brain cyp2d metabolism of opioids impacts brain levels ... · graduate department of pharmacology...

229
Brain CYP2D Metabolism of Opioids Impacts Brain Levels, Analgesia, and Tolerance by Douglas McMillan A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Pharmacology and Toxicology University of Toronto © Copyright by Douglas McMillan 2018

Upload: others

Post on 20-Jun-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

Brain CYP2D Metabolism of Opioids Impacts Brain Levels, Analgesia, and Tolerance

by

Douglas McMillan

A thesis submitted in conformity with the requirements

for the degree of Doctor of Philosophy

Department of Pharmacology and Toxicology

University of Toronto

© Copyright by Douglas McMillan 2018

Page 2: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

ii

Brain CYP2D Metabolism of Opioids Impacts Brain Levels, Analgesia, and Tolerance

Douglas McMillan

Doctor of Philosophy

Graduate Department of Pharmacology and Toxicology

University of Toronto

2018

Abstract

Cytochrome P450 2D (CYP2D) is a subfamily of enzymes expressed in both liver and brain that

metabolizes clinically used drugs, neurotoxins, and endogenous neurochemicals. Opioid

analgesics are metabolized by CYP2D to more potent analgesic metabolites, and variation in this

metabolism may alter opioid response. While human CYP2D expression in the liver is primarily

regulated by genetics, brain CYP2D activity may be altered by genetics, environmental inducers,

and hormonal regulation. Smokers have higher brain, but not liver, CYP2D and nicotine induces

rat brain, but not liver, CYP2D. This thesis investigated the impact of altering rat brain CYP2D

activity in vivo on the brain metabolism of opioids and resulting analgesia. Rats were administered

opioids and analgesia was measured; drug levels were assessed in brain and plasma after sacrifice,

or in brain in vivo by microdialysis. Rat brain CYP2D was inhibited by intracerebroventricular

injection of a CYP2D mechanism-based inhibitor or induced by seven-day subcutaneous nicotine

treatment. We found that rat brain CYP2D activation of codeine to morphine mediates acute

codeine analgesia; inhibiting brain CYP2D decreased, and inducing brain CYP2D increased, brain

morphine from codeine and resulting codeine analgesia. In a rat model of codeine analgesic

tolerance, inducing brain CYP2D increased acute analgesia and the rate of analgesic tolerance,

producing a greater absolute decrease in analgesia per codeine dose. Administering higher doses

of codeine similarly increased acute analgesia and the rate of tolerance. The rate of tolerance

correlated to acute analgesia indicating that brain CYP2D influenced codeine tolerance by altering

acute analgesic-response. Lastly, we found that rat brain CYP2D metabolism of oxycodone to

oxymorphone decreased acute oxycodone analgesia; inhibiting brain CYP2D increased brain

oxycodone levels and resulting analgesia, while inducing brain CYP2D increased brain

oxymorphone levels but decreased oxycodone analgesia. This thesis demonstrated an impact of rat

Page 3: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

iii

brain CYP2D on acute codeine and oxycodone induced analgesia, as well as the rate of codeine

analgesic tolerance. Variation in human brain CYP2D metabolism may be a clinically relevant

source of variation in opioid response. Novel approaches to understanding interindividual

differences in opioid analgesia and adverse effects remain critical for improving opioid efficacy

and mitigating abuse.

Page 4: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

iv

Acknowledgements

First, I wanted to thank my supervisor Rachel Tyndale for her support throughout my research.

Your mentorship has helped immeasurably in developing my skills in scientific writing, critical

thinking, presenting, and independent research. The training and opportunities I have been given

will be invaluable moving forward in my career and I am very grateful. I also wanted to thank my

PhD supervisory committee members David Riddick and Jose Nobrega for sharing their scientific

expertise by way of continued insight and helpful suggestions.

I also wanted to extend many thanks to all past and present members of the Tyndale lab.

Specifically, thanks to Sharon Miksys for her kindness and approachability in all matters scientific

and otherwise, Bin Zhao for his resilience in running my thousands of samples, Fariba Baghai

Wadji for her masterful surgical hands, Ewa Hoffman for providing welcomed distractions and an

outlet for my hockey addiction, Meghan Chenoweth for ensuring I was always well-caffeinated,

and JAT for leading the way and providing me with a graduate school blueprint which without, I

would be nowhere near finishing this PhD.

Lastly, beyond the Tyndale lab, I wanted to thank my friends and family. Thanks to Lauren, my

better-half through the majority of this PhD. Your passion, work ethic, and avid curiosity continues

to inspire me and you have provided me with an incredibly solid foundation throughout my studies.

Thank you to my sister Katie, my brother-in-law Justyn, and my nephews Tyler and Jackson, for

providing fun diversions from work whenever needed. And finally, to my parents, I’ll never be

able to thank you enough for everything you’ve done for me. Your support through my decade of

university has allowed my only source of stress to be academic, and you’ve provided me with

endless encouragement, validation, love, clean Barrie air, and Leafs tickets. Without you this

wouldn’t have been possible.

Page 5: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

v

Table of Contents

List of Publications ........................................................................................................................ ix

List of Tables ................................................................................................................................. xi

List of Figures ............................................................................................................................... xii

List of Abbreviations .................................................................................................................... xv

List of Appendices ...................................................................................................................... xvii

Statement of Research Problem ................................................................................................. xviii

Main Research Objectives ........................................................................................................... xxi

1. General