Unfortunately, no prospective studies assessing the clinical and

economic implications of this test have been conducted.

Despite the lack of success in developing a predictive test for CLZ response, advancements

in other areas of pharmacogenetics such as the ability to predict drug side effects, offer hope

for the future. Relatively recently, our colleagues at CAMH in Toronto developed a gene

panel consisting of 5 gene markers that is able to explain 27% of the variance associated with

developing antipsychotic induced weight gain (AIWG) (Tiwari et al, unpublished). AIWG is

125#

#

a debilitating side effect of second-generation antipsychotics such as CLZ and OLZ that

frequently results in obesity, metabolic syndrome and premature morbidity. The ability to

predict the likelihood of developing such negative side effects will allow physicians to make

more informed decisions regarding medication choice.

The benefits of personalized medicine to psychiatric care cannot be understated: outcomes

from personalized treatment may allow physicians the ability to define diseases more

precisely and to distinguish subtypes/groups of SCZ patients that are based on a drug’s

limited group specificity. In addition, the increased availability of genome sequencing data

may greater our understanding of the underlying pathophysiological basis of SCZ and lead to

the development of novel treatments that are more effective for treating specific disease

subtypes (reviewed by Insel & Scolnick, 2006). For instance, research has suggested that

patients suffering from TRS may present with a different underlying etiology than patients

who respond to conventional dopamine-blocking antipsychotic medications (Egerton et al.,

2012; de la Fuente-Sandoval et al., 2013; Demjaha et al., 2014). Demarcation between

individuals likely to respond and those who are not at first episode would enable physicians

to select a drug, once developed, that is more specific to this subgroup of patients.

There are several issues surrounding the use of genetic testing in personalized medicine that

must be mentioned. For instance, drug response likely results from a complex interaction

between genes, brain and environmental factors such as a patient’s lifestyle, age, gender,

diet, ethnicity and amount of social support. Therefore, it is imperative that researchers

investigating pharmacogenetic processes sufficiently acknowledge the uncertainties

pertaining to their research and report their findings with caution (Evers, 2009). As well, the

contribution of epigenetic interactions must also be considered regarding nonresponse to

126#

#

medications. Additional concerns include increased drug costs associated with targeted drug

development and an increased risk of pharmacological exclusion due to ethnicity or under-

represented genetic groups. As with all novel areas of research, appropriate infrastructure to

handle challenges associated with using pharmacogenomic drugs in psychiatric care will also

need to be implemented. Additional aspects that will need to be addressed are reliability of

genotyping, equal access to testing, as well as clinician and patient education (Costa e Silva,

2013).

Given the advancements in genetic technology and the interest in developing genetic panels

capable of response prediction, the future of personalized medicine in psychiatry looks

promising. As with all new areas of research, educating physicians and patients, developing

the appropriate infrastructure and anticipating ethical issues will need to be considered as the

field of personalized medicine continues to evolve. To highlight the prospective benefits,

personalized medicine in psychiatry has the potential to guide clinician choice regarding

therapeutic dose, decrease the occurrence of adverse drug reactions, and improve overall

disease prognosis. Results from pharmacogenetics studies and personalized treatment may

also guide development of novel treatments and identify more homogeneous disease

subclasses based on the propensity of certain individuals to respond to select medications.

4.5 CONCLUDING REMARKS

Glutamate signaling pathways are emerging as top candidates in SCZ risk and as targets for

novel therapies for the treatment of SCZ. As the field of psychiatric genetics continues to

expand with the advent of more cost effective methods for analysing genomic data,

personalized medicine in psychiatry is becoming more of a reality.

127#

#

Herein, we investigated the association of 17 glutamate system gene variants and clinical

response to CLZ. We reported no significant associations after correction for multiple

testing. Prior to correction, several nominally significant associations were observed. The

most noteworthy preliminary finding was for the glycine transporter 1 gene variant

rs16831558: patients carrying the C-allele exhibited a marked improvement in positive

symptoms compared to T-allele carriers following six months of CLZ therapy. Our findings

provide further credence to the ‘glutamate hypofunction hypothesis’ of SCZ and support

future research initiatives to investigate the potential functionality of rs16831558 and further

explore this variant in larger CLZ response samples.

Taylor DL V. REFERENCES !

128!!

REFERENCES

Abdul*Monim!Z,!Reynolds!GP!&!Neill!JC.!(2006).!The!effect!of!atypical!and!classical!antipsychotics!on!sub*chronic! PCP*induced! cognitive!deficits! in! a! reversal*learning!paradigm.! Behavioural! Brain!Research,!169(2):!263*273.!!

Abrams! DJ,! Rojas! DC! &! Arciniegas! DB.! (2008).! Is! schizoaffective! disorder! a! distinct! categorical!diagnosis?! A! critical! review! of! the! literature.! Neuropsychiatric! Disease! and! Treatment,! 4(6):!1089*1109.!!

Adams!DH,!Kinon!BJ,!Baygani!S,!Millen!BA,!Velona!I,!Kollack*Walker!S!&!Walling!DP.!(2013).!A!long*term,! phase! 2,! multicenter,! randomized,! open*label,! comparative! safety! study! of!pomaglumetad!methionil! (LY2140023!monohydrate)!versus!atypical!antipsychotic!standard!of!care!in!patients!with!schizophrenia.!BMC!Psychiatry,!13:!143*151.!!

Addington!AM,!Gornick!M,!Duckworth! J,!Sporn!A,!Gogtay!N,!Bobb!A,! .! .! .!Straub!RE.! (2005).!GAD1!(2q31.1),!which! encodes! glutamic! acid! decarboxylase! (GAD67),! is! associated!with! childhood*onset!schizophrenia!and!cortical!gray!matter!volume!loss.!Molecular!Psychiatry,!10(6):!581*588.!!

Aghajanian!GK!&!Marek!GJ.! (2000).!Serotonin!model!of!schizophrenia:!Emerging!role!of!glutamate!mechanisms.!Brain!Research!Reviews,!31(2*3):!302*312.!!

Aleman!A,!Kahn!RS!&!Selten!JP.!(2003).!Sex!differences!in!the!risk!of!schizophrenia:!Evidence!from!meta*analysis.!Archives!of!General!Psychiatry,!60(6):!565*571.!!

Alonso!P,!Gratacos!M,!Segalas!C,!Escaramis!G,!Real!E,!Bayes!M,!.!.!.!Menchon!JM.!(2012).!Association!between! the! NMDA! glutamate! receptor! GRIN2B! gene! and! obsessive*compulsive! disorder.!Journal!of!Psychiatry!&!Neuroscience,!37(4):!273*281.!

Altar!CA,!Wasley!AM,!Neale!RF!&!Stone!GA.!(1986).!Typical!and!atypical!antipsychotic!occupancy!of!D2!and!S2!receptors:!An!autoradiographic!analysis! in!rat!brain.!Brain!Research!Bulletin,!16(4):!517*525.!!

Altar! CA,! Carhart! JM,!Allen! JD,!Hall*Flavin!DK,!Dechairo!BM!&!Winner! JG.! (2015).! Clinical! validity:!Combinatorial! pharmacogenomics! predicts! antidepressant! responses! and! healthcare!utilizations! better! than! single! gene! phenotypes.! The! Pharmacogenomics! Journal,! advanced!online!publication.!!

Altmann!A,!Quast!C!&!Weber!P.!(2013).!Detecting!rare!variants!for!psychiatric!disorders!using!next!generation!sequencing:!A!methods!primer.!Current!Psychiatry!Reports,!15(1):!333*141.!!

American! Psychiatric! Association.! (1987).! Diagnostic! and! statistical! manual! of! mental! disorders!(DSMPIIIPR)!(3rd!ed).!Washington!DC:!American!Psychiatric!Association!(revised).!!

American! Psychiatric! Association.! (1994).! Diagnostic! and! statistical! manual! of! mental! disorders!(DSMPV)!(4th!ed).!Washington!DC:!American!Psychiatric!Association.!!

129!!

!!

American! Psychiatric! Association.! (2013).! Diagnostic! and! statistical! manual! of! mental! disorders!(DSMP5)!(5th!ed).!Arlington,!VA:!American!Psychiatric!Publishing.!!

Andreasen! NC,! Rezai! K,! Alliger! R,! Swayze! VW,! Flaum! M,! Kirchner! P,! .! .! .! O'Leary! DS.! (1992).!Hypofrontality! in! neuroleptic*naive! patients! and! in! patients! with! chronic! schizophrenia.!Assessment!with!xenon!133!single*photon!emission!computed!tomography!and!the!Tower!of!London.!Archives!of!General!Psychiatry,!49(12):!943*958.!!

Andreasen!NC,! Arndt! S,! Swayze! V,! Cizadlo! T,! Flaum!M,!O'Leary! D,! .! .! .! Yuh!WT.! (1994).! Thalamic!abnormalities! in! schizophrenia! visualized! through! magnetic! resonance! image! averaging.!Science,!266(5183):!294*298.!!

Angst! J,! Jaenicke!U,!Padrutt!A!&!Scharfetter!C.! (1971).!Ergebnisse!eines!doppelblindversuches!von!HF! 1854! (8*chlor*11*(4*methyl*1*piperazinyl)*5H*dibenzo! (b,e)! (1,4)diazepin)! im! vergleich! zu!levomepromazin.!Pharrnackopsychiatrie,!4:!192*200.!!

Anson! LC,! Chen! PE,! Wyllie! DJ,! Colquhoun! D! &! Schoepfer! R.! (1998).! Identification! of! amino! acid!residues!of!the!NR2A!subunit!that!control!glutamate!potency!in!recombinant!NR1/NR2A!NMDA!receptors.!The!Journal!of!Neuroscience,!18(2):!581*589.!!

Arango! C,! Breier! A,! McMahon! R,! Carpenter! WT! Jr! &! Buchanan! RW.! (2003).! The! relationship! of!clozapine!and!haloperidol! treatment! response! to!prefrontal,!hippocampal,!and!caudate!brain!volumes.!The!American!Journal!of!Psychiatry,!160(8):!1421*1427.!!

Arranz!M,!Collier!D,!Sodhi!M,!Ball!D,!Roberts!G,!Price!J,! .! .! .!Kerwin!R.!(1995).!Association!between!clozapine!response!and!allelic!variation!in!5*HT2A!receptor!gene.!Lancet,!346(8970):!281*282.!!

Arranz! MJ,! Munro! J,! Birkett! J,! Bolonna! A,! Mancama! D,! Sodhi! M,! .! .! .! Kerwin! RW.! (2000).!Pharmacogenetic!prediction!of!clozapine!response.!Lancet,!355(9215):!1615*1616.!!

Arranz!MJ,!Munro!J,!Osborne!S,!Collier!D!&!Kerwin!RW.!(2000).!Difficulties!in!replication!of!results.!Lancet,!356(9238):!1359*1360.!!

Arranz! MJ! &! Munro! JC.! (2011).! Toward! understanding! genetic! risk! for! differential! antipsychotic!response!in! individuals!with!schizophrenia.!Expert!Review!of!Clinical!Pharmacology,!4(3):!389*405.!!

Arvanov! VL,! Liang! X,! Schwartz! J,! Grossman! S! &! Wang! RY.! (1997).! Clozapine! and! haloperidol!modulate! N*methyl*D*aspartate*! and! non*N*methyl*D*aspartate! receptor*mediated!neurotransmission!in!rat!prefrontal!cortical!neurons!in!vitro.!The!Journal!of!Pharmacology!and!Experimental!Therapeutics,!283(1):!226*234.!!

Asada!H,!Kawamura!Y,!Maruyama!K,!Kume!H,!Ding!RG,!Kanbara!N,!.!.!.!Obata!K.!(1997).!Cleft!palate!and!decreased!brain!gamma*aminobutyric!acid!in!mice!lacking!the!67*kDa!isoform!of!glutamic!acid!decarboxylase.! Proceedings!of! the!National!Academy!of! Sciences!of! the!United! States!of!America,!94(12):!6496*6499.!!

130!!

!!

Asztely! F! &! Gustafsson! B.! (1996).! Ionotropic! glutamate! receptors.! Their! possible! role! in! the!expression!of!hippocampal!synaptic!plasticity.!Molecular!Neurobiology,!12(1):!1*11.!!

Ayalew! M,! Le*Niculescu! H,! Levey! DF,! Jain! N,! Changala! B,! Patel! SD,! .! .! .! Niculescu! AB.! (2012).!Convergent! functional! genomics! of! schizophrenia:! From! comprehensive! understanding! to!genetic!risk!prediction.!Molecular!Psychiatry,!17(9):!887*905.!!

Bakshi!VP,!Swerdlow!NR!&!Geyer!MA.!(1994).!Clozapine!antagonizes!phencyclidine*induced!deficits!in!sensorimotor!gating!of!the!startle!response.!The!Journal!of!Pharmacology!and!Experimental!Therapeutics,!271(2):!787*794.!!

Balding! DJ.! (2006).! A! tutorial! on! statistical! methods! for! population! association! studies.! Nature!Reviews,!Genetics,!7(10):!781*791.!!

Banerjee!SP,!Zuck!LG,!Yablonsky*Alter!E!&!Lidsky!TI.!(1995).!Glutamate!agonist!activity:!Implications!for!antipsychotic!drug!action!and!schizophrenia.!Neuroreport,!6(18):!2500*2504.!!

Barbato! A.! (1998).! Schizophrenia! and! public! health,! Chapter! 4:! Consequences! of! schizophrenia.!Geneva:!World!Heath!Organization.!!

Bar*Peled! O,! Ben*Hur! H,! Biegon! A,! Groner! Y,! Dewhurst! S,! Furuta! A! &! Rothstein! JD.! (1997).!Distribution! of! glutamate! transporter! subtypes! during! human! brain! development.! Journal! of!Neurochemistry,!69(6):!2571*2580.!!

Barrett! JC,! Fry! B,! Maller! J! &! Daly! MJ.! (2005).! Haploview:! Analysis! and! visualization! of! LD! and!haplotype!maps.!Bioinformatics,!21(2):!263*265.!!

Benjamini! Y! &! Hochberg! Y.! (1995).! Controlling! the! false! discovery! rate:! A! practical! and! powerful!approach! to! multiple! testing.! Journal! of! the! Royal! Statistical! Society! Series! B,! Statistical!Methodology,!57B(1):!289*300.!!

Benson! DA,! Cavanaugh! M,! Clark! K,! Karsch*Mizrachi! I,! Lipman! DJ,! Ostell! J! &! Sayers! EW.! (2013).!GenBank.!Nucleic!Acids!Research,!Database!Issue,!41:!D36*D42.!!

Bente!D,!Engelmeier!M,!Heinrich!K,!Schmitt!W!&!Hippius!H.!(1966).!Klinische!untersungen!mit!einem!neuroleptisch!wirksamen!dibenzothiazepin*derivat.!Arzneimittelforschung,!16:!314*316.!!

Bergeron! R,! Meyer! TM,! Coyle! JT! &! Greene! RW.! (1998).! Modulation! of! N*methyl*D*aspartate!receptor!function!by!glycine!transport.!Proceedings!of!the!National!Academy!of!Sciences!of!the!United!States!of!America,!95(26):!15730*15734.!!

Bernstein!BE,!Kamal!M,!Lindblad*Toh!K,!Bekiranov!S,!Bailey!DK,!Huebert!DJ,! .! .! .!Lander!ES.!(2005).!Genomic!maps!and!comparative!analysis!of!histone!modifications! in!human!and!mouse.!Cell,!120(2):!169*181.!!

131!!

!!

Bernstein!BE,!Stamatoyannopoulos!JA,!Costello!JF,!Ren!B,!Milosavljevic!A,!Meissner!A,!.!.!.!Thomson!JA.!(2010).!The!NIH!roadmap!epigenomics!mapping!consortium.!Nature!Biotechnology,!28(10):!1045*1048.!!

Bertilsson!L,!Carrillo! JA,!Dahl!ML,! Llerena!A,!Alm!C,!Bondesson!U,! .! .! .!Benitez! J.! (1994).!Clozapine!disposition! covaries! with! CYP1A2! activity! determined! by! a! caffeine! test.! British! Journal! of!Clinical!Pharmacology,!38(5):!471*473.!!

Beutler!E,!Dern!RJ!&!Alving!AS.! (1955).!The!hemolytic!effect!of!primaquine.!VI.!An! in!vitro! test! for!sensitivity! of! erythrocytes! to! primaquine.! The! Journal! of! Laboratory! and! Clinical! Medicine,!45(1):!40*50.!!

Black!IB.!(1999).!Trophic!regulation!of!synaptic!plasticity.!Journal!of!Neurobiology,!41(1):!108*118.!!

Bleehen!T.!(1993).!Clozapine:!Literature!review.!Basel,!Switzerland:!Sandoz!Pharma.!!

Bleuer!E.!(1911).!Dementia!praecox!oder!gruppe!der!schizophrenien!(dementia!praecox!or!the!group!of!schizophrenias).!Leipzig,!Germany:!Franz!Deuticke.!!

Bobes! J,! Garcia*Portilla! MP,! Bascaran! MT,! Saiz! PA! &! Bousono! M.! (2007).! Quality! of! life! in!schizophrenic!patients.!Dialogues!in!Clinical!Neuroscience,!9(2):!215*226.!!

Bonferroni! C.! (1936).! Teoria! statistica! delle! classi! e! calcolo! delle! probabilità.! Pubblicazioni! Del! R!Istituto!Superiore!Di!Scienze!Economiche!e!Commerciali!Di!Firenze,!8:!3*62.!!

Borgio!JG,!Bressan!RA,!Barbosa!Neto!JB!&!Daltio!CS.! (2007).!Refractory!schizophrenia:!A!neglected!clinical!problem.!Revista!Brasileira!De!Psiquiatria,!29(3):!292*293.!!

Boydell!J,!Dean!K,!Dutta!R,!Giouroukou!E,!Fearon!P!&!Murray!R.!(2007).!A!comparison!of!symptoms!and! family!history! in! schizophrenia!with!and!without!prior! cannabis!use:! Implications! for! the!concept!of!cannabis!psychosis.!Schizophrenia!Research,!93(1*3):!203*210.!!

Boyle! AP,! Hong! EL,! Hariharan! M,! Cheng! Y,! Schaub! MA,! Kasowski! M,! .! .! .! Snyder! M.! (2012).!Annotation!of!functional!variation!in!personal!genomes!using!RegulomeDB.!Genome!Research,!22(9):!1790*1797.!!

Braff!DL,!Grillon!C!&!Geyer!MA.!(1992).!Gating!and!habituation!of!the!startle!reflex!in!schizophrenic!patients.!Archives!of!General!Psychiatry,!49(3):!206*215.!!

Brown!AS,! Begg!MD,! Gravenstein! S,! Schaefer! CA,!Wyatt! RJ,! Bresnahan!M,! .! .! .! Susser! ES.! (2004).!Serologic!evidence!of!prenatal! influenza! in! the!etiology!of! schizophrenia.!Archives!of!General!Psychiatry,!61(8):!774*780.!!

Brzustowicz! LM,!Hodgkinson! KA,! Chow! EW,!Honer!WG!&!Bassett! AS.! (2000).! Location! of! a!major!susceptibility! locus! for! familial! schizophrenia! on! chromosome! 1q21*q22.! Science,! 288(5466):!678*682.!!

132!!

!!

Bu! DF,! Erlander!MG,! Hitz! BC,! Tillakaratne!NJ,! Kaufman!DL,!Wagner*McPherson! CB,! .! .! .! Tobin! AJ.!(1992).!Two!human!glutamate!decarboxylases,!65*kDa!GAD!and!67*kDa!GAD,!are!each!encoded!by! a! single! gene.! Proceedings! of! the! National! Academy! of! Sciences! of! the! United! States! of!America,!89(6):!2115*2119.!!

Bubenikova! V,! Votava! M,! Horacek! J,! Palenicek! T! &! Dockery! C.! (2005).! The! effect! of! zotepine,!risperidone,! clozapine! and! olanzapine! on! MK*801*disrupted! sensorimotor! gating.!Pharmacology,!Biochemistry,!and!Behavior,!80(4):!591*596.!

Buckley!PF.!(2005).!Neuroimaging!of!schizophrenia:!Structural!abnormalities!and!pathophysiological!implications.!Neuropsychiatric!Disease!and!Treatment,!1(3):!193*204.!!

Buka!SL,!Tsuang!MT,!Torrey!EF,!Klebanoff!MA,!Bernstein!D!&!Yolken!RH.!(2001).!Maternal!infections!and!subsequent!psychosis!among!offspring.!Archives!of!General!Psychiatry,!58(11):!1032*1037.!!

Burnet! PW! &! Harrison! PJ.! (1995).! Genetic! variation! of! the! 5*HT2A! receptor! and! response! to!clozapine.!Lancet,!346(8979):!909.!!

Cai!J,!Zhang!W,!Yi!Z,!Lu!W,!Wu!Z,!Chen!J,!.!.!.!Zhang!C.!(2013).!Influence!of!polymorphisms!in!genes!SLC1A1,! GRIN2B,! and! GRIK2! on! clozapine*induced! obsessive*compulsive! symptoms.!Psychopharmacology,!230(1):!49*55.!!

Canadian! Psychiatric! Association.! (2005).! Clinical! practice! guidelines.! Treatment! of! schizophrenia.!Canadian!Journal!of!Psychiatry,!Supplemental,!50(13,!S1):!7S*57S.!!

Cantor*Graae!E!&!Selten!JP.! (2005).!Schizophrenia!and!migration:!A!meta*analysis!and!review.!The!American!Journal!of!Psychiatry,!162(1):!12*24.!!

Cardno! AG! &! Gottesman! II.! (2000).! Twin! studies! of! schizophrenia:! From! bow*and*arrow!concordances!to!star!wars!Mx!and!functional!genomics.!American!Journal!of!Medical!Genetics,!97(1):!12*17.!!

Cardon! LR! &! Palmer! LJ.! (2003).! Population! stratification! and! spurious! allelic! association.! Lancet,!361(9357):!598*604.!

Carlquist!JF,!McKinney!JT,!Nicholas!ZP,!Clark!JL,!Kahn!SF,!Horne!BD,!.! .! .!Anderson!JL.!(2008).!Rapid!melting! curve! analysis! for! genetic! variants! that! underlie! inter*individual! variability! in! stable!warfarin!dosing.!Journal!of!Thrombosis!and!Thrombolysis,!26(1):!1*7.!!

Carlsson! A! &! Lindqvist! M.! (1963).! Effect! of! chlorpromazine! or! haloperidol! on! formation! of! 3*methoxytyramine!and!normetanephrine!in!mouse!brain.!Acta!Pharmacologica!et!Toxicologica,!20:!140*144.!!

Cartmell! J! &! Schoepp! DD.! (2000).! Regulation! of! neurotransmitter! release! by! metabotropic!glutamate!receptors.!Journal!of!Neurochemistry,!75(3):!889*907.!!

133!!

!!

Caspi! A! &! Moffitt! TE.! (2006).! Gene*environment! interactions! in! psychiatry:! Joining! forces! with!neuroscience.!Nature!Reviews,!Neuroscience,!7(7):!583*590.!!

Centorrino! F,! Hernan! MA,! Drago*Ferrante! G,! Rendall! M,! Apicella! A,! Langar! G! &! Baldessarini! RJ.!(2001).! Factors! associated! with! noncompliance! with! psychiatric! outpatient! visits.! Psychiatric!Services,!52(3):!378*380.!!

Centre! for! the! Evaluation! of! Value! and! Risk! in! Health.! (2011).! The! cost*effectiveness! analysis!registry,! Tufts! Medical! Centre.! Retrieved! July! 27th,! 2014,! from! https://research.tufts*nemc.org/cear4/!!

Chen! L,! Muhlhauser! M! &! Yang! CR.! (2003).! Glycine! tranporter*1! blockade! potentiates! NMDA*mediated! responses! in! rat! prefrontal! cortical! neurons! in! vitro! and! in! vivo.! Journal! of!Neurophysiology,!89(2):!691*703.!!

Chen! L! &! Yang! CR.! (2002).! Interaction! of! dopamine! D1! and! NMDA! receptors! mediates! acute!clozapine!potentiation!of!glutamate!EPSPs!in!rat!prefrontal!cortex.!Journal!of!Neurophysiology,!87(5):!2324*2336.!!

Chiu!HJ,!Wang!YC,!Liou!YJ,!Lai!IC!&!Chen!JY.!(2003).!Association!analysis!of!the!genetic!variants!of!the!N*methyl! D*aspartate! receptor! subunit! 2b! (NR2b)! and! treatment*refractory! schizophrenia! in!the!Chinese.!Neuropsychobiology,!47(4):!178*181.!!

Choudhry! S,! Seibold! MA,! Borrell! LN,! Tang! H,! Serebrisky! D,! Chapela! R,! .! .! .! Burchard! EG.! (2007).!Dissecting!complex!diseases!in!complex!populations:!Asthma!in!Latino!Americans.!Proceedings!of!the!American!Thoracic!Society,!4(3):!226*233.!!

Chowdhury! NI,! Tiwari! AK,! Souza! RP,! Zai! CC,! Shaikh! SA,! Chen! S,! .! .! .! Muller! DJ.! (2013).! Genetic!association!study!between!antipsychotic*induced!weight!gain!and!the!melanocortin*4!receptor!gene.!The!Pharmacogenomics!Journal,!13(3):!272*279.!!

Chumakov! I,! Blumenfeld!M,!Guerassimenko!O,!Cavarec! L,! Palicio!M,!Abderrahim!H,! .! .! .! Cohen!D.!(2002).!Genetic!and!physiological!data!implicating!the!new!human!gene!G72!and!the!gene!for!D*amino!acid!oxidase!in!schizophrenia.!Proceedings!of!the!National!Academy!of!Sciences!of!the!United!States!of!America,!99(21):!13675*13680.!!

Claghorn!J,!Honigfeld!G,!Abuzzahab!FS!Sr,!Wang!R,!Steinbook!R,!Tuason!V!&!Klerman!G.!(1987).!The!risks!and!benefits!of!clozapine!versus!chlorpromazine.!Journal!of!Clinical!Psychopharmacology,!7(6):!377*384.!!

Clayton!D!&!McKeigue!PM.!(2001).!Epidemiological!methods!for!studying!genes!and!environmental!factors!in!complex!diseases.!Lancet,!358(9290):!1356*1360.!!

Clinton! SM!&!Meador*Woodruff! JH.! (2004).! Abnormalities! of! the! NMDA! receptor! and! associated!intracellular! molecules! in! the! thalamus! in! schizophrenia! and! bipolar! disorder.!Neuropsychopharmacology,!29(7):!1353*1362.!!

134!!

!!

Clouston!TS.!(1904).!Clinical!Lectures!on!Mental!Diseases.!6th!ed!London,!UK:!J&A!Churchill.!!

Conley! RR,! Tamminga! CA,! Kelly! DL! &! Richardson! CM.! (1999).! Treatment*resistant! schizophrenic!patients!respond!to!clozapine!after!olanzapine!non*response.!Biological!Psychiatry,!46(1):!73*77.!!

Conn!PJ!&!Pin!JP.!(1997).!Pharmacology!and!functions!of!metabotropic!glutamate!receptors.!Annual!Review!of!Pharmacology!and!Toxicology,!37:!205*237.!!

Corbett!R,!Camacho!F,!Woods!AT,!Kerman!LL,!Fishkin!RJ,!Brooks!K!&!Dunn!RW.!(1995).!Antipsychotic!agents! antagonize! non*competitive! N*methyl*D*aspartate! antagonist*induced! behaviors.!Psychopharmacology,!120(1):!67*74.!!

Corvin!AP,!Morris!DW,!McGhee!K,!Schwaiger!S,!Scully!P,!Quinn! J,! .! .! .!Gill!M.! (2004).!Confirmation!and!refinement!of!an! 'at*risk'!haplotype!for!schizophrenia!suggests!the!EST!cluster,!Hs.97362,!as!a!potential!susceptibility!gene!at!the!neuregulin*1!locus.!Molecular!Psychiatry,!9(2):!208*213.!!

Cosgrove!VE!&!Suppes!T.!(2013).!Informing!DSM*5:!Biological!boundaries!between!bipolar!I!disorder,!schizoaffective!disorder,!and!schizophrenia.!BMC!Medicine,!11(127):!1*7.!!

Costa! e! Silva! JA.! (2013).! Personalized!medicine! in! psychiatry:! New! technologies! and! approaches.!Metabolism:!Clinical!and!Experimental,!Supplemental,!62(S1):!S40*S44.!!

Coyle! JT! &! Puttfarcken! P.! (1993).! Oxidative! stress,! glutamate,! and! neurodegenerative! disorders.!Science,!262(5134):!689*695.!!

Creese! I,! Burt! DR! &! Snyder! SH.! (1976).! Dopamine! receptors! and! average! clinical! doses.! Science,!194(4264):!546.!

CRESTAR.! (2011).! CRESTAR:! Development! of! pharmacogenetic! biomarkers! for! schizophrenia.!Retrieved!March!14th,!2014,!from!http://www.crestar*project.eu/!!

Csernansky!JG,!Wang!L,!Jones!D,!Rastogi*Cruz!D,!Posener!JA,!Heydebrand!G,! .! .! .!Miller!MI.!(2002).!Hippocampal! deformities! in! schizophrenia! characterized! by! high! dimensional! brain!mapping.!The!American!Journal!of!Psychiatry,!159(12):!2000*2006.!!

Cubelos! B,! Gimenez! C! &! Zafra! F.! (2005).! Localization! of! the! GLYT1! glycine! transporter! at!glutamatergic!synapses!in!the!rat!brain.!Cerebral!Cortex,!15(4):!448*459.!!

Curthoys!NP!&!Watford!M.! (1995).! Regulation!of! glutaminase! activity! and! glutamine!metabolism.!Annual!Review!of!Nutrition,!15:!133*159.!

Curtis! DR,! Phillis! JW!&!Watkins! JC.! (1960).! The! chemical! excitation! of! spinal! neurones! by! certain!acidic!amino!acids.!The!Journal!of!Physiology,!150:!656*682.!!

Daikhin!Y!&!Yudkoff!M.! (2000).!Compartmentation!of!brain!glutamate!metabolism! in!neurons!and!glia.!The!Journal!of!Nutrition,!Supplemental,!130(4S):!1026S*31S.!!

135!!

!!

Dallaspezia! S,! Poletti! S,! Lorenzi! C,! Pirovano! A,! Colombo! C!&! Benedetti! F.! (2012).! Influence! of! an!interaction!between! lithium!salts!and!a! functional!polymorphism! in!SLC1A2!on! the!history!of!illness!in!bipolar!disorder.!Molecular!Diagnosis!&!Therapy,!16(5):!303*309.!!

Daly!DA!&!Moghaddam!B.!(1993).!Actions!of!clozapine!and!haloperidol!on!the!extracellular!levels!of!excitatory! amino! acids! in! the! prefrontal! cortex! and! striatum! of! conscious! rats.! Neuroscience!Letters,!152(1*2):!61*64.!!

Danysz! W! &! Parsons! CG.! (1998).! Glycine! and! N*methyl*D*aspartate! receptors:! Physiological!significance!and!possible!therapeutic!applications.!Pharmacological!Reviews,!50(4):!597*664.!!

Davis! JM,! Chen! N! &! Glick! ID.! (2003).! A! meta*analysis! of! the! efficacy! of! second*generation!antipsychotics.!Archives!of!General!Psychiatry,!60(6):!553*564.!!

Davis! KL,! Kahn! RS,! Ko! G! &! Davidson! M.! (1991).! Dopamine! in! schizophrenia:! A! review! and!reconceptualization.!The!American!Journal!of!Psychiatry,!148(11):!1474*1486.!!

de!la!Fuente*Sandoval!C,!Leon*Ortiz!P,!Azcarraga!M,!Favila!R,!Stephano!S!&!Graff*Guerrero!A.!(2013).!Striatal!glutamate!and!the!conversion!to!psychosis:!A!prospective!1H*MRS!imaging!study.!The!International!Journal!of!Neuropsychopharmacology,!16(2):!471*475.!!

de! Leon! J.! (2004).! Atypical! antipsychotic! dosing:! The! effect! of! smoking! and! caffeine.! Psychiatric!Services,!55(5):!491*493.!!

De! Sousa! KR,! Tiwari! AK,! Giuffra! DE,! Mackenzie! B,! Zai! CC! &! Kennedy! JL.! (2013).! Age! at! onset! of!schizophrenia:!Cannabis,!COMT!gene,!and!their!interactions.!Schizophrenia!Research,!151(1*3):!289*290.!!

Delay! J,! Deniker! E! &! Had! JM.! (1952).! Traitement! des! etats! d'excitation! et! d'agitation! par! une!methode!medicamentense!derivee!de!l'hibernotherapie.!Annales!Médico!Psychologiques,!110:!267*273.!!

Demjaha! A,! Egerton! A,! Murray! RM,! Kapur! S,! Howes! OD,! Stone! JM! &! McGuire! PK.! (2014).!Antipsychotic!treatment!resistance!in!schizophrenia!associated!with!elevated!glutamate!levels!but!normal!dopamine!function.!Biological!Psychiatry,!75(5):!11*13.!!

Deng!X,!Shibata!H,!Ninomiya!H,!Tashiro!N,!Iwata!N,!Ozaki!N!&!Fukumaki!Y.!(2004).!Association!study!of!polymorphisms!in!the!excitatory!amino!acid!transporter!2!gene!(SLC1A2)!with!schizophrenia.!BMC!Psychiatry,!4:!21.!!

Detera*Wadleigh!SD!&!McMahon!FJ.!(2006).!G72/G30!in!schizophrenia!and!bipolar!disorder:!Review!and!meta*analysis.!Biological!Psychiatry,!60(2):!106*114.!

!Di!Maria!E,!Gulli!R,!Begni!S,!De!Luca!A,!Bignotti!S,!Pasini!A,!.!.!.!Mandich!P.!(2004).!Variations!in!the!NMDA!receptor!subunit!2B!gene!(GRIN2B)!and!schizophrenia:!A!case*control!study.!American!Journal!of!Medical!Genetics!Part!B,!Neuropsychiatric!Genetics,!128B(1):!27*29.!!

136!!

!!

Distlerath!LM,!Reilly!PE,!Martin!MV,!Davis!GG,!Wilkinson!GR!&!Guengerich!FP.! (1985).!Purification!and! characterization! of! the! human! liver! cytochromes! P*450! involved! in! debrisoquine! 4*hydroxylation! and! phenacetin! O*deethylation,! two! prototypes! for! genetic! polymorphism! in!oxidative!drug!metabolism.!The!Journal!of!Biological!Chemistry,!260(15):!9057*9067.!!

Dixon! M! &! Kleppe! K.! (1965).! D*amino! acid! oxidase.! I.! Dissociation! and! recombination! of! the!holoenzyme.!Biochimica!et!Biophysica!Acta,!96:!357*367.!!

Dong! E,! Nelson! M,! Grayson! DR,! Costa! E! &! Guidotti! A.! (2008).! Clozapine! and! sulpiride! but! not!haloperidol! or! olanzapine! activate! brain! DNA! demethylation.! Proceedings! of! the! National!Academy!of!Sciences!of!the!United!States!of!America,!105(36):!13614*13619.!!

Dorval!KM,!Wigg!KG,!Crosbie!J,!Tannock!R,!Kennedy!JL,!Ickowicz!A,!.!.!.!Barr!CL.!(2007).!Association!of!the! glutamate! receptor! subunit! gene! GRIN2B! with! attention*deficit/hyperactivity! disorder.!Genes,!Brain,!and!Behavior,!6(5):!444*452.!!

Dracheva! S,! Marras! SA,! Elhakem! SL,! Kramer! FR,! Davis! KL! &! Haroutunian! V.! (2001).! N*methyl*D*aspartic!acid!receptor!expression!in!the!dorsolateral!prefrontal!cortex!of!elderly!patients!with!schizophrenia.!The!American!Journal!of!Psychiatry,!158(9):!1400*1410.!!

Drazen! JM,! Yandava! CN,! Dube! L,! Szczerback! N,! Hippensteel! R,! Pillari! A,! .! .! .! Drajesk! J.! (1999).!Pharmacogenetic! association! between!ALOX5! promoter! genotype! and! the! response! to! anti*asthma!treatment.!Nature!Genetics,!22(2):!168*170.!!

Dudbridge! F.! (2008).! Likelihood*based! association! analysis! for! nuclear! families! and! unrelated!subjects!with!missing!genotype!data.!Human!Heredity,!66(2):!87*98.!

Dunbar*Jacob!J!&!Mortimer*Stephens!MK.!(2001).!Treatment!adherence!in!chronic!disease.!Journal!of!Clinical!Epidemiology,!Supplemental,!54(S1):!S57*S60.!!

Dunlop! DS! &! Neidle! A.! (1997).! The! origin! and! turnover! of! D*serine! in! brain.! Biochemical! and!Biophysical!Research!Communications,!235(1):!26*30.!!

Dunwiddie! TV.! (1980).! Endogenously! released! adenosine! regulates! excitability! in! the! in! vitro!hippocampus.!Epilepsia,!21(5):!541*548.!!

Dunwiddie! TV,! Hoffer! BJ! &! Fredholm! BB.! (1981).! Alkylxanthines! elevate! hippocampal! excitability.!evidence! for! a! role! of! endogenous! adenosine.! NaunynPSchmiedeberg's! Archives! of!Pharmacology,!316(4):!326*330.!!

Egerton! A,! Brugger! S,! Raffin!M,! Barker! GJ,! Lythgoe! DJ,!McGuire! PK!&! Stone! JM.! (2012).! Anterior!cingulate! glutamate! levels! related! to! clinical! status! following! treatment! in! first*episode!schizophrenia.!Neuropsychopharmacology,!37(11):!2515*2521.!!

Elkis! H! &! Meltzer! HY.! (2007).! Refractory! schizophrenia.! Revista! Brasileira! De! Psiquiatria,!Supplemental,!29(S2):!S41*S7.!!

137!!

!!

El*Saadi!O,!Pedersen!CB,!McNeil!TF,!Saha!S,!Welham!J,!O'Callaghan!E,!.!.!.!McGrath!J.!(2004).!Paternal!and!maternal!age!as!risk!factors!for!psychosis:!Findings!from!Denmark,!Sweden!and!Australia.!Schizophrenia!Research,!67(2*3):!227*236.!!

Emsley!R,!Rabinowitz!J,!Medori!R!&!Early!Psychosis!Global!Working!Group.!(2007).!Remission!in!early!psychosis:! Rates,! predictors,! and! clinical! and! functional! outcome! correlates.! Schizophrenia!Research,!89(1*3):!129*139.!!

ENCODE! Project! Consortium.! (2012).! An! integrated! encyclopedia! of! DNA! elements! in! the! human!genome.!Nature,!489(7414):!57*74.!!

Endele!S,!Rosenberger!G,!Geider!K,!Popp!B,!Tamer!C,!Stefanova!I,!.!.!.!Kutsche!K.!(2010).!Mutations!in!GRIN2A! and! GRIN2B! encoding! regulatory! subunits! of! NMDA! receptors! cause! variable!neurodevelopmental!phenotypes.!Nature!Genetics,!42(11):!1021*1026.!!

Erlander!MG,! Tillakaratne!NJ,! Feldblum! S,! Patel!N!&! Tobin! AJ.! (1991).! Two! genes! encode! distinct!glutamate!decarboxylases.!Neuron,!7(1):!91*100.!!

Ernst!J!&!Kellis!M.!(2012).!ChromHMM:!Automating!chromatin*state!discovery!and!characterization.!Nature!Methods,!9(3):!215*216.!!

Evans!DA,!Manley!KA!&!Mckusick!VA.!(1960).!Genetic!control!of!isoniazid!metabolism!in!man.!British!Medical!Journal,!2(5197):!485*491.!!

Evans!DM!&!Purcell!S.! (2012).!Power!calculations! in!genetic!studies.!Cold!Spring!Harbor!Protocols,!2012(6):!664*674.!!

Evers!K.!(2009).!Personalized!medicine!in!psychiatry:!Ethical!challenges!and!opportunities.!Dialogues!in!Clinical!Neuroscience,!11(4):!427*434.!!

Evins!AE,!Amico!ET,!Shih!V!&!Goff!DC.!(1997).!Clozapine!treatment!increases!serum!glutamate!and!aspartate! compared! to! conventional! neuroleptics.! Journal! of! Neural! Transmission,! 104(6*7):!761*766.!!

Evins! AE,! Fitzgerald! SM,!Wine! L,! Rosselli! R! &! Goff! DC.! (2000).! Placebo*controlled! trial! of! glycine!added!to!clozapine!in!schizophrenia.!The!American!Journal!of!Psychiatry,!157(5):!826*828.!!

Fagiolini!M,!Fritschy!JM,!Low!K,!Mohler!H,!Rudolph!U!&!Hensch!TK.!(2004).!Specific!GABAA!circuits!for!visual!cortical!plasticity.!Science,!303(5664):!1681*1683.!

Fan! F!&!Wood! KV.! (2007).! Bioluminescent! assays! for! high*throughput! screening.! Assay! and! Drug!Development!Technologies,!5(1):!127*136.!!

Farin!HF,!Bussen!M,!Schmidt!MK,!Singh!MK,!Schuster*Gossler!K!&!Kispert!A.!(2007).!Transcriptional!repression! by! the! T*box! proteins! Tbx18! and! Tbx15! depends! on! groucho! corepressors.! The!Journal!of!Biological!Chemistry,!282(35):!25748*25759.!!

138!!

!!

Fell!MJ,! Svensson! KA,! Johnson! BG!&! Schoepp! DD.! (2008).! Evidence! for! the! role! of!metabotropic!glutamate! (mGlu)2!not!mGlu3! receptors! in! the!preclinical! antipsychotic!pharmacology!of! the!mGlu2/3! receptor! agonist! (*)*(1R,4S,5S,6S)*4*amino*2*sulfonylbicyclo[3.1.0]hexane*4,6*dicarboxylic! acid! (LY404039).! The! Journal! of! Pharmacology! and! Experimental! Therapeutics,!326(1):!209*217.!!

Ferreira! R,! Ohneda! K,! Yamamoto! M! &! Philipsen! S.! (2005).! GATA1! function,! a! paradigm! for!transcription!factors!in!hematopoiesis.!Molecular!and!Cellular!Biology,!25(4):!1215*1227.!!

Fitzgerald! LW,!Deutch!AY,!Gasic!G,!Heinemann!SF!&!Nestler! EJ.! (1995).! Regulation!of! cortical! and!subcortical! glutamate! receptor! subunit! expression! by! antipsychotic! drugs.! The! Journal! of!Neuroscience,!15(3!Pt!2):!2453*2461.!!

Flint! AC,!Maisch! US,!Weishaupt! JH,! Kriegstein! AR! &!Monyer! H.! (1997).! NR2A! subunit! expression!shortens! NMDA! receptor! synaptic! currents! in! developing! neocortex.! The! Journal! of!Neuroscience,!17(7):!2469*2476.!!

Foster!GA!&!Roberts!PJ.!(1981).!Stimulation!of!rat!cerebellar!guanosine!3',5'*cyclic!monophosphate!(cyclic!GMP)! levels:!Effects!of!amino!acid!antagonists.!British! Journal!of!Pharmacology,!74(3):!723*729.!!

Frackiewicz! EJ,! Sramek! JJ,! Herrera! JM,! Kurtz! NM!&! Cutler! NR.! (1997).! Ethnicity! and! antipsychotic!response.!The!Annals!of!Pharmacotherapy,!31(11):!1360*1369.!!

Freed! WJ,! Bing! LA! &! Wyatt! RJ.! (1984).! Effects! of! neuroleptics! on! phencyclidine! (PCP)*induced!locomotor!stimulation!in!mice.!Neuropharmacology,!23(2A):!175*181.!!

Fribourg! M,! Moreno! JL,! Holloway! T,! Provasi! D,! Baki! L,! Mahajan! R,! .! .! .! Logothetis! DE.! (2011).!Decoding!the!signaling!of!a!GPCR!heteromeric!complex!reveals!a!unifying!mechanism!of!action!of!antipsychotic!drugs.!Cell,!147(5):!1011*1023.!!

Funke!B,!Finn!CT,!Plocik!AM,!Lake!S,!DeRosse!P,!Kane!JM,!.!.!.!Malhotra!AK.!(2004).!Association!of!the!DTNBP1! locus!with! schizophrenia! in! a!U.S.! population.! American! Journal! of!Human!Genetics,!75(5):!891*898.!!

Furuta!A,!Rothstein! JD!&!Martin!LJ.! (1997).!Glutamate!transporter!protein!subtypes!are!expressed!differentially!during!rat!CNS!development.!The!Journal!of!Neuroscience,!17(21):!8363*8375.!!

Gabernet! L,! Pauly*Evers!M,! Schwerdel! C,! Lentz!M,! Bluethmann! H,! Vogt! K,! .! .! .! Boison! D.! (2005).!Enhancement!of!the!NMDA!receptor!function!by!reduction!of!glycine!transporter*1!expression.!Neuroscience!Letters,!373(1):!79*84.!!

GAIN!Collaborative!Research!Group,!Manolio!TA,!Rodriguez!LL,!Brooks!L,!Abecasis!G,!Collaborative!Association!Study!of!Psoriasis,!.!.!.!Collins!FS.!(2007).!New!models!of!collaboration!in!genome*wide!association!studies:!The!genetic!association!information!network.!Nature!Genetics,!39(9):!1045*1051.!!

139!!

!!

Garrod!A.!(1909).!Inborn!errors!of!metabolism!(1st!ed).!England:!Oxford!University!Press.!!

Gauderman! WJ.! (2002).! Sample! size! requirements! for! matched! case*control! studies! of! gene*environment!interaction.!Statistics!in!Medicine,!21(1):!35*50.!!

Gauderman!WJ!&!Morrison!JM.! (2006).!QUANTO:!A!computer!program!for!power!and!sample!size!calculations!for!geneticPepidemiology!studies!(Version!1.2.4).!!

Ge!S,!Goh!EL,!Sailor!KA,!Kitabatake!Y,!Ming!GL!&!Song!H.!(2006).!GABA!regulates!synaptic!integration!of!newly!generated!neurons!in!the!adult!brain.!Nature,!439(7076):!589*593.!!

Ghassabian! S,! Chetty!M,! Tattam!BN,!Glen! J,! Rahme! J,! Stankovic! Z,! .! .! .!McLachlan!AJ.! (2010).! The!participation! of! cytochrome! P450! 3A4! in! clozapine! biotransformation! is! detected! in! people!with! schizophrenia! by! high*throughput! in! vivo! phenotyping.! Journal! of! Clinical!Psychopharmacology,!30(5):!629*631.!!

Giardino!L,!Bortolotti!F,!Orazzo!C,!Pozza!M,!Monteleone!P,!Calza!L!&!Maj!M.!(1997).!Effect!of!chronic!clozapine!administration!on![3H]MK801*binding!sites!in!the!rat!brain:!A!side*preference!action!in!cortical!areas.!Brain!Research,!762(1*2):!216*218.!!

Gilman!H,!van!Ess!PR!&!Shirley!DA.!(1944).!The!methalation!of!10*phenylphenothiazine!and!of!10*ethylphenothiazine.!Journal!of!the!American!Chemical!Society,!66:!1214*1216.!!

Glatt! SJ,! Faraone! SV! &! Tsuang! MT.! (2003).! Meta*analysis! identifies! an! association! between! the!dopamine!D2!receptor!gene!and!schizophrenia.!Molecular!Psychiatry,!8(11):!911*915.!!

Goeree!R,!Farahati!F,!Burke!N,!Blackhouse!G,!O'Reilly!D,!Pyne!J!&!Tarride!JE.!(2005).!The!economic!burden! of! schizophrenia! in! Canada! in! 2004.! Current! Medical! Research! and! Opinion,! 21(12):!2017*2028.!!

Goff! DC,! Henderson! DC,! Evins! AE! &! Amico! E.! (1999).! A! placebo*controlled! crossover! trial! of! D*cycloserine!added!to!clozapine!in!patients!with!schizophrenia.!Biological!Psychiatry,!45(4):!512*514.!!

Goff! DC,! Tsai! G,! Levitt! J,! Amico! E,! Manoach! D,! Schoenfeld! DA,! .! .! .! Coyle! JT.! (1999).! A! placebo*controlled! trial! of! D*cycloserine! added! to! conventional! neuroleptics! in! patients! with!schizophrenia.!Archives!of!General!Psychiatry,!56(1):!21*27.!!

Goffman! E.! (1963).! Stigma:! Notes! on! the! management! of! spoiled! identity.! Englewood! Cliffs,! NJ:!Prentice*Hall.!!

Goldstein! L.! (1967).! Pathways! of! glutamine! deamination! and! their! control! in! the! rat! kidney.! The!American!Journal!of!Physiology,!213(4):!983*989.!!

Gomes! I,! Collins! A,! Lonjou! C,! Thomas! NS,! Wilkinson! J,! Watson! M! &! Morton! N.! (1999).! Hardy*Weinberg!quality!control.!Annals!of!Human!Genetics,!63(6):!535*538.!!

140!!

!!

Gonzalez*Maeso!J,!Ang!RL,!Yuen!T,!Chan!P,!Weisstaub!NV,!Lopez*Gimenez!JF,!.!.!.!Sealfon!SC.!(2008).!Identification! of! a! serotonin/glutamate! receptor! complex! implicated! in! psychosis.! Nature,!452(7183):!93*97.!!

Gottesman!II.!(1991).!Schizophrenia!genesis:!The!origin!of!madness.!New!York:!Freeman.!!

Gottesman! II!&!Shields! J.! (1967).!A!polygenic!theory!of!schizophrenia.!Proceedings!of! the!National!Academy!of!Sciences!of!the!United!States!of!America,!58(1):!199*205.!!

Grace! AA.! (2012).! Dopamine! system! dysregulation! by! the! hippocampus:! Implications! for! the!pathophysiology!and!treatment!of!schizophrenia.!Neuropharmacology,!62(3):!1342*1348.!!

Grimwood! S,! Slater! P,! Deakin! JF! &! Hutson! PH.! (1999).! NR2B*containing! NMDA! receptors! are! up*regulated!in!temporal!cortex!in!schizophrenia.!Neuroreport,!10(3):!461*465.!!

Groc! L,! Choquet! D! &! Chaouloff! F.! (2008).! The! stress! hormone! corticosterone! conditions! AMPAR!surface!trafficking!and!synaptic!potentiation.!Nature!Neuroscience,!11(8):!868*870.!!

Gross! H! &! Langner! E.! (1966).! Das! wirkungprofil! eines! chemischen! neuartigen! breitband*neuroleptikums! der! dibenzodiazepingruppe.! Wiener! Medizinische! Wochenschrift,! 116:! 814*816.!!

Grucza! RA,! Johnson! EO,! Krueger! RF,! Breslau! N,! Saccone! NL,! Chen! LS,! .! .! .! Bierut! LJ.! (2010).!Incorporating!age!at!onset!of!smoking!into!genetic!models!for!nicotine!dependence:!Evidence!for!interaction!with!multiple!genes.!Addiction!Biology,!15(3):!346*357.!!

Grueter! BA! &! Winder! DG.! (2005).! Group! II! and! III! metabotropic! glutamate! receptors! suppress!excitatory! synaptic! transmission! in! the! dorsolateral! bed! nucleus! of! the! stria! terminalis.!Neuropsychopharmacology,!30(7):!1302*1311.!!

Gu!Z,!Jiang!Q,!Fu!AK,!Ip!NY!&!Yan!Z.!(2005).!Regulation!of!NMDA!receptors!by!neuregulin!signaling!in!prefrontal!cortex.!The!Journal!of!Neuroscience,!25(20):!4974*4984.!!

Guengerich!FP.!(2008).!Cytochrome!P450!and!chemical!toxicology.!Chemical!Research!in!Toxicology,!21(1):!70*83.!!

Gunduz*Bruce!H,!McMeniman!M,!Robinson!DG,!Woerner!MG,!Kane!JM,!Schooler!NR!&!Lieberman!JA.!(2005).!Duration!of!untreated!psychosis!and!time!to!treatment!response!for!delusions!and!hallucinations.!The!American!Journal!of!Psychiatry,!162(10):!1966*1969.!!

Gurling!HM,!Kalsi!G,!Brynjolfson!J,!Sigmundsson!T,!Sherrington!R,!Mankoo!BS,!.! .! .!Curtis!D.!(2001).!Genomewide! genetic! linkage! analysis! confirms! the! presence! of! susceptibility! loci! for!schizophrenia,!on!chromosomes!1q32.2,!5q33.2,!and!8p21*22!and!provides!support!for!linkage!to!schizophrenia,!on!chromosomes!11q23.3*24!and!20q12.1*11.23.!American!Journal!of!Human!Genetics,!68(3):!661*673.!!!

141!!

!!

Gutierrez*Maldonado!J,!Caqueo*Urizar!A!&!Kavanagh!DJ.!(2005).!Burden!of!care!and!general!health!in! families! of! patients! with! schizophrenia.! Social! Psychiatry! and! Psychiatric! Epidemiology,!40(11):!899*904.!!

Guy!W.!(1976).!ECDEU!assessment!manual!for!psychopharmacology.!Rockville,!MD:!US!Department!of!Health,!Education,!and!Welfare.!!

Haak! LL.! (1999).! Metabotropic! glutamate! receptor! modulation! of! glutamate! responses! in! the!suprachiasmatic!nucleus.!Journal!of!Neurophysiology,!81(3):!1308*1317.!!

Hafner!H,!Maurer!K,!Loffler!W,!Fatkenheuer!B,!an!der!Heiden!W,!Riecher*Rossler!A,!.!.!.!Gattaz!WF.!(1994).! The! epidemiology! of! early! schizophrenia.! Influence! of! age! and! gender! on! onset! and!early!course.!The!British!Journal!of!Psychiatry,!Supplemental,!(23):!S29*S38.!!

Hafner! H,! Loffler!W,!Maurer! K,! Hambrecht!M! &! an! der! Heiden!W.! (1999).! Depression,! negative!symptoms,! social! stagnation! and! social! decline! in! the! early! course! of! schizophrenia.! Acta!Psychiatrica!Scandinavica,!100(2):!105*118.!!

Hagg! S,! Spigset! O,! Mjorndal! T! &! Dahlqvist! R.! (2000).! Effect! of! caffeine! on! clozapine!pharmacokinetics!in!healthy!volunteers.!British!Journal!of!Clinical!Pharmacology,!49(1):!59*63.!!

Hagger!C,!Buckley!P,!Kenny!JT,!Friedman!L,!Ubogy!D!&!Meltzer!HY.!(1993).!Improvement!in!cognitive!functions! and! psychiatric! symptoms! in! treatment*refractory! schizophrenic! patients! receiving!clozapine.!Biological!Psychiatry,!34(10):!702*712.!!

Hall*Flavin!DK,!Winner!JG,!Allen!JD,!Carhart!JM,!Proctor!B,!Snyder!KA,!.!.!.!Mrazek!DA.!(2013).!Utility!of!integrated!pharmacogenomic!testing!to!support!the!treatment!of!major!depressive!disorder!in!a!psychiatric!outpatient!setting.!Pharmacogenetics!and!Genomics,!23(10):!535*548.!!

Hamon!J,!Paraire!J!&!Velluz!J.!(1952).!Remarques!sur!l'action!du!4560!R.P!sur!l'agitation!maniaque.!Annales!Médico!Psychologiques,!110(13):!331*335.!!

Hanson!A!&!Levin!B.!(2013).!Mental!health!informatics.!Part!3!P!Competencies!and!strategies:!Types!of!data.!New!York,!NY:!Oxford!University!Press.!!

Hardy!GH.!(1908).!Mendelian!proportions!in!a!mixed!population.!Science,!28(706):!49*50.!!

Hashimoto! K.! (2010).! Glycine! transport! inhibitors! for! the! treatment! of! schizophrenia.! The! Open!Medicinal!Chemistry!Journal,!4:!10*19.!!

Harris!EW,!Ganong!AH!&!Cotman!CW.!(1984).!Long*term!potentiation!in!the!hippocampus!involves!activation!of!N*methyl*D*aspartate!receptors.!Brain!Research,!323(1):!132*137.!!

Harris!LW,!Sharp!T,!Gartlon!J,!Jones!DN!&!Harrison!PJ.!(2003).!Long*term!behavioural,!molecular!and!morphological! effects! of! neonatal! NMDA! receptor! antagonism.! The! European! Journal! of!Neuroscience,!18(6):!1706*1710.!!

142!!

!!

Harrison! PJ! &! Law! AJ.! (2006).! Neuregulin! 1! and! schizophrenia:! Genetics,! gene! expression,! and!neurobiology.!Biological!Psychiatry,!60(2):!132*140.!!

Harrow! M,! Grossman! LS,! Herbener! ES! &! Davies! EW.! (2000).! Ten*year! outcome:! Patients! with!schizoaffective! disorders,! schizophrenia,! affective! disorders! and!mood*incongruent! psychotic!symptoms.!The!British!Journal!of!Psychiatry,!177:!421*426.!!

Harvey! RJ! &! Yee! BK.! (2013).! Glycine! transporters! as! novel! therapeutic! targets! in! schizophrenia,!alcohol!dependence!and!pain.!Nature!Reviews,!Drug!Discovery,!12(11):!866*885.!!

Heresco*Levy! U.! (2003).! Glutamatergic! neurotransmission! modulation! and! the! mechanisms! of!antipsychotic!atypicality.!Progress!in!NeuroPPsychopharmacology!&!Biological!Psychiatry,!27(7):!1113*1123.!!

Heresco*Levy! U,! Javitt! DC,! Ermilov! M,! Mordel! C,! Horowitz! A! &! Kelly! D.! (1996).! Double*blind,!placebo*controlled,! crossover! trial! of! glycine! adjuvant! therapy! for! treatment*resistant!schizophrenia.!The!British!Journal!of!Psychiatry,!169(5):!610*617.!!

Hermans! E! &! Challiss! RA.! (2001).! Structural,! signalling! and! regulatory! properties! of! the! group! I!metabotropic! glutamate! receptors:! Prototypic! family! C! G*protein*coupled! receptors.! The!Biochemical!Journal,!359(3):!465*484.!!

Heston! LL.! (1966).! Psychiatric! disorders! in! foster! home! reared! children! of! schizophrenic!mothers.!The!British!Journal!of!Psychiatry,!112(489):!819*825.!!

Higgins!J.!(1966).!Effects!of!child!rearing!by!schizophrenic!mothers.!Journal!of!Psychiatric!Research,!4(3):!153*167.!!

Higgins! J.! (1976).! Effects! of! child! rearing! by! schizophrenic! mothers:! A! follow*up.! Journal! of!Psychiatric!Research,!13(1):!1*9.!!

Higuchi!M,!Single!FN,!Kohler!M,!Sommer!B,!Sprengel!R!&!Seeburg!PH.!(1993).!RNA!editing!of!AMPA!receptor! subunit! GluR*B:! A! base*paired! intron*exon! structure! determines! position! and!efficiency.!Cell,!75(7):!1361*1370.!!

Hippius!H.! (1996).!The! founding! of! the! CINP! and! the! discovery! of! clozapine.! In!D.!Healy! (Ed),!The!psychopharmacologists.!New!York,!NY:!Chapman!&!Hall.!!

Hirai! H,! Kirsch! J,! Laube! B,! Betz! H! &! Kuhse! J.! (1996).! The! glycine! binding! site! of! the! N*methyl*D*aspartate!receptor!subunit!NR1:!Identification!of!novel!determinants!of!co*agonist!potentiation!in!the!extracellular!M3*M4!loop!region.!Proceedings!of!the!National!Academy!of!Sciences!of!the!United!States!of!America,!93(12):!6031*6036.!!

Hoggart!CJ,!Parra!EJ,!Shriver!MD,!Bonilla!C,!Kittles!RA,!Clayton!DG!&!McKeigue!PM.!(2003).!Control!of! confounding! of! genetic! associations! in! stratified! populations.! American! Journal! of! Human!Genetics,!72(6):!1492*1504.!!

143!!

!!

Hong!CJ,!Yu!YW,!Lin!CH,!Cheng!CY!&!Tsai!SJ.!(2001).!Association!analysis!for!NMDA!receptor!subunit!2B! (GRIN2B)! genetic! variants! and! psychopathology! and! clozapine! response! in! schizophrenia.!Psychiatric!Genetics,!11(4):!219*222.!!

Hopkins! CR.! (2011).! ACS! Chemical! Neuroscience! molecule! spotlight! on! RG1678.! ACS! Chemical!Neuroscience,!2(12):!685*686.!!

Horacek! J,! Libiger! C,! Hoschl! K,! Borzova! I,! Hendrychova! J.! (2001).! Clozapine*induced! concordant!agranulocytosis! in!monozygotic! twins.! International! Journal! of! Psychiatry! in! Clinical! Practice,!5(1):!71*73.!!

Hosking! L,! Lumsden! S,! Lewis! K,! Yeo! A,! McCarthy! L,! Bansal! A,! .! .! .! Xu! CF.! (2004).! Detection! of!genotyping! errors! by! Hardy*Weinberg! equilibrium! testing.! European! Journal! of! Human!Genetics,!12(5):!395*399.!!

Hovatta! I,! Varilo! T,! Suvisaari! J,! Terwilliger! JD,! Ollikainen! V,! Arajarvi! R,! .! .! .! Peltonen! L.! (1999).! A!genomewide! screen! for! schizophrenia!genes! in!an! isolated!Finnish! subpopulation,! suggesting!multiple!susceptibility!loci.!American!Journal!of!Human!Genetics,!65(4):!1114*1124.!!

Hoyer!C,!Kranaster!L,!Leweke!FM,!Klosterkotter!J,!Meyer*Lindenberg!A!&!Koethe!D.!(2010).!Familial!differential! treatment! response! in! schizophrenia! *! lessons! from! a! case! of! three! affected!siblings.!Pharmacopsychiatry,!43(5):!196*197.!!

Hu! G,! Schones! DE,! Cui! K,! Ybarra! R,! Northrup! D,! Tang! Q,! .! .! .! Zhao! K.! (2011).! Regulation! of!nucleosome!landscape!and!transcription!factor!targeting!at!tissue*specific!enhancers!by!BRG1.!Genome!Research,!21(10):!1650*1658.!!

Huettner! JE.! (2003).!Kainate! receptors!and!synaptic! transmission.!Progress! in!Neurobiology,!70(5):!387*407.!!

Hwang! R,! Souza! RP,! Tiwari! AK,! Zai! CC,!Muller! DJ,! Potkin! SG,! .! .! .! Kennedy! JL.! (2011).! Gene*gene!interaction! analyses! between!NMDA! receptor! subunit! and! dopamine! receptor! gene! variants!and!clozapine!response.!Pharmacogenomics,!12(2):!277*291.!!

Ichise!T,!Kano!M,!Hashimoto!K,!Yanagihara!D,!Nakao!K,!Shigemoto!R,!.!.!.!Aiba!A.!(2000).!mGluR1!in!cerebellar! purkinje! cells! essential! for! long*term! depression,! synapse! elimination,! and!motor!coordination.!Science,!288(5472):!1832*1835.!!

Idanpaan*Heikkila! J,! Alhava! E,! Olkinuora!M! &! Palva! IP.! (1997).! Agranulocytosis! during! treatment!with!clozapine.!European!Journal!of!Clinical!Pharmacology,!11:!193*198.!!

Insel! TR! &! Scolnick! EM.! (2006).! Cure! therapeutics! and! strategic! prevention:! Raising! the! bar! for!mental!health!research.!Molecular!Psychiatry,!11(1):!11*17.!!

International! HapMap! Consortium.! (2003).! The! international! HapMap! project.! Nature,! 426(6968):!789*796.!!

144!!

!!

International! Schizophrenia! Consortium,! Purcell! SM,!Wray! NR,! Stone! JL,! Visscher! PM,! O'Donovan!MC,! .! .! .!Sklar!P.! (2009).!Common!polygenic!variation!contributes!to!risk!of!schizophrenia!and!bipolar!disorder.!Nature,!460(7256):!748*752.!!

Ishimaru! M,! Kurumaji! A! &! Toru! M.! (1992).! NMDA*associated! glycine! binding! site! increases! in!schizophrenic!brains.!Biological!Psychiatry,!32(4):!379*381.!!

Jablensky! A,! Sartorius! N,! Ernberg! G,! Anker! M,! Korten! A,! Cooper! JE,! .! .! .! Bertelsen! A.! (1992).!Schizophrenia:! Manifestations,! incidence! and! course! in! different! cultures.! A! world! health!organization!ten*country!study.!Psychological!Medicine!Monograph,!Supplemental,!20:!S1*S97.!!

Janca!A,!Kastrup!M,!Katschnig!H,!Lopez*Ibor!JJ!Jr,!Mezzich!JE!&!Sartorius!N.!(1996).!The!World!Health!Organization!short!disability!assessment!schedule! (WHO!DAS*S):!A!tool! for! the!assessment!of!difficulties!in!selected!areas!of!functioning!of!patients!with!mental!disorders.!Social!Psychiatry!and!Psychiatric!Epidemiology,!31(6):!349*354.!!

Javitt! DC! &! Zukin! SR.! (1991).! Recent! advances! in! the! phencyclidine!model! of! schizophrenia.! The!American!Journal!of!Psychiatry,!148(10):!1301*1308.!!

Javitt!DC,!Sershen!H,!Hashim!A!&!Lajtha!A.!(1997).!Reversal!of!phencyclidine*induced!hyperactivity!by! glycine! and! the! glycine! uptake! inhibitor! glycyldodecylamide.! Neuropsychopharmacology,!17(3):!202*204.!

Javitt!DC.! (2004).!Glutamate!as!a! therapeutic! target! in!psychiatric!disorders.!Molecular!Psychiatry,!9(11):!984*97.!!

Javitt!DC,!Duncan!L,!Balla!A!&!Sershen!H.!(2005).!Inhibition!of!system!A*mediated!glycine!transport!in! cortical! synaptosomes! by! therapeutic! concentrations! of! clozapine:! Implications! for!mechanisms!of!action.!Molecular!Psychiatry,!10(3):!275*287.!!

Javitt!DC,!Silipo!G,!Cienfuegos!A,!Shelley!AM,!Bark!N,!Park!M,!.!.!.!Zukin!SR.!(2001).!Adjunctive!high*dose! glycine! in! the! treatment! of! schizophrenia.! The! International! Journal! of!Neuropsychopharmacology,!4(4):!385*391.!

Jernigan!TL,!Zisook!S,!Heaton!RK,!Moranville!JT,!Hesselink!JR!&!Braff!DL.!(1991).!Magnetic!resonance!imaging! abnormalities! in! lenticular! nuclei! and! cerebral! cortex! in! schizophrenia.! Archives! of!General!Psychiatry,!48(10):!881*890.!!

Jia!P,!Wang!L,!Fanous!AH,!Pato!CN,!Edwards!TL,! International!Schizophrenia!Consortium!&!Zhao!Z.!(2012).! Network*assisted! investigation! of! combined! causal! signals! from! genome*wide!association!studies!in!schizophrenia.!PLoS!Computational!Biology,!8(7):!e1002587.!!

Johnson! JA,!Gong! L,!Whirl*Carrillo!M,!Gage!BF,! Scott! SA,! Stein!CM,! .! .! .! Clinical! Pharmacogenetics!Implementation! Consortium.! (2011).! Clinical! Pharmacogenetics! Implementation! Consortium!guidelines!for!CYP2C9!and!VKORC1!genotypes!and!warfarin!dosing.!Clinical!Pharmacology!and!Therapeutics,!90(4):!625*629.!

145!!

!!

Johnson! JW!&!Ascher!P.! (1987).!Glycine!potentiates! the!NMDA! response! in! cultured!mouse!brain!neurons.!Nature,!325(6104):!529*531.!!

Johnstone!EC,!Crow!TJ,!Frith!CD,!Husband!J!&!Kreel!L.!(1976).!Cerebral!ventricular!size!and!cognitive!impairment!in!chronic!schizophrenia.!Lancet,!2(7992):!924*926.!!

Jones!EM,!Fernald!A,!Bell!GI!&!Le!Beau!MM.!(1995).!Assignment!of!SLC6A9!to!human!chromosome!band!1p33!by!in!situ!hybridization.!Cytogenetics!and!Cell!Genetics,!71(3):!211.!!

Joo! A,! Shibata! H,! Ninomiya! H,! Kawasaki! H,! Tashiro! N! &! Fukumaki! Y.! (2001).! Structure! and!polymorphisms!of!the!human!metabotropic!glutamate!receptor!type!2!gene!(GRM2):!Analysis!of!association!with!schizophrenia.!Molecular!Psychiatry,!6(2):!186*192.!!

Kalow!W.! (1962).! Pharmacogenetics:! Heredity! and! the! response! to! drugs.! Philadelphia,! USA:!WB!Saunders!&!Co.!!

Kalsi!G,!Kuo!PH,!Aliev!F,!Alexander!J,!McMichael!O,!Patterson!DG,!.!.!.!Riley!BP.!(2010).!A!systematic!gene*based!screen!of!chr4q22*q32! identifies!association!of!a!novel! susceptibility!gene,!DKK2,!with!the!quantitative!trait!of!alcohol!dependence!symptom!counts.!Human!Molecular!Genetics,!19(12):!2497*2506.!!

Kane! J,! Honigfeld! G,! Singer! J! &! Meltzer! H.! (1988).! Clozapine! for! the! treatment*resistant!schizophrenic.!A!double*blind!comparison!with!chlorpromazine.!Archives!of!General!Psychiatry,!45(9):!789*796.!!

Kargieman!L,!Santana!N,!Mengod!G,!Celada!P!&!Artigas!F.! (2007).!Antipsychotic!drugs! reverse! the!disruption! in! prefrontal! cortex! function! produced! by! NMDA! receptor! blockade! with!phencyclidine.! Proceedings! of! the! National! Academy! of! Sciences! of! the! United! States! of!America,!104(37):!14843*14848.!!

Karreman! M! &! Moghaddam! B.! (1996).! The! prefrontal! cortex! regulates! the! basal! release! of!dopamine! in! the! limbic! striatum:! An! effect! mediated! by! ventral! tegmental! area.! Journal! of!Neurochemistry,!66(2):!589*598.!!

Kasai! K,! Shenton!ME,! Salisbury! DF,! Hirayasu! Y,! Lee! CU,! Ciszewski! AA,! .! .! .! McCarley! RW.! (2003).!Progressive!decrease!of!left!superior!temporal!gyrus!gray!matter!volume!in!patients!with!first*episode!schizophrenia.!The!American!Journal!of!Psychiatry,!160(1):!156*164.!!

Kattenstroth!G,!Tantalaki!E,!Sudhof!TC,!Gottmann!K!&!Missler!M.!(2004).!Postsynaptic!N*methyl*D*aspartate!receptor!function!requires!alpha*neurexins.!Proceedings!of!the!National!Academy!of!Sciences!of!the!United!States!of!America,!101(8):!2607*2612.!!

Kay! SR,! Fiszbein! A! &! Opler! LA.! (1987).! The! positive! and! negative! syndrome! scale! (PANSS)! for!schizophrenia.!Schizophrenia!Bulletin,!13(2):!261*276.!!

146!!

!!

Kelleher!RJ!3rd,!Geigenmuller!U,!Hovhannisyan!H,!Trautman!E,!Pinard!R,!Rathmell!B,!.!.!.!Margulies!D.!(2012).!High*throughput!sequencing!of!mGluR!signaling!pathway!genes!reveals!enrichment!of!rare!variants!in!autism.!PloS!One,!7(4):!e35003.!!

Kemether!EM,!Buchsbaum!MS,!Byne!W,!Hazlett!EA,!Haznedar!M,!Brickman!AM,!.!.!.!Bloom!R.!(2003).!Magnetic! resonance! imaging! of! mediodorsal,! pulvinar,! and! centromedian! nuclei! of! the!thalamus!in!patients!with!schizophrenia.!Archives!of!General!Psychiatry,!60(10):!983*991.!!

Kendler!KS.!(1983).!Overview:!A!current!perspective!on!twin!studies!of!schizophrenia.!The!American!Journal!of!Psychiatry,!140(11):!1413*1425.!!

Kennedy!JL,!Altar!CA,!Taylor!DL,!Degtiar!I!&!Hornberger!JC.!(2014).!The!social!and!economic!burden!of! treatment*resistant! schizophrenia:! A! systematic! literature! review.! International! Clinical!Psychopharmacology,!29(2):!63*76.!!

Kenny!EM,!Cormican!P,!Furlong!S,!Heron!E,!Kenny!G,!Fahey!C,!.!.!.!Morris!DW.!(2014).!Excess!of!rare!novel! loss*of*function! variants! in! synaptic! genes! in! schizophrenia! and! autism! spectrum!disorders.!Molecular!Psychiatry,!19(8):!872*879.!!

Kent!WJ,!Sugnet!CW,!Furey!TS,!Roskin!KM,!Pringle!TH,!Zahler!AM!&!Haussler!D.!(2002).!The!human!genome!browser!at!UCSC.!Genome!Research,!12(6):!996*1006.!!

Kety!SS.!(1959).!Biochemical!theories!of!schizophrenia.!II.!Science,!129(3363):!1590*1596.!!

Khakh!BS!&!Henderson!G.! (2000).!Modulation!of! fast! synaptic! transmission!by!presynaptic! ligand*gated!cation!channels.!Journal!of!the!Autonomic!Nervous!System,!81(1*3):!110*121.!!

Kharazia!VN,!Phend!KD,!Rustioni!A!&!Weinberg!RJ.! (1996).!EM!colocalization!of!AMPA!and!NMDA!receptor!subunits!at!synapses!in!rat!cerebral!cortex.!Neuroscience!Letters,!210(1):!37*40.!!

Kikuchi!G.!(1973).!The!glycine!cleavage!system:!Composition,!reaction!mechanism,!and!physiological!significance.!Molecular!and!Cellular!Biochemistry,!1(2):!169*187.!!

Kinney!GG,!Sur!C,!Burno!M,!Mallorga!PJ,!Williams!JB,!Figueroa!DJ,! .! .! .!Conn!PJ.!(2003).!The!glycine!transporter! type! 1! inhibitor! N*[3*(4'*fluorophenyl)*3*(4'*phenylphenoxy)propyl]sarcosine!potentiates!NMDA!receptor*mediated!responses! in!vivo!and!produces!an!antipsychotic!profile!in!rodent!behavior.!The!Journal!of!Neuroscience,!23(20):!7586*7591.!!

Kinon! BJ,! Liu*Seifert! H,! Adams! DH! &! Citrome! L.! (2006).! Differential! rates! of! treatment!discontinuation! in! clinical! trials! as! a!measure! of! treatment! effectiveness! for! olanzapine! and!comparator!atypical!antipsychotics!for!schizophrenia.! Journal!of!Clinical!Psychopharmacology,!26(6):!632*637.!

Kinon!BJ,!Zhang!L,!Millen!BA,!Osuntokun!OO,!Williams!JE,!Kollack*Walker!S,! .! .! .!HBBI!Study!Group.!(2011).!A!multicenter,!inpatient,!phase!2,!double*blind,!placebo*controlled!dose*ranging!study!of! LY2140023! monohydrate! in! patients! with! DSM*IV! schizophrenia.! Journal! of! Clinical!Psychopharmacology,!31(3):!349*355.!!

147!!

!!

Kinon!BJ,!Millen!BA!&!Downing!AC.! (2013).! LY2140023!monohydrate! in! the! treatment!of!patients!with!schizophrenia:!Results!of!2!clinical! trials!assessing!efficacy! in! treating!acutely! ill!patients!and! those! with! prominent! negative! symptoms.! Schizophrenia! Bulletin,! Supplemental,! 39(1):!S338.!!

Kleckner! NW! &! Dingledine! R.! (1988).! Requirement! for! glycine! in! activation! of! NMDA*receptors!expressed!in!xenopus!oocytes.!Science,!241(4867):!835*837.!!

Klosterkotter!J,!Hellmich!M,!Steinmeyer!EM!&!Schultze*Lutter!F.!(2001).!Diagnosing!schizophrenia!in!the!initial!prodromal!phase.!Archives!of!General!Psychiatry,!58(2):!158*164.!!

Kohlrausch! FB.! (2013).! Pharmacogenetics! in! schizophrenia:! A! review! of! clozapine! studies.! Revista!Brasileira!De!Psiquiatria,!35(3):!305*317.!!

Kolachana! BS,! Saunders! RC! &! Weinberger! DR.! (1995).! Augmentation! of! prefrontal! cortical!monoaminergic! activity! inhibits! dopamine! release! in! the! caudate! nucleus:! An! in! vivo!neurochemical!assessment!in!the!rhesus!monkey.!Neuroscience,!69(3):!859*868.!!

Komuro! H! &! Rakic! P.! (1993).! Modulation! of! neuronal! migration! by! NMDA! receptors.! Science,!260(5104):!95*97.!!

Kraepelin! E.! (1899).! Diagnosis! and! prognosis! of! dementia! praecox.! Allgemeine! Zeitschrift! Fur!Psychiatrie,!56:!254*263.!!

Kraguljac! NV,! White! DM,! Reid! MA! &! Lahti! AC.! (2013).! Increased! hippocampal! glutamate! and!volumetric!deficits!in!unmedicated!patients!with!schizophrenia.!JAMA!Psychiatry,!70(12):!1294*1302.!!

Kreyenbuhl! J,! Buchanan!RW,!Dickerson! FB,!Dixon! LB!&! Schizophrenia! Patient!Outcomes!Research!Team! (PORT).! (2010).! The! schizophrenia! patient! outcomes! research! team! (PORT):! Updated!treatment!recommendations!2009.!Schizophrenia!Bulletin,!36(1):!94*103.!!

Kubota! T,! Jibiki! I,! Kishizawa! S! &! Kurokawa! K.! (1996).! Clozapine*induced! potentiation! of! synaptic!responses! in! the! perforant! path*dentate! gyrus! pathway! in! chronically! prepared! rabbits.!Neuroscience!Letters,!211(1):!21*24.!!

Kubota!T,! Jibiki! I,! Enokido!F,!Nakagawa!H!&!Watanabe!K.! (2000).! Effects!of!MK*801!on!clozapine*induced! potentiation! of! excitatory! synaptic! responses! in! the! perforant! path*dentate! gyrus!pathway!in!chronically!prepared!rabbits.!European!Journal!of!Pharmacology,!395(1):!37*42.!!

Kuryatov!A,!Laube!B,!Betz!H!&!Kuhse!J.!(1994).!Mutational!analysis!of!the!glycine*binding!site!of!the!NMDA!receptor:!Structural!similarity!with!bacterial!amino!acid*binding!proteins.!Neuron,!12(6):!1291*1300.!!

Laborit! H! &! Huguenard! P.! (1951).! L'hibernation! artificielle! par! moyen! pharmacodynamiques! et!physiques.!La!Presse!Médical,!59:!1329.!!

148!!

!!

Lacro! JP,!Dunn! LB,!Dolder!CR,! Leckband! SG!&! Jeste!DV.! (2002).! Prevalence!of! and! risk! factors! for!medication! nonadherence! in! patients!with! schizophrenia:! A! comprehensive! review!of! recent!literature.!The!Journal!of!Clinical!Psychiatry,!63(10):!892*909.!!

Lahiri!DK!&!Nurnberger! JI! Jr.! (1991).!A! rapid!non*enzymatic!method! for! the!preparation!of!HMW!DNA!from!blood!for!RFLP!studies.!Nucleic!Acids!Research,!19(19):!5444.!!

Lahti!AC,!Weiler!MA,!Tamara*Michaelidis!BA,!Parwani!A!&!Tamminga!CA.!(2001).!Effects!of!ketamine!in!normal!and!schizophrenic!volunteers.!Neuropsychopharmacology,!25(4):!455*467.!!

Lander!ES,!Linton!LM,!Birren!B,!Nusbaum!C,!Zody!MC,!Baldwin!J,!.!.!.!International!Human!Genome!Sequencing!Consortium.!(2001).!Initial!sequencing!and!analysis!of!the!human!genome.!Nature,!409(6822):!860*921.!!

Larson! MK,! Walker! EF! &! Compton! MT.! (2010).! Early! signs,! diagnosis! and! therapeutics! of! the!prodromal! phase! of! schizophrenia! and! related! psychotic! disorders.! Expert! Review! of!Neurotherapeutics,!10(8):!1347*1359.!

Laube! B,! Hirai! H,! Sturgess! M,! Betz! H! &! Kuhse! J.! (1997).! Molecular! determinants! of! agonist!discrimination!by!NMDA!receptor!subunits:!Analysis!of!the!glutamate!binding!site!on!the!NR2B!subunit.!Neuron,!18(3):!493*503.!!

Laursen!TM,!Munk*Olsen!T!&!Vestergaard!M.!(2012).!Life!expectancy!and!cardiovascular!mortality!in!persons!with!schizophrenia.!Current!Opinion!in!Psychiatry,!25(2):!83*88.!!

Lee!SH,!Kubicki!M,!Asami!T,!Seidman!LJ,!Goldstein!JM,!Mesholam*Gately!RI,!.!.!.!Shenton!ME.!(2013).!Extensive!white!matter!abnormalities! in!patients!with! first*episode!schizophrenia:!A!diffusion!tensor!imaging!(DTI)!study.!Schizophrenia!Research,!143(2*3):!231*238.!!

Lee!YL,!Teitelbaum!S,!Wolff!MS,!Wetmur!JG!&!Chen!J.!(2010).!Comparing!genetic!ancestry!and!self*reported!race/ethnicity!in!a!multiethnic!population!in!New!York!City.!Journal!of!Genetics,!89(4):!417*423.!!

Lemonde! S,! Turecki! G,! Bakish! D,! Du! L,! Hrdina! PD,! Bown! CD,! .! .! .! Albert! PR.! (2003).! Impaired!repression! at! a! 5*hydroxytryptamine! 1A! receptor! gene! polymorphism! associated!with!major!depression!and!suicide.!The!Journal!of!Neuroscience,!23(25):!8788*8799.!!

Lemonde! S,! Du! L,! Bakish! D,! Hrdina! P! &! Albert! PR.! (2004).! Association! of! the! C(*1019)G! 5PHT1A!functional!promoter!polymorphism!with!antidepressant!response.!The!International!Journal!of!Neuropsychopharmacology,!7(4):!501*506.!

Lett!TA,!Tiwari!AK,!Meltzer!HY,!Lieberman!JA,!Potkin!SG,!Voineskos!AN,! .! .! .!Muller!DJ.! (2011).!The!putative! functional! rs1045881!marker!of!neurexin*1! in!schizophrenia!and!clozapine!response.!Schizophrenia!Research,!132(2*3):!121*124.!!

149!!

!!

Levin!ED,!Caldwell!DP!&!Perraut!C.!(2007).!Clozapine!treatment!reverses!dizocilpine*induced!deficits!of!pre*pulse! inhibition!of! tactile! startle! response.! Pharmacology,!Biochemistry,!and!Behavior,!86(3):!597*605.!!

Lewis! CM! &! Knight! J.! (2012).! Introduction! to! genetic! association! studies.! Cold! Spring! Harbor!Protocols,!2012(3):!297*306.!!

Lewis!SW,!Ford!RA,!Syed!GM,!Reveley!AM!&!Toone!BK.!(1992).!A!controlled!study!of!99mTc*HMPAO!single*photon!emission!imaging!in!chronic!schizophrenia.!Psychological!Medicine,!22(1):!27*35.!!

Li!ML,!Hu!XQ,!Li!F!&!Gao!WJ.!(2015).!Perspectives!on!the!mGluR2/3!agonists!as!a!therapeutic!target!for! schizophrenia:! Still! promising! or! a! dead! end?! Progress! in! NeuroPPsychopharmacology! &!Biological!Psychiatry,!60:!66*76.!!

Li! X! &! Francke! U.! (1995).! Assignment! of! the! gene! SLC1A2! coding! for! the! human! glutamate!transporter!EAAT2!to!human!chromosome!11!bands!p13*p12.!Cytogenetics!and!Cell!Genetics,!71(3):!212*213.!!

Lieberman! JA,! Kane! JM! &! Alvir! J.! (1987).! Provocative! tests! with! psychostimulant! drugs! in!schizophrenia.!Psychopharmacology,!91(4):!415*433.!!

Lieberman!JA,!Safferman!AZ,!Pollack!S,!Szymanski!S,!Johns!C,!Howard!A,!.!.!.!Kane!JM.!(1994).!Clinical!effects! of! clozapine! in! chronic! schizophrenia:! Response! to! treatment! and! predictors! of!outcome.!The!American!Journal!of!Psychiatry,!151(12):!1744*1752.!!

Lieberman!JA,!Bymaster!FP,!Meltzer!HY,!Deutch!AY,!Duncan!GE,!Marx!CE,!.!.!.!Csernansky!JG.!(2008).!Antipsychotic! drugs:! Comparison! in! animal! models! of! efficacy,! neurotransmitter! regulation,!and!neuroprotection.!Pharmacological!Reviews,!60(3):!358*403.!!

Life! Technologies.! (2011).! Ion! torrent! amplicon! sequencing! applicaiton! note.! Unpublished!manuscript.!!

Lin!MW,!Curtis!D,!Williams!N,!Arranz!M,!Nanko!S,!Collier!D,!.!.!.!Gill!M.!(1995).!Suggestive!evidence!for! linkage! of! schizophrenia! to! markers! on! chromosome! 13q14.1*q32.! Psychiatric! Genetics,!5(3):!117*126.!!

Lindenmayer! J! &! Apergi! F.! (1996).! The! relationship! between! clozapine! plasma! levels! and! clinical!response.!Psychiatric!Annals,!26(7):!406*412.!!

Lindstrom! LH.! (1988).! The! effect! of! long*term! treatment! with! clozapine! in! schizophrenia:! A!retrospective!study!in!96!patients!treated!with!clozapine!for!up!to!13!years.!Acta!Psychiatrica!Scandinavica,!77(5):!524*529.!!

Linn! GS,! Negi! SS,! Gerum! SV! &! Javitt! DC.! (2003).! Reversal! of! phencyclidine*induced! prepulse!inhibition!deficits!by!clozapine!in!monkeys.!Psychopharmacology,!169(3*4):!234*239.!!

150!!

!!

Lipska! BK,! Swerdlow! NR,! Geyer! MA,! Jaskiw! GE,! Braff! DL! &! Weinberger! DR.! (1995).! Neonatal!excitotoxic!hippocampal!damage!in!rats!causes!post*pubertal!changes!in!prepulse!inhibition!of!startle!and!its!disruption!by!apomorphine.!Psychopharmacology,!122(1):!35*43.!!

Liu!N,! Zhao!H,! Patki! A,! Limdi!NA!&!Allison!DB.! (2011).! Controlling! population! structure! in! human!genetic! association! studies!with! samples! of! unrelated! individuals.! Statistics! and! its! Interface,!4(3):!317*326.!!

Liu! W,! Downing! AC,! Munsie! LM,! Chen! P,! Reed! MR,! Ruble! CL,! .! .! .! Nisenbaum! LK.! (2012).!Pharmacogenetic!analysis!of!the!mGlu2/3!agonist!LY2140023!monohydrate!in!the!treatment!of!schizophrenia.!The!Pharmacogenomics!Journal,!12(3):!246*254.!!

Lohmueller! KE,! Pearce! CL,! Pike! M,! Lander! ES! &! Hirschhorn! JN.! (2003).! Meta*analysis! of! genetic!association! studies! supports! a! contribution! of! common! variants! to! susceptibility! to! common!disease.!Nature!Genetics,!33(2):!177*182.!!

Long! AD! &! Langley! CH.! (1999).! The! power! of! association! studies! to! detect! the! contribution! of!candidate!genetic!loci!to!variation!in!complex!traits.!Genome!Research,!9(8):!720*731.!!

Luby! ED,! Cohen! BD,! Rosenbaum! G,! Gottlieb! JS! &! Kelley! R.! (1959).! Study! of! a! new!schizophrenomimetic!drug;!sernyl.!AMA!Archives!of!Neurology!and!Psychiatry,!81(3):!363*369.!!

Luisada! PV.! (1978).! The! phencyclidine! psychosis:! Phenomenology! and! treatment.! NIDA! Research!Monograph,!(21):!241*253.!!

Lundorf!MD,!Buttenschon!HN,!Foldager!L,!Blackwood!DH,!Muir!WJ,!Murray!V,! .! .! .!Mors!O.!(2005).!Mutational! screening! and! association! study! of! glutamate! decarboxylase! 1! as! a! candidate!susceptibility! gene! for! bipolar! affective! disorder! and! schizophrenia.! American! Journal! of!Medical!Genetics!Part!B,!135B(1):!94*101.!!

Lynex! CN,! Carr! IM,! Leek! JP,! Achuthan! R,! Mitchell! S,! Maher! ER,! .! .! .! Markham! AF.! (2004).!Homozygosity!for!a!missense!mutation!in!the!67!kDa!isoform!of!glutamate!decarboxylase!in!a!family!with!autosomal!recessive!spastic!cerebral!palsy:!Parallels!with!stiff*person!syndrome!and!other!movement!disorders.!BMC!Neurology,!4(1):!20.!!

Ma! Q! &! Lu! AY.! (2011).! Pharmacogenetics,! pharmacogenomics,! and! individualized! medicine.!Pharmacological!Reviews,!63(2):!437*459.!!

Malaspina!D,!Harlap! S,! Fennig! S,! Heiman!D,!Nahon!D,! Feldman!D!&! Susser! ES.! (2001).! Advancing!paternal!age!and!the!risk!of!schizophrenia.!Archives!of!General!Psychiatry,!58(4):!361*367.!!

Malhotra! AK,! Adler! CM,! Kennison! SD,! Elman! I,! Pickar! D! &! Breier! A.! (1997).! Clozapine! blunts! N*methyl*D*aspartate!antagonist*induced!psychosis:!A!study!with!ketamine.!Biological!Psychiatry,!42(8):!664*668.!!

151!!

!!

Malhotra!AK,!Correll!CU,!Chowdhury!NI,!Muller!DJ,!Gregersen!PK,!Lee!AT,! .! .! .!Kennedy! JL.! (2012).!Association! between! common! variants! near! the! melanocortin! 4! receptor! gene! and! severe!antipsychotic!drug*induced!weight!gain.!Archives!of!General!Psychiatry,!69(9):!904*912.!!

Mallolas! J,! Hurtado! O,! Castellanos! M,! Blanco! M,! Sobrino! T,! Serena! J,! .! .! .! Davalos! A.! (2006).! A!polymorphism!in!the!EAAT2!promoter!is!associated!with!higher!glutamate!concentrations!and!higher!frequency!of!progressing!stroke.!The!Journal!of!Experimental!Medicine,!203(3):!711*717.!!

Mandich!P,!Schito!AM,!Bellone!E,!Antonacci!R,!Finelli!P,!Rocchi!M!&!Ajmar!F.!(1994).!Mapping!of!the!human! NMDAR2B! receptor! subunit! gene! (GRIN2B)! to! chromosome! 12p12.! Genomics,! 22(1):!216*218.!!

Marcelis!M,!Navarro*Mateu!F,!Murray!R,!Selten!JP!&!Van!Os!J.!(1998).!Urbanization!and!psychosis:!A!study!of!1942*1978!birth!cohorts!in!the!Netherlands.!Psychological!Medicine,!28(4):!871*879.!!

Martin! S,! Henley! JM,! Holman! D,! Zhou! M,! Wiegert! O,! van! Spronsen! M,! .! .! .! Krugers! HJ.! (2009).!Corticosterone!alters!AMPAR!mobility!and!facilitates!bidirectional!synaptic!plasticity.!PloS!One,!4(3):!e4714.!!

Martinez*Hernandez!A,!Bell!KP!&!Norenberg!MD.!(1977).!Glutamine!synthetase:!Glial!localization!in!brain.!Science,!195(4284):!1356*1358.!!

Martucci! L!&!Kennedy! JL.! (2010).!Predicting!negative! symptom!change!during!drug! treatment.!US!Patent!7,794,934.!!

Masellis! M,! Paterson! AD,! Badri! F,! Lieberman! JA,! Meltzer! HY,! Cavazzoni! P! &! Kennedy! JL.! (1995).!Genetic!variation!of!5*HT2A!receptor!and!response!to!clozapine.!Lancet,!346(8982):!1108.!!

Massey! PV,! Johnson! BE,! Moult! PR,! Auberson! YP,! Brown! MW,! Molnar! E,! .! .! .! Bashir! ZI.! (2004).!Differential! roles! of! NR2A! and! NR2B*containing! NMDA! receptors! in! cortical! long*term!potentiation!and!long*term!depression.!The!Journal!of!Neuroscience,!24(36):!7821*7828.!!

Mata! I,!Madoz! V,! Arranz!MJ,! Sham! P!&!Murray! RM.! (2001).! Olanzapine:! Concordant! response! in!monozygotic!twins!with!schizophrenia.!The!British!Journal!of!Psychiatry,!178(1):!86.!!

Mathalon!DH,!Sullivan!EV,!Lim!KO!&!Pfefferbaum!A.!(2001).!Progressive!brain!volume!changes!and!the!clinical!course!of!schizophrenia!in!men:!A!longitudinal!magnetic!resonance!imaging!study.!Archives!of!General!Psychiatry,!58(2):!148*157.!!

Mathieu!G,! Groisman! IJ! &!Godard! B.! (2013).! Next! generation! sequencing! in! psychiatric! research:!What! study! participants! need! to! know! about! research! findings.! The! International! Journal! of!Neuropsychopharmacology,!16(9):!2119*2127.!!

Matsui! T,! Sekiguchi! M,! Hashimoto! A,! Tomita! U,! Nishikawa! T! &! Wada! K.! (1995).! Functional!comparison!of!D*serine!and!glycine!in!rodents:!The!effect!on!cloned!NMDA!receptors!and!the!extracellular!concentration.!Journal!of!Neurochemistry,!65(1):!454*458.!!

152!!

!!

Matys! V,! Kel*Margoulis! OV,! Fricke! E,! Liebich! I,! Land! S,! Barre*Dirrie! A,! .! .! .! Wingender! E.! (2006).!TRANSFAC! and! its! module! TRANSCompel:! Transcriptional! gene! regulation! in! eukaryotes.!Nucleic!Acids!Research,!Database!Issue,!34:!D108*D110.!!

Mayer! ML,! Westbrook! GL! &! Guthrie! PB.! (1984).! Voltage*dependent! block! by! Mg2+! of! NMDA!responses!in!spinal!cord!neurones.!Nature,!309(5965):!261*263.!!

Mayer! ML,! Vyklicky! L! Jr! &! Clements! J.! (1989).! Regulation! of! NMDA! receptor! desensitization! in!mouse!hippocampal!neurons!by!glycine.!Nature,!338(6214):!425*427.!!

Maziade!M,!Raymond!V,!Cliche!D,! Fournier! JP,!Caron!C,!Garneau!Y,! .! .! .! Simard!C.! (1995).! Linkage!results!on!11Q21*22!in!Eastern!Quebec!pedigrees!densely!affected!by!schizophrenia.!American!Journal!of!Medical!Genetics,!60(6):!522*528.!!

McBain! CJ! &! Mayer! ML.! (1994).! N*methyl*D*aspartic! acid! receptor! structure! and! function.!Physiological!Reviews,!74(3):!723*760.!!

McCarley! RW,!Wible! CG,! Frumin!M,! Hirayasu! Y,! Levitt! JJ,! Fischer! IA! &! Shenton!ME.! (1999).! MRI!anatomy!of!schizophrenia.!Biological!Psychiatry,!45(9):!1099*1119.!!

McClellan! JM,! Susser! E!&! King!MC.! (2007).! Schizophrenia:! A! common! disease! caused! by!multiple!rare!alleles.!The!British!Journal!of!Psychiatry,!190:!194*199.!!

McEvoy!JP,!Lieberman!JA,!Stroup!TS,!Davis!SM,!Meltzer!HY,!Rosenheck!RA,!.!.!.!CATIE!Investigators.!(2006).! Effectiveness! of! clozapine! versus! olanzapine,! quetiapine,! and! risperidone! in! patients!with!chronic!schizophrenia!who!did!not!respond!to!prior!atypical!antipsychotic!treatment.!The!American!Journal!of!Psychiatry,!163(4):!600*610.!!

McGlashan!TH.! (1988).!A!selective!review!of!recent!North!American! long*term!followup!studies!of!schizophrenia.!Schizophrenia!Bulletin,!14(4):!515*542.!!

McGrath!J,!Saha!S,!Welham!J,!El!Saadi!O,!MacCauley!C!&!Chant!D.!(2004).!A!systematic!review!of!the!incidence! of! schizophrenia:! The! distribution! of! rates! and! the! influence! of! sex,! urbanicity,!migrant!status!and!methodology.!BMC!Medicine,!2:!13.!!

McGrath! J,! Saha! S,! Chant!D!&!Welham! J.! (2008).! Schizophrenia:!A! concise! overview!of! incidence,!prevalence,!and!mortality.!Epidemiologic!Reviews,!30:!67*76.!!

McGurk!SR.!(1999).!The!effects!of!clozapine!on!cognitive!functioning!in!schizophrenia.!The!Journal!of!Clinical!Psychiatry,!Supplemental,!60(S12):!S24*S29.!!

McIlwain!ME,!Harrison!J,!Wheeler!AJ!&!Russell!BR.!(2011).!Pharmacotherapy!for!treatment*resistant!schizophrenia.!Neuropsychiatric!Disease!and!Treatment,!7:!135*149.!!

Meister!A.!(1974).!Glutamine!synthetase!of!mammals.!In!PD!Boyer!(Ed.),!The!enzymes!(Vol!10).!New!York:!Academic!Press.!!

153!!

!!

Melone!M,!Vitellaro*Zuccarello! L,!Vallejo*Illarramendi!A,!Perez*Samartin!A,!Matute!C,!Cozzi!A,! .! .! .!Conti! F.! (2001).! The! expression! of! glutamate! transporter! GLT*1! in! the! rat! cerebral! cortex! is!down*regulated!by!the!antipsychotic!drug!clozapine.!Molecular!Psychiatry,!6(4):!380*386.!!

Meltzer!HY,!Matsubara!S!&!Lee!JC.!(1989a).!Classification!of!typical!and!atypical!antipsychotic!drugs!on! the! basis! of! dopamine!D*1,!D*2! and! serotonin*2! pKi! values.! The! Journal! of! Pharmacology!and!Experimental!Therapeutics,!251(1):!238*246.!!

Meltzer!HY,!Bastani!B,!Kwon!KY,!Ramirez!LF,!Burnett!S!&!Sharpe!J.!(1989b).!A!prospective!study!of!clozapine! in! treatment*resistant! schizophrenic! patients.! I.! Preliminary! report.!Psychopharmacology,!Supplemental,!99:!S68*S72.!!

Meltzer! HY.! (1992).! Treatment! of! the! neuroleptic*nonresponsive! schizophrenic! patient.!Schizophrenia!Bulletin,!18(3):!515*542.!!

Meltzer!HY.! (1995a).! The! role! of! serotonin! in! schizophrenia! and! the!place!of! serotonin*dopamine!antagonist!antipsychotics.!Journal!of!Clinical!Psychopharmacology,!Supplemental,!15(1,!S1):!2S*3S.!!

Meltzer!HY!&!Okayli!G.!(1995b).!Reduction!of!suicidality!during!clozapine!treatment!of!neuroleptic*resistant!schizophrenia:!Impact!on!risk*benefit!assessment.!The!American!Journal!of!Psychiatry,!152(2):!183*190.!!

Meltzer!HY,!Bobo!WV,!Roy!A,!Jayathilake!K,!Chen!Y,!Ertugrul!A,!.!.!.!Small!JG.!(2008).!A!randomized,!double*blind!comparison!of!clozapine!and!high*dose!olanzapine!in!treatment*resistant!patients!with!schizophrenia.!The!Journal!of!Clinical!Psychiatry,!69(2):!274*285.!!

Mendel!G.!(1866).!Versuche!über!pflanzen*hybriden.!Verhandlungen!Des!Naturforschenden!Vereines!in!Brünn!(Abhandlungen),!4:!3*47.!!

Meshul!CK,!Bunker!GL,!Mason!JN,!Allen!C!&!Janowsky!A.!(1996).!Effects!of!subchronic!clozapine!and!haloperidol!on!striatal!glutamatergic!synapses.!Journal!of!Neurochemistry,!67(5):!1965*1973.!!

Meyer! JM.! (2001).! Individual! changes! in! clozapine! levels! after! smoking! cessation:! Results! and! a!predictive!model.!Journal!of!Clinical!Psychopharmacology,!21(6):!569*574.!!

Miyamoto!S,!Duncan!GE,!Marx!CE!&!Lieberman!JA.!(2005).!Treatments!for!schizophrenia:!A!critical!review! of! pharmacology! and! mechanisms! of! action! of! antipsychotic! drugs.! Molecular!Psychiatry,!10(1):!79*104.!!

Miyoshi!K,!Honda!A,!Baba!K,!Taniguchi!M,!Oono!K,!Fujita!T,! .! .! .!Tohyama!M.! (2003).!Disrupted*in*schizophrenia! 1,! a! candidate! gene! for! schizophrenia,! participates! in! neurite! outgrowth.!Molecular!Psychiatry,!8(7):!685*694.!!

Moghaddam! B! &! Javitt! D.! (2012).! From! revolution! to! evolution:! The! glutamate! hypothesis! of!schizophrenia!and!its!implication!for!treatment.!Neuropsychopharmacology,!37(1):!4*15.!!

154!!

!!

Mohn! AR,! Gainetdinov! RR,! Caron! MG! &! Koller! BH.! (1999).! Mice! with! reduced! NMDA! receptor!expression!display!behaviors!related!to!schizophrenia.!Cell,!98(4):!427*436.!!

Moniz! E.! (1937).! Prefrontal! leucotomy! in! the! treatment! of!mental! disorders.! Am! J! Psychiatry,! 93:!1379*1385.!!

Monn!JA,!Britton!TC,!Valli!MJ,!Massey!SM,!Henry!SS,!De!Dios!A,!.!.!.!Lindstrom!T.!(2005).!Discovery!of!LY2140023,! a! peptide! prodrug! of! the! mGlu2/3! receptor! agonist! LY404039.!Neuropharmacology,!49:!89.!!

Monyer! H,! Sprengel! R,! Schoepfer! R,! Herb! A,! Higuchi! M,! Lomeli! H,! .! .! .! Seeburg! PH.! (1992).!Heteromeric! NMDA! receptors:! Molecular! and! functional! distinction! of! subtypes.! Science,!256(5060):!1217*1221.!!

Moore! JH.! (2004).! Computational! analysis! of! gene*gene! interactions! using! multifactor!dimensionality!reduction.!Expert!Review!of!Molecular!Diagnostics,!4(6):!795*803.!!

Moore!TA,!Buchanan!RW,!Buckley!PF,!Chiles!JA,!Conley!RR,!Crismon!ML,!.! .! .!Miller!AL.!(2007).!The!Texas! medication! algorithm! project! antipsychotic! algorithm! for! schizophrenia:! 2006! update.!The!Journal!of!Clinical!Psychiatry,!68(11):!1751*1762.!!

Morel! BA.! (1860).!Traité! des! maladies! mentales.![Treatise! on! mental! diseases].! Paris,! France:!Masson.!

Morris! RG,! Anderson! E,! Lynch! GS! &! Baudry! M.! (1986).! Selective! impairment! of! learning! and!blockade! of! long*term! potentiation! by! an! N*methyl*D*aspartate! receptor! antagonist,! AP5.!Nature,!319(6056):!774*776.!!

Mortensen!PB,!Norgaard*Pedersen!B,!Waltoft!BL,!Sorensen!TL,!Hougaard!D,!Torrey!EF!&!Yolken!RH.!(2007).!Toxoplasma!gondii!as!a!risk!factor!for!early*onset!schizophrenia:!Analysis!of!filter!paper!blood!samples!obtained!at!birth.!Biological!Psychiatry,!61(5):!688*693.!!

Mortensen!PB,!Pedersen!CB,!Westergaard!T,!Wohlfahrt!J,!Ewald!H,!Mors!O,!.!.! .!Melbye!M.!(1999).!Effects!of! family!history!and!place!and!season!of!birth!on! the! risk!of! schizophrenia.! The!New!England!Journal!of!Medicine,!340(8):!603*608.!!

Mortimer!AM.!(2007).!Symptom!rating!scales!and!outcome!in!schizophrenia.!The!British!Journal!of!Psychiatry,!Supplemental,!50:!S7*S14.!!

Motsinger! AA,! Ritchie! MD! &! Reif! DM.! (2007).! Novel! methods! for! detecting! epistasis! in!pharmacogenomics!studies.!Pharmacogenomics,!8(9):!1229*1241.!!

Mouri!A,!Noda!Y,!Enomoto!T!&!Nabeshima!T.!(2007).!Phencyclidine!animal!models!of!schizophrenia:!Approaches! from! abnormality! of! glutamatergic! neurotransmission! and! neurodevelopment.!Neurochemistry!International,!51(2*4):!173*184.!!

155!!

!!

Mueller!HT,!Haroutunian!V,!Davis!KL!&!Meador*Woodruff! JH.! (2004).!Expression!of! the! ionotropic!glutamate! receptor! subunits! and! NMDA! receptor*associated! intracellular! proteins! in! the!substantia!nigra!in!schizophrenia.!Molecular!Brain!Research,!121(1*2):!60*69.!!

Mullis!K,!Faloona!F,!Scharf!S,!Saiki!R,!Horn!G!&!Erlich!H.!(1986).!Specific!enzymatic!amplification!of!DNA! in! vitro:! The! polymerase! chain! reaction.! Cold! Spring! Harbor! Symposia! on! Quantitative!Biology,!51(1):!263*273.!!

Murray!C!&!Lopez!A.! (Eds.).! (1996).!The!global!burden!of!disease:!A!comprehensive!assessment!of!mortality,! injuries,! and! risk! factors! in! 1990! and! projected! to! 2020! (Vol! 1).! Cambridge,! MA:!Harvard!University!Press.!!

Myles*Worsley!M,!Tiobech!J,!Browning!SR,!Korn!J,!Goodman!S,!Gentile!K,!.!.!.!Middleton!FA.!(2013).!Deletion! at! the! SLC1A1! glutamate! transporter! gene! co*segregates! with! schizophrenia! and!bipolar!schizoaffective!disorder!in!a!5*generation!family.!American!Journal!of!Medical!Genetics!Part!B,!Neuropsychiatric!Genetics,!162B(2):!87*95.!!

Nabeshima!T,!Fukaya!H,!Yamaguchi!K,!Ishikawa!K,!Furukawa!H!&!Kameyama!T.!(1987).!Development!of! tolerance! and! supersensitivity! to! phencyclidine! in! rats! after! repeated! administration! of!phencyclidine.!European!Journal!of!Pharmacology,!135(1):!23*33.!!

Nagai! Y,! Ohnuma! T,! Karibe! J,! Shibata! N,! Maeshima! H,! Baba! H,! .! .! .! Arai! H.! (2009).! No! genetic!association!between!the!SLC1A2!gene!and!Japanese!patients!with!schizophrenia.!Neuroscience!Letters,!463(3):!223*227.!!

Nakamura!K,!Kawasaki!Y,!Takahashi!T,!Furuichi!A,!Noguchi!K,!Seto!H!&!Suzuki!M.! (2012).!Reduced!white! matter! fractional! anisotropy! and! clinical! symptoms! in! schizophrenia:! A! voxel*based!diffusion!tensor!imaging!study.!Psychiatry!Research,!202(3):!233*238.!!

Nakatsu! Y,! Tyndale! RF,! DeLorey! TM,! Durham*Pierre! D,! Gardner! JM,! McDanel! HJ,! .! .! .! Sikela! JM.!(1993).!A!cluster!of!three!GABAA!receptor!subunit!genes!is!deleted!in!a!neurological!mutant!of!the!mouse!p!locus.!Nature,!364(6436):!448*450.!

Nakazawa! K,! Zsiros! V,! Jiang! Z,! Nakao! K,! Kolata! S,! Zhang! S! &! Belforte! JE.! (2012).! GABAergic!interneuron!origin!of!schizophrenia!pathophysiology.!Neuropharmacology,!62(3):!1574*1583.!!

Nanko! S,! Gill! M,! Owen! M,! Takazawa! N,! Moridaira! J! &! Kazamatsuri! H.! (1992).! Linkage! study! of!schizophrenia! with! markers! on! chromosome! 11! in! two! Japanese! pedigrees.! The! Japanese!Journal!of!Psychiatry!and!Neurology,!46(1):!155*159.!!

Neale!BM!&!Sham!PC.!(2004).!The!future!of!association!studies:!Gene*based!analysis!and!replication.!American!Journal!of!Human!Genetics,!75(3):!353*362.!!

Neeman!G,!Blanaru!M,!Bloch!B,!Kremer!I,!Ermilov!M,!Javitt!DC!&!Heresco*Levy!U.!(2005).!Relation!of!plasma! glycine,! serine,! and! homocysteine! levels! to! schizophrenia! symptoms! and!medication!type.!The!American!Journal!of!Psychiatry,!162(9):!1738*1740.!!

156!!

!!

Newman*Tancredi! A,! Chaput! C,! Verriele! L! &!Millan!M.! J.! (1996).! Clozapine! is! a! partial! agonist! at!cloned,!human!serotonin!5*HT1A!receptors.!Neuropharmacology,!35(1):!119*121.!!

Niciu!MJ,! Kelmendi! B! &! Sanacora! G.! (2012).! Overview! of! glutamatergic! neurotransmission! in! the!nervous!system.!Pharmacology,!Biochemistry,!and!Behavior,!100(4):!656*664.!!

Nielsen!DM,!Ehm!MG!&!Weir!BS.!(1998).!Detecting!marker*disease!association!by!testing!for!Hardy*Weinberg!disequilibrium!at!a!marker!locus.!American!Journal!of!Human!Genetics,!63(5):!1531*1540.!!

Ninan! I,! Jardemark!KE!&!Wang!RY.! (2003).!Differential!effects!of!atypical!and!typical!antipsychotic!drugs!on!N*methyl*D*aspartate*!and!electrically!evoked!responses!in!the!pyramidal!cells!of!the!rat!medial!prefrontal!cortex.!Synapse,!48(2):!66*79.!!

Noda!Y,!Kamei!H,!Mamiya!T,!Furukawa!H!&!Nabeshima!T.!(2000).!Repeated!phencyclidine!treatment!induces! negative! symptom*like! behavior! in! forced! swimming! test! in! mice:! Imbalance! of!prefrontal! serotonergic! and! dopaminergic! functions.! Neuropsychopharmacology,! 23(4):! 375*387.!!

Noda! Y,! Yamada! K,! Furukawa! H! &! Nabeshima! T.! (1995).! Enhancement! of! immobility! in! a! forced!swimming! test! by! subacute! or! repeated! treatment! with! phencyclidine:! A! new! model! of!schizophrenia.!British!Journal!of!Pharmacology,!116(5):!2531*2537.!!

Nothen! MM,! Rietschel! M,! Erdmann! J,! Oberlander! H,! Moller! HJ,! Nober! D! &! Propping! P.! (1995).!Genetic! variation!of! the!5*HT2A! receptor!and! response! to! clozapine.! Lancet,!346(8979):!908*909.!!

Nowak!L,!Bregestovski!P,!Ascher!P,!Herbet!A!&!Prochiantz!A.! (1984).!Magnesium!gates!glutamate*activated!channels!in!mouse!central!neurones.!Nature,!307(5950):!462*465.!!

Nudmamud*Thanoi!S!&!Reynold!GP.! (2004).!The!NR1!subunit!of! the!glutamate/NMDA!receptor! in!the! superior! temporal! cortex! in! schizophrenia! and! affective! disorders.! Neuroscience! Letters,!372(1*2):!173*177.!!

Numakawa!T,!Yagasaki!Y,!Ishimoto!T,!Okada!T,!Suzuki!T,!Iwata!N,!.!.!.!Hashimoto!R.!(2004).!Evidence!of! novel! neuronal! functions! of! dysbindin,! a! susceptibility! gene! for! schizophrenia.! Human!Molecular!Genetics,!13(21):!2699*2708.!!

Nyholt!DR.! (2004).!A!simple!correction! for!multiple! testing! for!single*nucleotide!polymorphisms! in!linkage!disequilibrium!with!each!other.!American!Journal!of!Human!Genetics,!74(4):!765*769.!!

Odom*White!A!&!de!Leon!J.!(1996).!Clozapine!levels!and!caffeine.!The!Journal!of!Clinical!Psychiatry,!57(4):!175*176.!!

Ohnuma!T,!Augood!SJ,!Arai!H,!McKenna!PJ!&!Emson!PC.!(1998).!Expression!of!the!human!excitatory!amino! acid! transporter! 2! and! metabotropic! glutamate! receptors! 3! and! 5! in! the! prefrontal!

157!!

!!

cortex! from! normal! individuals! and! patients! with! schizophrenia.! Molecular! Brain! Research,!56(1*2):!207*217.!!

Ohtsuki!T,!Sakurai!K,!Dou!H,!Toru!M,!Yamakawa*Kobayashi!K!&!Arinami!T.!(2001).!Mutation!analysis!of!the!NMDAR2B!(GRIN2B)!gene!in!schizophrenia.!Molecular!Psychiatry,!6(2):!211*216.!!

Olney! JW! &! Farber! NB.! (1995).! Glutamate! receptor! dysfunction! and! schizophrenia.! Archives! of!General!Psychiatry,!52(12):!998*1007.!!

Olney! JW,! Newcomer! JW! &! Farber! NB.! (1999).! NMDA! receptor! hypofunction! model! of!schizophrenia.!Journal!of!Psychiatric!Research,!33(6):!523*533.!!

Omkumar! RV,! Kiely! MJ,! Rosenstein! AJ,! Min! KT! &! Kennedy! MB.! (1996).! Identification! of! a!phosphorylation!site! for!calcium/calmodulin*dependent!protein!kinase! II! in! the!NR2B!subunit!of! the! N*methyl*D*aspartate! receptor.! The! Journal! of! Biological! Chemistry,! 271(49):! 31670*31678.!!

Ong!M,!Carreira!S,!Goodall!J,!Mateo!J,!Figueiredo!I,!Rodrigues!DN,!.!.!.!de!Bono!JS.!(2014).!Validation!and! utilisation! of! high*coverage! next*generation! sequencing! to! deliver! the! pharmacological!audit!trail.!British!Journal!of!Cancer,!111(5):!828*836.!!

O'Shea!RD.! (2002).!Roles! and! regulation!of! glutamate! transporters! in! the! central! nervous! system.!Clinical!and!Experimental!Pharmacology!&!Physiology,!29(11):!1018*1023.!!

Ossowska! K,! Pietraszek! M,! Wardas! J,! Nowak! G! &! Wolfarth! S.! (1999).! Chronic! haloperidol! and!clozapine! administration! increases! the! number! of! cortical! NMDA! receptors! in! rats.! NaunynPSchmiedeberg's!Archives!of!Pharmacology,!359(4):!280*287.!!

Overall!EJ!&!Gorham!DR.! (1962).!The!Brief!Psychiatric!Rating!Scale.!Psychological!Reports!10:!799*812.!!

Owen! RR! Jr,! Beake! BJ,!Marby! D,! Dessain! EC!&! Cole! JO.! (1989).! Response! to! clozapine! in! chronic!psychotic!patients.!Psychopharmacology!Bulletin,!25(2):!253*256.!!

Ozeki! Y,! Tomoda! T,! Kleiderlein! J,! Kamiya! A,! Bord! L,! Fujii! K,! .! .! .! Sawa! A.! (2003).! Disrupted*in*schizophrenia*1! (DISCP1):! Mutant! truncation! prevents! binding! to! NudE*like! (NUDEL)! and!inhibits! neurite! outgrowth.! Proceedings! of! the! National! Academy! of! Sciences! of! the! United!States!of!America,!100(1):!289*294.!!

Palego!L,!Biondi!L,!Giannaccini!G,!Sarno!N,!Elmi!S,!Ciapparelli!A,!.!.!.!Dell'Osso!L.!(2002a).!Clozapine,!norclozapine!plasma!levels,!their!sum!and!ratio! in!50!psychotic!patients:! Influence!of!patient*related! variables.! Progress! in!NeuroPPsychopharmacology!&!Biological! Psychiatry,! 26(3):! 473*480.!!

Palmer! BA,! Pankratz! VS! &! Bostwick! JM.! (2005).! The! lifetime! risk! of! suicide! in! schizophrenia:! A!reexamination.!Archives!of!General!Psychiatry,!62(3):!247*253.!!

158!!

!!

Park!S,! Jung!SW,!Kim!BN,!Cho!SC,!Shin!MS,!Kim!JW,! .! .! .!Kim!HW.!(2013).!Association!between!the!GRM7! rs3792452! polymorphism! and! attention! deficit! hyperacitiveity! disorder! in! a! Korean!sample.!Behavioral!and!Brain!Functions,!9(9):!1*8.!

Parker! JG,! Zweifel! LS,! Clark! JJ,! Evans! SB,! Phillips! PE! &! Palmiter! RD.! (2010).! Absence! of! NMDA!receptors! in! dopamine! neurons! attenuates! dopamine! release! but! not! conditioned! approach!during!pavlovian!conditioning.!Proceedings!of!the!National!Academy!of!Sciences!of!the!United!States!of!America,!107(30):!13491*13496.!!

Parliament!of!Canada.!(2011).!Canadian!Human!Rights!Act!Amendment!(Genetic!Characteristics).!Bill!C*445:! The! Government! of! Canada.! http://www.parl.gc.ca/content/hoc/Bills/412/Private/C*445/C*445_1/C*445_1.pdf!

Parliament!of!Canada.!(2013).!Genetic!Nondiscrimination!Act.!Bill!S*201:!The!Government!of!Canada.!http://www.parl.gc.ca/content/hoc/Bills/412/Private/S*201/S*201_1/S*201_1.pdf!!

Parsey!RV,!Olvet!DM,!Oquendo!MA,!Huang!YY,!Ogden!RT!&!Mann!JJ.!(2006).!Higher!5*HT1A!receptor!binding! potential! during! a! major! depressive! episode! predicts! poor! treatment! response:!Preliminary!data!from!a!naturalistic!study.!Neuropsychopharmacology,!31(8):!1745*1749.!!

Paschou!P,!Drineas!P,!Lewis!J,!Nievergelt!CM,!Nickerson!DA,!Smith!JD,!.!.!.!Ziv!E.!(2008).!Tracing!sub*structure! in!the!European!American!population!with!PCA*informative!markers.!PLoS!Genetics,!4(7):!e1000114.!!

Patil!ST,!Zhang!L,!Martenyi!F,!Lowe!SL,!Jackson!KA,!Andreev!BV,!.!.!.!Schoepp!DD.!(2007).!Activation!of!mGlu2/3!receptors!as!a!new!approach!to!treat!schizophrenia:!A!randomized!phase!2!clinical!trial.!Nature!Medicine,!13(9):!1102*1107.!!

Patnala!R,!Clements!J!&!Batra!J.!(2013).!Candidate!gene!association!studies:!A!comprehensive!guide!to!useful!in!silico!tools.!BMC!Genetics,!14(39):!1*11.!

Pearce! IA,! Cambray*Deakin! MA! &! Burgoyne! RD.! (1987).! Glutamate! acting! on! NMDA! receptors!stimulates!neurite!outgrowth!from!cerebellar!granule!cells.!FEBS!Letters,!223(1):!143*147.!!

Peduzzi! P,! Concato! J,! Feinstein! AR! &! Holford! TR.! (1995).! Importance! of! events! per! independent!variable! in! proportional! hazards! regression! analysis.! II.! Accuracy! and! precision! of! regression!estimates.!Journal!of!Clinical!Epidemiology,!48(12):!1503*1510.!!

Peduzzi!P,!Concato!J,!Kemper!E,!Holford!TR!&!Feinstein!AR.!(1996).!A!simulation!study!of!the!number!of! events! per! variable! in! logistic! regression! analysis.! Journal! of! Clinical! Epidemiology,! 49(12):!1373*1379.!!

Pennacchio!LA,!Ahituv!N,!Moses!AM,!Prabhakar!S,!Nobrega!MA,!Shoukry!M,!.!.!.!Rubin!EM.!(2006).!In!vivo! enhancer! analysis! of! human! conserved! non*coding! sequences.! Nature,! 444(7118):! 499*502.!!

159!!

!!

Perkins!DO,!Gu!H,! Boteva! K!&! Lieberman! JA.! (2005).! Relationship! between!duration!of! untreated!psychosis!and!outcome!in!first*episode!schizophrenia:!A!critical!review!and!meta*analysis.!The!American!Journal!of!Psychiatry,!162(10):!1785*1804.!!

Perneger! TV.! (1998).! What's! wrong! with! Bonferroni! adjustments.! BMJ! (Clinical! Research! Ed),!316(7139):!1236*1238.!!

Phillips!KA,!Veenstra!DL,!Oren!E,!Lee!JK!&!Sadee!W.!(2001).!Potential!role!of!pharmacogenomics!in!reducing!adverse!drug!reactions:!A!systematic!review.!Jama,!286(18):!2270*2279.!!

Phillips!PC.!(2008).!Epistasis!*!The!essential!role!of!gene!interactions!in!the!structure!and!evolution!of!genetic!systems.!Nature!Reviews,!Genetics,!9(11):!855*867.!!

Pickar!D,!Owen!RR,!Litman!RE,!Konicki!E,!Gutierrez!R!&!Rapaport!MH.! (1992).!Clinical!and!biologic!response!to!clozapine!in!patients!with!schizophrenia.!Crossover!comparison!with!fluphenazine.!Archives!of!General!Psychiatry,!49(5):!345*353.!!

Pilowsky!LS,!Bressan!RA,!Stone!JM,!Erlandsson!K,!Mulligan!RS,!Krystal!JH!&!Ell!PJ.!(2006).!First!in!vivo!evidence! of! an! NMDA! receptor! deficit! in! medication*free! schizophrenic! patients.! Molecular!Psychiatry,!11(2):!118*119.!!

Pin! JP! &! Duvoisin! R.! (1995).! The! metabotropic! glutamate! receptors:! Structure! and! functions.!Neuropharmacology,!34(1):!1*26.!!

Pin!JP,!Galvez!T!&!Prezeau!L.!(2003).!Evolution,!structure,!and!activation!mechanism!of!family!3/C!G*protein*coupled!receptors.!Pharmacology!&!Therapeutics,!98(3):!325*354.!!

Pinard!E,!Alanine!A,!Alberati!D,!Bender!M,!Borroni!E,!Bourdeaux!P,!.!.!.!Zimmerli!D.!(2010).!Selective!GlyT1! inhibitors:! Discovery! of! [4*(3*fluoro*5*trifluoromethylpyridin*2*yl)piperazin*1*yl][5*methanesulfonyl*2*((S)*2,2,2*trifluoro*1*methylethoxy)phenyl]methanone! (RG1678),! a!promising!novel!medicine!to!treat!schizophrenia.!Journal!of!Medicinal!Chemistry,!53(12):!4603*4614.!!

Pique*Regi!R,!Degner! JF,! Pai!AA,!Gaffney!DJ,!Gilad!Y!&!Pritchard! JK.! (2011).!Accurate! inference!of!transcription! factor! binding! from! DNA! sequence! and! chromatin! accessibility! data.! Genome!Research,!21(3):!447*455.!!

Plomen! R,! DeFries! JC,!McClearn! GE! &! Rutter!M.! (1997).!Behavioral! genetics! (3rd! ed).! New! York:!Freeman.!!

Potkin! SG,! Costa! J,! Roy! S,! Sramek! J,! Jin! Y! &! Gulasekaram! B.! (1992).! Glycine! in! the! treatment! of!schizophrenia!*!Theory!and!preliminary!results.! In!HY!Meltzer!(Ed),!Novel!antipsychotic!drugs.!New!York:!Raven!Press!Ltd.!!

Price!AL,!Butler!J,!Patterson!N,!Capelli!C,!Pascali!VL,!Scarnicci!F,!.!.!.!Hirschhorn!JN.!(2008).!Discerning!the!ancestry!of!European!Americans!in!genetic!association!studies.!PLoS!Genetics,!4(1):!e236.!!

160!!

!!

Pritchard! JK.,!Stephens!M,!Rosenberg!NA!&!Donnelly!P.! (2000).!Association!mapping! in!structured!populations.!American!Journal!of!Human!Genetics,!67(1):!170*181.!!

Puckett!C,!Gomez!CM,!Korenberg!JR,!Tung!H,!Meier!TJ,!Chen!XN!&!Hood!L.!(1991).!Molecular!cloning!and!chromosomal! localization!of!one!of!the!human!glutamate!receptor!genes.!Proceedings!of!the!National!Academy!of!Sciences!of!the!United!States!of!America,!88(17):!7557*7561.!!

Pulver!AE,!Lasseter!VK,!Kasch!L,!Wolyniec!P,!Nestadt!G,!Blouin!JL,!.!.!.!Chen!H.!(1995).!Schizophrenia:!A! genome! scan! targets! chromosomes! 3p! and! 8p! as! potential! sites! of! susceptibility! genes.!American!Journal!of!Medical!Genetics,!60(3):!252*260.!!

Purcell!S,!Neale!B,!Todd*Brown!K,!Thomas!L,!Ferreira!MA,!Bender!D,!.!.!.!Sham!PC.!(2007).!PLINK:!A!tool! set! for! whole*genome! association! and! population*based! linkage! analyses.! American!Journal!of!Human!Genetics,!81(3):!559*575.!!

Purkayastha! S,! Ford! J,! Kanjilal! B,!Diallo! S,!Del!Rosario! Inigo! J,!Neuwirth! L,! .! .! .! Banerjee!P.! (2012).!Clozapine!functions!through!the!prefrontal!cortex!serotonin!1A!receptor!to!heighten!neuronal!activity! via! calmodulin! kinase! II*NMDA! receptor! interactions.! Journal! of! Neurochemistry,!120(3):!396*407.!!

Pycock! CJ,! Kerwin! RW! &! Carter! CJ.! (1980).! Effect! of! lesion! of! cortical! dopamine! terminals! on!subcortical!dopamine!receptors!in!rats.!Nature,!286(5768):!74*76.!!

Qin!S,!Zhao!X,!Pan!Y,!Liu!J,!Feng!G,!Fu!J,! .! .! .!He!L.!(2005).!An!association!study!of!the!N*methyl*D*aspartate! receptor! NR1! subunit! gene! (GRIN1)! and! NR2B! subunit! gene! (GRIN2B)! in!schizophrenia!with!universal!DNA!microarray.!European!Journal!of!Human!Genetics,!13(7):!807*814.!!

Reicher!A,!Maurer!K,!Loffler!W,!Fatkenheuer!B,!van!der!Heiden!W,!Munk*Jorgenson!P,!.!.!Hafner!H.!(1990).!Gender!differences!in!age!at!onset!and!course!of!schizophrenic!disorders.!In!H!Hafner!&!W!Gattaz!(Eds),!Search!for!the!causes!of!schizophrenia.!Berlin:!Springer*Verlag.!!

Remington!G,!Saha!A,!Chong!SA!&!Shammi!C.!(2005).!Augmentation!strategies!in!clozapine*resistant!schizophrenia.!CNS!Drugs,!19(10):!843*872.!!

Riley!BP!&!McGuffin!P.!(2000).!Linkage!and!associated!studies!of!schizophrenia.!American!Journal!of!Medical!Genetics,!97(1):!23*44.!!

Riley! B! &! Kendler! KS.! (2006).! Molecular! genetic! studies! of! schizophrenia.! European! Journal! of!Human!Genetics,!14(6):!669*680.!!

Risch!N!&!Merikangas!K.!(1996).!The!future!of!genetic!studies!of!complex!human!diseases.!Science,!273(5281):!1516*1517.!!

Ritchie!MD,! Hahn! LW,! Roodi! N,! Bailey! LR,! Dupont!WD,! Parl! FF! &!Moore! JH.! (2001).!Multifactor*dimensionality!reduction!reveals!high*order!interactions!among!estrogen*metabolism!genes!in!sporadic!breast!cancer.!American!Journal!of!Human!Genetics,!69(1):!138*147.!!

161!!

!!

Ritchie!MD!&!Motsinger!AA.! (2005).!Multifactor!dimensionality! reduction! for!detecting!gene*gene!and! gene*environment! interactions! in! pharmacogenomics! studies.! Pharmacogenomics,! 6(8):!823*834.!!

Rodriguez*Moreno!A.! (2006).!The!role!of!kainate!receptors! in!the!regulation!of!excitatory!synaptic!transmission!in!the!hippocampus.!Revista!De!Neurologia,!42(5):!282*287.!!

Rorick*Kehn!LM,!Johnson!BG,!Burkey!JL,!Wright!RA,!Calligaro!DO,!Marek!GJ,!.!.!.!Schoepp!DD.!(2007).!Pharmacological!and!pharmacokinetic!properties!of!a!structurally!novel,!potent,!and!selective!metabotropic! glutamate! 2/3! receptor! agonist:! In! vitro! characterization! of! agonist! (*)*(1R,4S,5S,6S)*4*amino*2*sulfonylbicyclo[3.1.0]*hexane*4,6*dicarboxylic! acid! (LY404039).! The!Journal!of!Pharmacology!and!Experimental!Therapeutics,!321(1):!308*317.!!

Rosenheck!R,!Evans!D,!Herz!L,!Cramer!J,!Xu!W,!Thomas!J,!.!.!.!Charney!D.!(1999).!How!long!to!wait!for!a! response! to! clozapine:! A! comparison! of! time! course! of! response! to! clozapine! and!conventional! antipsychotic! medication! in! refractory! schizophrenia.! Schizophrenia! Bulletin,!25(4):!709*719.!!

Rosenthal!D,!Werder! PH,! Kety! SS,! Schulsinger! F,!Welner! J,!Ostergaard! L,! .! .! .! Kety! SS.! (1968).!The!transmission!of!schizophrenia.!Oxford:!Pergamon!Press.!!

Ross! JS! &! Cronin! M.! (2011).! Whole! cancer! genome! sequencing! by! next*generation! methods.!American!Journal!of!Clinical!Pathology,!136(4):!527*539.!!

Rothstein!JD,!Dykes*Hoberg!M,!Pardo!CA,!Bristol!LA,!Jin!L,!Kuncl!RW,!.!.!.!Welty!DF.!(1996).!Knockout!of! glutamate! transporters! reveals! a! major! role! for! astroglial! transport! in! excitotoxicity! and!clearance!of!glutamate.!Neuron,!16(3):!675*686.!!

Rubin! P,! Holm! S,! Friberg! L,! Videbech! P,! Andersen! HS,! Bendsen! BB,! .! .! .! Hemmingsen! R.! (1991).!Altered! modulation! of! prefrontal! and! subcortical! brain! activity! in! newly! diagnosed!schizophrenia!and!schizophreniform!disorder.!A!regional!cerebral!blood!flow!study.!Archives!of!General!Psychiatry,!48(11):!987*995.!!

Sadee! W.! (2012).! The! relevance! of! "missing! heritability"! in! pharmacogenomics.! Clinical!Pharmacology!and!Therapeutics,!92(4):!428*430.!!

Sadee!W.!(2013).!Gene*gene*environment!interactions!between!drugs,!transporters,!receptors,!and!metabolizing!enzymes:!Statins,!SLCO1B1,!and!CYP3A4!as!an!example.!Journal!of!Pharmaceutical!Sciences,!102(9):!2924*2929.!

Saha! S,! Chant! D! &! McGrath! J.! (2007).! A! systematic! review! of! mortality! in! schizophrenia:! Is! the!differential!mortality!gap!worsening!over! time?!Archives!of!General!Psychiatry,!64(10):!1123*1131.!

Sala! C,! Rudolph*Correia! S! &! Sheng! M.! (2000).! Developmentally! regulated! NMDA! receptor*dependent! dephosphorylation! of! cAMP! response! element*binding! protein! (CREB)! in!hippocampal!neurons.!The!Journal!of!Neuroscience,!20(10):!3529*3536.!!

162!!

!!

Salmi!P,!Chergui!K!&!Fredholm!BB.! (2005).!Adenosine*dopamine! interactions!revealed! in!knockout!mice.!Journal!of!Molecular!Neuroscience,!26(2*3):!239*244.!!

Sams*Dodd!F.!(1995).!Distinct!effects!of!d*amphetamine!and!phencyclidine!on!the!social!behaviour!of!rats.!Behavioural!Pharmacology,!6(1):!55*65.!!

Sams*Dodd! F.! (1998).! Effects! of! continuous! D*amphetamine! and! phencyclidine! administration! on!social! behaviour,! stereotyped! behaviour,! and! locomotor! activity! in! rats.!Neuropsychopharmacology,!19(1):!18*25.!!

Sanger! F,! Nicklen! S! &! Coulson! AR.! (1977).! DNA! sequencing! with! chain*terminating! inhibitors.!Proceedings!of!the!National!Academy!of!Sciences!of!the!United!States!of!America,!74(12):!5463*5467.!!

Santos! SD,! Carvalho! AL,! Caldeira! MV! &! Duarte! CB.! (2009).! Regulation! of! AMPA! receptors! and!synaptic!plasticity.!Neuroscience,!158(1):!105*125.!!

Sasieni! PD.! (1997).! From! genotypes! to! genes:! Doubling! the! sample! size.! Biometrics,! 53(4):! 1253*1261.!!

Schene!AH,!van!Wijngaarden!B!&!Koeter!MW.!(1998).!Family!caregiving! in!schizophrenia:!Domains!and!distress.!Schizophrenia!Bulletin,!24(4):!609*618.!!

Schiller! J,! Schiller! Y! &! Clapham!DE.! (1998).! NMDA! receptors! amplify! calcium! influx! into! dendritic!spines!during!associative!pre*!and!postsynaptic!activation.!Nature!Neuroscience,!1(2):!114*118.!!

Schizophrenia!Linkage!Collaborative!Group.!(1996).!Additional!support!for!schizophrenia!linkage!on!chromosomes!6!and!8:!A!multicenter!study.!American!Journal!of!Medical!Genetics,!67(6):!580*594.!

Schizophrenia!Working! Group! of! the! Psychiatric! Genomics! Consortium.! (2014).! Biological! insights!from!108!schizophrenia*associated!genetic!loci.!Nature,!511(7510):!421*427.!!

Schmitt! A,! Zink!M,!Muller! B,!May! B,! Herb! A,! Jatzko! A,! .! .! .! Henn! FA.! (2003).! Effects! of! long*term!antipsychotic! treatment! on! NMDA! receptor! binding! and! gene! expression! of! subunits.!Neurochemical!Research,!28(2):!235*241.!!

Schork!NJ,!Murray!SS,!Frazer!KA!&!Topol!EJ.!(2009).!Common!vs.!rare!allele!hypotheses!for!complex!diseases.!Current!Opinion!in!Genetics!&!Development,!19(3):!212*219.!!

Schotte!A,!Janssen!PF,!Gommeren!W,!Luyten!WH,!Van!Gompel!P,!Lesage!AS,!.!.!.!Leysen!JE.!(1996).!Risperidone! compared! with! new! and! reference! antipsychotic! drugs:! In! vitro! and! in! vivo!receptor!binding.!Psychopharmacology,!124(1*2):!57*73.!!

Schumacher!J,!Schulze!TG,!Wienker!TF,!Rietschel!M!&!Nothen!MM.!(2000).!Pharmacogenetics!of!the!clozapine!response.!Lancet,!356(9228):!506*507.!!

163!!

!!

Schumacher! J,! Jamra! RA,! Freudenberg! J,! Becker! T,! Ohlraun! S,! Otte! AC,! .! .! .! Cichon! S.! (2004).!Examination! of! G72! and! D*amino*acid! oxidase! as! genetic! risk! factors! for! schizophrenia! and!bipolar!affective!disorder.!Molecular!Psychiatry,!9(2):!203*207.!!

Schwieler!L,!Engberg!G!&!Erhardt!S.!(2004).!Clozapine!modulates!midbrain!dopamine!neuron!firing!via!interaction!with!the!NMDA!receptor!complex.!Synapse,!52(2):!114*122.!!

Schwieler! L! &! Erhardt! S.! (2003).! Inhibitory! action! of! clozapine! on! rat! ventral! tegmental! area!dopamine! neurons! following! increased! levels! of! endogenous! kynurenic! acid.!Neuropsychopharmacology,!28(10):!1770*1777!!

Schwieler!L,!Linderholm!KR,!Nilsson*Todd!LK,!Erhardt!S!&!Engberg!G.!(2008).!Clozapine!interacts!with!the!glycine!site!of!the!NMDA!receptor:!Electrophysiological!studies!of!dopamine!neurons!in!the!rat!ventral!tegmental!area.!Life!Sciences,!83(5*6):!170*175.!!

Seeman! MV! &! Lang! M.! (1990).! The! role! of! estrogens! in! schizophrenia! gender! differences.!Schizophrenia!Bulletin,!16(2):!185*194.!!

Seeman! P! &! Lee! T.! (1975).! Antipsychotic! drugs:! Direct! correlation! between! clinical! potency! and!presynaptic!action!on!dopamine!neurons.!Science,!188(4194):!1217*1219.!!

Seeman! P.! (1987).! Dopamine! receptors! and! the! dopamine! hypothesis! of! schizophrenia.! Synapse,!1(2):!133*152.!!

Seppala!NH,! Leinonen!EV,! Lehtonen!ML!&!Kivisto! KT.! (1999).! Clozapine! serum! concentrations! are!lower! in! smoking! than! in! non*smoking! schizophrenic! patients.! Pharmacology! &! Toxicology,!85(5):!244*246.!!

Serretti!A,!Drago!A!&!De!Ronchi!D.!(2007).!HTR2A!gene!variants!and!psychiatric!disorders:!A!review!of! current! literature! and! selection! of! SNPs! for! future! studies.! Current! Medicinal! Chemistry,!14(19):!2053*2069.!!

Setsirichok! D,! Piroonratana! T,! Assawamakin! A,! Usavanarong! T,! Limwongse! C,!Wongseree!W,! .! .! .!Chaiyaratana!N.! (2012).! Small! ancestry! informative!marker! panels! for! complete! classification!between! the! original! four! HapMap! populations.! International! Journal! of! Data! Mining! and!Bioinformatics,!6(6):!651*674.!!

Shan! D,! Yates! S,! Roberts! RC! &! McCullumsmith! RE.! (2012).! Update! on! the! neurobiology! of!schizophrenia:!A!role!for!extracellular!microdomains.!Minerva!Psichiatrica,!53(3):!233*249.!!

Shank! RP!&!Aprison!MH.! (1981).! Present! status! and! significance! of! the! glutamine! cycle! in! neural!tissues.!Life!Sciences,!28(8):!837*842.!!

Shao!L,! Shuai!Y,!Wang! J,! Feng!S,! Lu!B,! Li! Z,! .! .! .! Zhong!Y.! (2011).! Schizophrenia! susceptibility!gene!dysbindin! regulates! glutamatergic! and! dopaminergic! functions! via! distinctive!mechanisms! in!drosophila.!Proceedings!of!the!National!Academy!of!Sciences!of!the!United!States!of!America,!108(46):!18831*18836.!!

164!!

!!

Shapiro!M.! (2001).! Plasticity,! hippocampal!place! cells,! and! cognitive!maps.! Archives!of!Neurology,!58(6):!874*881.!!

Shastry!P,!Basu!A!&!Rajadhyaksha!MS.!(2001).!Neuroblastoma!cell!lines:!A!versatile!in!vitro!model!in!neurobiology.!The!International!Journal!of!Neuroscience,!108(1*2):!109*126.!!

Shaw!SH,!Kelly!M,!Smith!AB,!Shields!G,!Hopkins!PJ,!Loftus! J,! .! .! .!DeLisi!LE.! (1998).!A!genome*wide!search!for!schizophrenia!susceptibility!genes.!American!Journal!of!Medical!Genetics,!81(5):!364*376.!!

Shendure! J!&! Ji!H.! (2008).!Next*generation!DNA! sequencing.!Nature!Biotechnology,! 26(10):! 1135*1145.!!

Sigwald! J,!Bouttier!D,!Raymond!C!&!Pichot!C.! (1959).!Four!cases!of! facio*bucco*linguo*masticatory!dyskinesia! of! prolonged! development! following! treatment! with! neuroleptics.! Revue!Neurologique,!100:!751*755.!!

Singh!SP!&!Singh!V.!(2011).!Meta*analysis!of!the!efficacy!of!adjunctive!NMDA!receptor!modulators!in!chronic!schizophrenia.!CNS!Drugs,!25(10):!859*885.!!

Sipos!A,!Rasmussen!F,!Harrison!G,!Tynelius!P,!Lewis!G,!Leon!DA!&!Gunnell!D.! (2004).!Paternal!age!and! schizophrenia:! A! population! based! cohort! study.! BMJ! (Clinical! Research! Ed),! 329(7474):!1070.!

Smith!KE,!Borden!LA,!Hartig!PR,!Branchek!T!&!Weinshank!RL.! (1992).!Cloning!and!expression!of! a!glycine!transporter!reveal!colocalization!with!NMDA!receptors.!Neuron,!8(5):!927*935.!!

Solanki! RK,! Singh! P! &! Munshi! D.! (2009).! Current! perspectives! in! the! treatment! of! resistant!schizophrenia.!Indian!Journal!of!Psychiatry,!51(4):!254*260.!!

Solberg! N! &! Krauss! S.! (2013).! Luciferase! assay! to! study! the! activity! of! a! cloned! promoter! DNA!fragment.!Methods!in!Molecular!Biology,!977:!65*78.!!

Solinas! M,! Ferre! S,! You! ZB,! Karcz*Kubicha! M,! Popoli! P! &! Goldberg! SR.! (2002).! Caffeine! induces!dopamine! and! glutamate! release! in! the! shell! of! the! nucleus! accumbens.! The! Journal! of!Neuroscience,!22(15):!6321*6324.!!

Sommer!B,!Keinanen!K,!Verdoorn!TA,!Wisden!W,!Burnashev!N,!Herb!A,!.!.!.!Seeburg!PH.!(1990).!Flip!and!flop:!A!cell*specific!functional!switch!in!glutamate*operated!channels!of!the!CNS.!Science,!249(4976):!1580*1585.!!

Song! L! &! Crawford! GE.! (2010).! DNase*seq:! A! high*resolution! technique! for! mapping! active! gene!regulatory!elements!across! the!genome!from!mammalian!cells.!Cold!Spring!Harbor!Protocols,!2010(2):!1*13.!

165!!

!!

Spurney!CF,!Baca!SM,!Murray!AM,!Jaskiw!GE,!Kleinman!JE!&!Hyde!TM.!(1999).!Differential!effects!of!haloperidol!and!clozapine!on!ionotropic!glutamate!receptors! in!rats.!Synapse,!34(4):!266*276.!doi:2*2!!

St! Clair! D,! Blackwood! D,! Muir! W,! Carothers! A,! Walker! M,! Spowart! G,! .! .! .! Evans! HJ.! (1990).!Association!within! a! family! of! a! balanced! autosomal! translocation!with!major!mental! illness.!Lancet,!336(8706):!13*16.!!

St! Clair! D,! Xu!M,!Wang! P,! Yu! Y,! Fang! Y,! Zhang! F,! .! .! .! He! L.! (2005).! Rates! of! adult! schizophrenia!following!prenatal!exposure!to!the!Chinese!famine!of!1959*1961.!Jama,!294(5):!557*562.!!

Stahl!SM.!(2007).!Beyond!the!dopamine!hypothesis!to!the!NMDA!glutamate!receptor!hypofunction!hypothesis!of!schizophrenia.!CNS!Spectrums,!12(4):!265*268.!!

Steck!H.! (1954).!Le!syndrome!extrapyramidal!et!diencephalique!au! largactil!et!au!serpasil.!Annales!Médico!Psychologique,!112:!737*743.!!

Stefansson!H,!Sarginson!J,!Kong!A,!Yates!P,!Steinthorsdottir!V,!Gudfinnsson!E,!.!.!.!St!Clair!D.!(2003).!Association!of! neuregulin! 1!with! schizophrenia! confirmed! in! a! Scottish! population.! American!Journal!of!Human!Genetics,!72(1):!83*87.!!

Stern! RG,! Kahn! RS,! Davidson! M,! Nora! RM! &! Davis! KL.! (1994).! Early! response! to! clozapine! in!schizophrenia.!The!American!Journal!of!Psychiatry,!151(12):!1817*1818.!!

Stowell! JN! &! Craig! AM.! (1999).! Axon/dendrite! targeting! of!metabotropic! glutamate! receptors! by!their!cytoplasmic!carboxy*terminal!domains.!Neuron,!22(3):!525*536.!!

Straub! RE,! Jiang! Y,!MacLean! CJ,!Ma! Y,!Webb!BT,!Myakishev!MV,! .! .! .! Kendler! KS.! (2002).! Genetic!variation! in! the! 6p22.3! gene!DTNBP1,! the! human! ortholog! of! the!mouse! dysbindin! gene,! is!associated!with!schizophrenia.!American!Journal!of!Human!Genetics,!71(2):!337*348.!!

Straub!RE,!Lipska!BK,!Egan!MF,!Goldberg!TE,!Callicott!JH,!Mayhew!MB,!.!.! .!Weinberger!DR.!(2007).!Allelic! variation! in! GAD1! (GAD67)! is! associated! with! schizophrenia! and! influences! cortical!function!and!gene!expression.!Molecular!Psychiatry,!12(9):!854*869.!!

Sturgeon!RD,!Fessler!RG!&!Meltzer!HY.!(1979).!Behavioral!rating!scales!for!assessing!phencyclidine*induced! locomotor! activity,! stereotyped! behavior! and! ataxia! in! rats.! European! Journal! of!Pharmacology,!59(3*4):!169*179.!!

Sumiyoshi! T,!Anil!AE,! Jin!D,! Jayathilake!K,! Lee!M!&!Meltzer!HY.! (2004).! Plasma!glycine!and! serine!levels! in! schizophrenia! compared! to! normal! controls! and! major! depression:! Relation! to!negative!symptoms.!The!International!Journal!of!Neuropsychopharmacology,!7(1):!1*8.!!

Sun!T,!Hu!G!&!Li!M.! (2009).!Repeated!antipsychotic! treatment!progressively!potentiates! inhibition!on! phencyclidine*induced! hyperlocomotion,! but! attenuates! inhibition! on! amphetamine*induced! hyperlocomotion:! Relevance! to! animal! models! of! antipsychotic! drugs.! European!Journal!of!Pharmacology,!602(2*3):!334*342.!!

166!!

!!

Susser! ES!&! Lin! SP.! (1992).! Schizophrenia! after! prenatal! exposure! to! the! Dutch! hunger!winter! of!1944*1945.!Archives!of!General!Psychiatry,!49(12):!983*988.!!

Suzuki!T,!Remington!G,!Arenovich!T,!Uchida!H,!Agid!O,!Graff*Guerrero!A!&!Mamo!DC.!(2011).!Time!course! of! improvement! with! antipsychotic! medication! in! treatment*resistant! schizophrenia.!British!Journal!of!Psychiatry,!199(4):!275*280.!!

Suzuki! T,! Remington!G,!Mulsant! BH,!Uchida!H,! Rajji! TK,! Graff*Guerrero! A,! .! .! .!Mamo!DC.! (2012).!Defining! treatment*resistant! schizophrenia! and! response! to! antipsychotics:! A! review! and!recommendation.!Psychiatry!Research,!197(1*2):!1*6.!!

Swerdlow! NR,! Bakshi! V,! Waikar! M,! Taaid! N! &! Geyer! MA.! (1998).! Seroquel,! clozapine! and!chlorpromazine! restore! sensorimotor! gating! in! ketamine*treated! rats.! Psychopharmacology,!140(1):!75*80.!!

Szatkowski!M!&!Attwell!D.! (1994).!Triggering!and!execution!of!neuronal!death! in!brain! ischaemia:!Two!phases!of!glutamate!release!by!different!mechanisms.!Trends!in!Neurosciences,!17(9):!359*365.!!

Szczepankiewicz!A,! Skibinska!M,! Suwalska!A,!Hauser! J!&!Rybakowski! JK.! (2009).!No!association!of!three! GRIN2B! polymorphisms! with! lithium! response! in! bipolar! patients.! Pharmacological!Reports,!61(3):!448*452.!!

Takahashi! T,! Svoboda! K! &! Malinow! R.! (2003).! Experience! strengthening! transmission! by! driving!AMPA!receptors!into!synapses.!Science,!299(5612):!1585*1588.!!

Takahashi! M,! Kakita! A,! Futamura! T,! Watanabe! Y,! Mizuno! M,! Sakimura! K,! .! .! .! Nawa! H.! (2006).!Sustained! brain*derived! neurotrophic! factor! up*regulation! and! sensorimotor! gating!abnormality! induced! by! postnatal! exposure! to! phencyclidine:! Comparison! with! adult!treatment.!Journal!of!Neurochemistry,!99(3):!770*780.!!

Talbot! K,! Eidem! WL,! Tinsley! CL,! Benson! MA,! Thompson! EW,! Smith! RJ,! .! .! .! Arnold! SE.! (2004).!Dysbindin*1! is! reduced! in! intrinsic,! glutamatergic! terminals! of! the! hippocampal! formation! in!schizophrenia.!The!Journal!of!Clinical!Investigation,!113(9):!1353*1363.!!

Tandon!R!&!Fleischhacker!WW.!(2005).!Comparative!efficacy!of!antipsychotics! in!the!treatment!of!schizophrenia:!A!critical!assessment.!Schizophrenia!Research,!79(2*3):!145*155.!!

Tandon! R.! (2011).! Antipsychotics! in! the! treatment! of! schizophrenia:! An! overview.! The! Journal! of!Clinical!Psychiatry,!Supplemental,!72(S1):!4*8.!!

Taylor!SF!&!Tso!IF!(2014).!GABA!abnormalities!in!schizophrenia:!A!methodological!review!of!in!vivo!studies.!Schizophrenia!Research,!advanced!online!publication.!!

Theisen! FM,!Gebhardt! S,! Haberhausen!M,!Heinzel*Gutenbrunner!M,!Wehmeier! PM,! Krieg! JC,! .! .! .!Hebebrand!J.!(2005).!Clozapine*induced!weight!gain:!A!study!in!monozygotic!twins!and!same*sex!sib!pairs.!Psychiatric!Genetics,!15(4):!285*289.!!

167!!

!!

Thierry*Mieg! D! &! Thierry*Mieg! J.! (2006).! AceView:! A! comprehensive! cDNA*supported! gene! and!transcripts!annotation.!Genome!Biology,!Supplemental,!7(S1):!S12*S14.!!

Thomson!AM.! (1990).!Glycine! is! a! coagonist! at! the!NMDA! receptor/channel! complex.! Progress! in!Neurobiology,!35(1):!53*74.!!

Tian!L,!Stefanidakis!M,!Ning!L,!Van!Lint!P,!Nyman*Huttunen!H,!Libert!C,! .! .! .!Gahmberg!CG.!(2007).!Activation!of!NMDA!receptors!promotes!dendritic!spine!development!through!MMP*mediated!ICAM*5!cleavage.!The!Journal!of!Cell!Biology,!178(4):!687*700.!!

Tienari!PJ!&!Wynne!LC.! (1994).!Adoption!studies!of!schizophrenia.!Annals!of!Medicine,!26(4):!233*237.!!

Tingley! WG,! Ehlers! MD,! Kameyama! K,! Doherty! C,! Ptak! JB,! Riley! CT! &! Huganir! RL.! (1997).!Characterization!of!protein!kinase!A!and!protein!kinase!C!phosphorylation!of!the!N*methyl*D*aspartate! receptor!NR1!subunit!using!phosphorylation! site*specific!antibodies.! The! Journal!of!Biological!Chemistry,!272(8):!5157*5166.!!

Torrey! E.! (2013).! Surviving! schizophrenia:! A!manual! for! families,! patients,! and! providers! (5th! ed).!New!York,!NY:!Harper!Perrenial.!!

Tran!B,!Brown!AM,!Bedard!PL,!Winquist!E,!Goss!GD,!Hotte!SJ,! .! .! .!Dancey!JE.! (2013).!Feasibility!of!real!time!next!generation!sequencing!of!cancer!genes!linked!to!drug!response:!Results!from!a!clinical!trial.!International!Journal!of!Cancer,!132(7):!1547*1555.!!

Tsai! GE,! Yang! P,! Chung! LC,! Lange! N!&! Coyle! JT.! (1998).! D*serine! added! to! antipsychotics! for! the!treatment!of!schizophrenia.!Biological!Psychiatry,!44(11):!1081*1089.!!

Tsai!GE,!Yang!P,!Chung!LC,!Tsai!IC,!Tsai!CW!&!Coyle!JT.!(1999).!D*serine!added!to!clozapine!for!the!treatment!of!schizophrenia.!The!American!Journal!of!Psychiatry,!156(11):!1822*1825.!!

Tsunoka!T,!Kishi!T,!Ikeda!M,!Kitajima!T,!Yamanouchi!Y,!Kinoshita!Y,!.!.!.!Iwata!N.!(2009).!Association!analysis! of! group! II! metabotropic! glutamate! receptor! genes! (GRM2! and! GRM3)! with! mood!disorders! and! fluvoxamine! response! in! a! Japanese! population.! Progress! in! NeuroPPsychopharmacology!&!Biological!Psychiatry,!33(5):!875*879.!!

Tsunoka!T,!Kishi!T,!Kitajima!T,!Okochi!T,!Okumura!T,!Yamanouchi!Y,!.!.!.!Iwata!N.!(2010).!Association!analysis!of!GRM2!and!HTR2A!with!methamphetamine*induced!psychosis!and!schizophrenia!in!the! Japanese! population.! Progress! in! NeuroPPsychopharmacology! &! Biological! Psychiatry,!34(4):!639*644.!!

Tu! JC,! Xiao! B,! Naisbitt! S,! Yuan! JP,! Petralia! RS,! Brakeman! P,! .! .! .! Worley! PF.! (1999).! Coupling! of!mGluR/Homer! and! PSD*95! complexes! by! the! shank! family! of! postsynaptic! density! proteins.!Neuron,!23(3):!583*592.!!

United!Kingdom!of!Great!Britain.!(2010).!Equality!Act!of!2010:!Government!of!the!United!Kingdom.!http://www.legislation.gov.uk/ukpga/2010/15/pdfs/ukpga_20100015_en.pdf!

168!!

!!

United!States!of!America.! (2008).!Genetic! Information!Nondiscrimination!Act!of!2008! (GINA).!H.R.,!493:! Government! of! the! United! States! of! America.! http://www.gpo.gov/fdsys/pkg/BILLS*110hr493enr/pdf/BILLS*110hr493enr.pdf!

van!den!Oord!EJ!&!Sullivan!PF.! (2003).!False!discoveries!and!models! for!gene!discovery.! Trends! in!Genetics,!19(10):!537*542.!!

Van!den!Pol!AN.! (1994).!Metabotropic!glutamate!receptor!mGluR1!distribution!and!ultrastructural!localization!in!hypothalamus.!The!Journal!of!Comparative!Neurology,!349(4):!615*632.!!

Vasudeva! S,! Sekhar! CK! &! Rao! PG.! (2013).! Caregivers! burden! of! patients! with! schizophrenia! and!bipolar!disorder:!A!sectional!study.!Indian!Journal!of!Psychological!Medicine,!35(4):!352*357.!!

Vesell!ES.!(1978).!Twin!studies!in!pharmacogenetics.!Human!Genetics,!Supplemental,!(1)(1):!19*30.!!

Vink! JM,! Smit! AB,! de! Geus! EJ,! Sullivan! P,! Willemsen! G,! Hottenga! JJ,! .! .! .! Boomsma! DI.! (2009).!Genome*wide!association!study!of!smoking!initiation!and!current!smoking.!American!Journal!of!Human!Genetics,!84(3):!367*379.!!

Visscher! PM,! Hill! WG! &! Wray! NR.! (2008).! Heritability! in! the! genomics! era:! Concepts! and!misconceptions.!Nature!Reviews,!Genetics,!9(4):!255*266.!!

Vogel! F.! (1959).! Moderne! problem! der! humangenetik.! Ergebnisse! der! Inneren! Medizin! und!Kinderheilkunde,!12:!52*125.!!

Voineskos! AN,! Foussias! G,! Lerch! J,! Felsky! D,! Remington! G,! Rajji! TK,! .! .! .! Mulsant! BH.! (2013).!Neuroimaging!evidence!for!the!deficit!subtype!of!schizophrenia.! JAMA!Psychiatry,!70(5):!472*480.!!

Vojvoda! D,! Grimmell! K,! Sernyak! M! &! Mazure! CM.! (1996).! Monozygotic! twins! concordant! for!response!to!clozapine.!Lancet,!347(8993):!61.!!

Volavka!J,!Czobor!P,!Sheitman!B,!Lindenmayer!JP,!Citrome!L,!McEvoy!JP,!.! .! .!Lieberman!JA.!(2002).!Clozapine,!olanzapine,! risperidone,!and!haloperidol! in! the! treatment!of!patients!with!chronic!schizophrenia! and! schizoaffective! disorder.! The! American! Journal! of! Psychiatry,! 159(2):! 255*262.!!

Wang!F,!Chen!H,! Steketee! JD!&!Sharp!BM.! (2007).!Upregulation!of! ionotropic! glutamate! receptor!subunits!within!specific!mesocorticolimbic!regions!during!chronic!nicotine!self*administration.!Neuropsychopharmacology,!32(1):!103*109.!!

Wang!J,!Zhuang!J,!Iyer!S,!Lin!X,!Whitfield!TW,!Greven!MC,!.!.!.!Weng!Z.!(2012).!Sequence!features!and!chromatin! structure! around! the! genomic! regions! bound!by! 119!human! transcription! factors.!Genome!Research,!22(9):!1798*1812.!!

169!!

!!

Ward!LD!&!Kellis!M.!(2012).!HaploReg:!A!resource!for!exploring!chromatin!states,!conservation,!and!regulatory!motif! alterations!within! sets!of! genetically! linked!variants.!Nucleic!Acids!Research,!Database!Issue,!(40):!D930*D934.!!

Wassef! A,! Baker! J! &! Kochan! LD.! (2003).! GABA! and! schizophrenia:! A! review! of! basic! science! and!clinical!studies.!Journal!of!Clinical!Psychopharmacology,!23(6):!601*640.!!

Watkins,! J! &! Collingridge! G.! (1994).! Phenylglycine! derivatives! as! antagonists! of! metabotropic!glutamate!receptors.!Trends!in!Pharmacological!Sciences,!15(9):!333*342.!!

Watkins!JC!&!Evans!RH.!(1981).!Excitatory!amino!acid!transmitters.!Annual!Review!of!Pharmacology!and!Toxicology,!21:!165*204.!!

Weickert!CS,! Straub!RE,!McClintock!BW,!Matsumoto!M,!Hashimoto!R,!Hyde!TM,! .! .! .! Kleinman! JE.!(2004).! Human! dysbindin! (DTNBP1)! gene! expression! in! normal! brain! and! in! schizophrenic!prefrontal!cortex!and!midbrain.!Archives!of!General!Psychiatry,!61(6):!544*555.!!

Weinberg!W.!(1908).!Über!den!nachweis!der!vererbung!beim!menschen.!Württemberg,!64:!369*382.!!

Weinberger! DR.! (1987).! Implications! of! normal! brain! development! for! the! pathogenesis! of!schizophrenia.!Archives!of!General!Psychiatry,!44(7):!660*669.!!

Weir! B.! (1996).! Genetic! data! analysis! II:! Methods! for! discrete! population! genetic! data!(2nd!ed).!Sunderland,!MA:!Sinauer!Associates.!!

Whitlock!JR,!Heynen!AJ,!Shuler!MG!&!Bear!MF.!(2006).!Learning! induces! long*term!potentiation!in!the!hippocampus.!Science,!313(5790):!1093*1097.!!

Williams!NM,!Preece!A,!Spurlock!G,!Norton!N,!Williams!HJ,!Zammit!S,!.!.!.!Owen!MJ.!(2003).!Support!for!genetic!variation!in!neuregulin!1!and!susceptibility!to!schizophrenia.!Molecular!Psychiatry,!8(5):!485*487.!!

Williams!NM,!Preece!A,!Morris!DW,!Spurlock!G,!Bray!NJ,!Stephens!M,! .! .! .!O'Donovan!MC.! (2004).!Identification! in! 2! independent! samples! of! a! novel! schizophrenia! risk! haplotype! of! the!dystrobrevin!binding!protein!gene!(DTNBP1).!Archives!of!General!Psychiatry,!61(4):!336*344.!!

Wilson! JF,! Weale! ME,! Smith! AC,! Gratrix! F,! Fletcher! B,! Thomas! MG,! .! .! .! Goldstein! DB.! (2001).!Population!genetic!structure!of!variable!drug!response.!Nature!Genetics,!29(3):!265*269.!!

Wilson!WH.!(1996).!Time!required!for!initial!improvement!during!clozapine!treatment!of!refractory!schizophrenia.!The!American!Journal!of!Psychiatry,!153(7):!951*952.!!

Winner! JG,! Carhart! JM,! Altar! CA,! Allen! JD! &! Dechairo! BM.! (2013).! A! prospective,! randomized,!double*blind! study! assessing! the! clinical! impact! of! integrated! pharmacogenomic! testing! for!major!depressive!disorder.!Discovery!Medicine,!16(89):!219*227.!!

170!!

!!

Wittke*Thompson! JK,! Pluzhnikov! A! &! Cox! NJ.! (2005).! Rational! inferences! about! departures! from!Hardy*Weinberg!equilibrium.!American!Journal!of!Human!Genetics,!76(6):!967*986.!!

Wolosker!H,!Blackshaw!S!&!Snyder!SH.!(1999).!Serine!racemase:!A!glial!enzyme!synthesizing!D*serine!to! regulate! glutamate*N*methyl*D*aspartate! neurotransmission.! Proceedings! of! the! National!Academy!of!Sciences!of!the!United!States!of!America,!96(23):!13409*13414.!!

World! Medical! Association! (WMA).! (2013).! WMA! Declaration! of! Helsinki:! Ethical! principles! for!medical! research! involving! human! subjects.! Retrieved! November! 6th,! 2014,! from!http://www.wma.net/en/30publications/10policies/b3/index.html!!

Wright! IC,! Rabe*Hesketh! S,! Woodruff! PW,! David! AS,! Murray! RM! &! Bullmore! ET.! (2000).! Meta*analysis!of!regional!brain!volumes!in!schizophrenia.!The!American!Journal!of!Psychiatry,!157(1):!16*25.!!

Wyatt!RJ!&!Henter! I.! (2001).!Rationale!for!the!study!of!early! intervention.!Schizophrenia!Research,!51(1):!69*76.!!

Xia!Z,!Dudek!H,!Miranti!CK!&!Greenberg!ME.!(1996).!Calcium!influx!via!the!NMDA!receptor!induces!immediate!early!gene!transcription!by!a!MAP!kinase/ERK*dependent!mechanism.!The!Journal!of!Neuroscience,!16(17):!5425*5436.!!

Xu!Z!&!Taylor!JA.!(2009).!SNPinfo:!Integrating!GWAS!and!candidate!gene!information!into!functional!SNP! selection! for! genetic! association! studies.! Nucleic! Acids! Research,! Web! Server! issue,! 37:!W600*W605.!!

Yamamori!H,!Hashimoto!R,!Fujita!Y,!Numata!S,!Yasuda!Y,!Fujimoto!M,!.!.!.!Takeda!M.!(2014).!Changes!in!plasma!D*serine,!L*serine,!and!glycine!levels!in!treatment*resistant!schizophrenia!before!and!after!clozapine!treatment.!Neuroscience!Letters,!582:!93*98.!!

Yamamoto!BK!&!Cooperman!MA.!(1994).!Differential!effects!of!chronic!antipsychotic!drug!treatment!on!extracellular!glutamate!and!dopamine!concentrations.!The! Journal!of!Neuroscience,!14(7):!4159*4166.!!

Yang! HC,! Liu! CM,! Liu! YL,! Chen! CW,! Chang! CC,! Fann! CS,! .! .! .! Hwu! HG.! (2013).! The! DAO! gene! is!associated! with! schizophrenia! and! interacts! with! other! genes! in! the! Taiwan! Han! Chinese!population.!PloS!One,!8(3):!e60099.!!

Yoo!HJ,!Cho!IH,!Park!M,!Yang!SY!&!Kim!SA.!(2012).!Family!based!association!of!GRIN2A!and!GRIN2B!with!Korean!autism!spectrum!disorders.!Neuroscience!Letters,!512(2):!89*93.!!

Yuan!HY,!Chen! JJ,! Lee!MT,!Wung! JC,!Chen!YF,!Charng!MJ,! .! .! .!Chen!YT.! (2005).!A!novel! functional!VKORC1! promoter! polymorphism! is! associated! with! inter*individual! and! inter*ethnic!differences!in!warfarin!sensitivity.!Human!Molecular!Genetics,!14(13):!1745*1751.!!

171!!

!!

Yuen!EY,!Liu!W,!Karatsoreos,!IN,!Ren!Y,!Feng!J,!McEwen!BS!&!Yan!Z.!(2011).!Mechanisms!for!acute!stress*induced! enhancement! of! glutamatergic! transmission! and! working!memory.! Molecular!Psychiatry,!16(2):!156*170.!!

Zafra! F,! Aragon! C,! Olivares! L,! Danbolt! NC,! Gimenez! C! &! Storm*Mathisen! J.! (1995).! Glycine!transporters!are!differentially!expressed!among!CNS!cells.! The! Journal!of!Neuroscience,!15(5,!2):!3952*3969.!!

Zai!CC,!Manchia!M,!De!Luca!V,!Tiwari!AK,!Chowdhury!NI,!Zai!GC,!.!.!.!Kennedy!JL.!(2012).!The!brain*derived! neurotrophic! factor! gene! in! suicidal! behaviour:! A! meta*analysis.! The! International!Journal!of!Neuropsychopharmacology,!15(8):!1037*1042.!!

Zhang!C,!Li!Z,!Wu!Z,!Chen!J,!Wang!Z,!Peng!D,! .! .! .!Fang!Y.! (2014).!A!study!of!N*methyl*D*aspartate!receptor! gene! (GRIN2B)! variants! as! predictors! of! treatment*resistant! major! depression.!Psychopharmacology,!231(4):!685*693.!!

Zhao!C,!Sun!T!&!Li!M.!(2012).!Neural!basis!of!the!potentiated!inhibition!of!repeated!haloperidol!and!clozapine! treatment! on! the! phencyclidine*induced! hyperlocomotion.! Progress! in! NeuroPPsychopharmacology!&!Biological!Psychiatry,!38(2):!175*182.!!

Zheng!X,!Zhang!L,!Wang!AP,!Bennett!MV!&!Zukin!RS.! (1997).!Ca2+! influx!amplifies!protein!kinase!C!potentiation!of!recombinant!NMDA!receptors.!The!Journal!of!Neuroscience,!17(22):!8676*8686.!!

Zuk! O,! Hechter! E,! Sunyaev! SR! &! Lander! ES.! (2012).! The! mystery! of! missing! heritability:! Genetic!interactions! create!phantom!heritability.! Proceedings!of! the!National!Academy!of! Sciences!of!the!United!States!of!America,!109(4):!1193*1198.!!

Zweifel!LS,!Argilli!E,!Bonci!A!&!Palmiter!RD.!(2008).!Role!of!NMDA!receptors!in!dopamine!neurons!for!plasticity!and!addictive!behaviors.!Neuron,!59(3):!486*496.!!

!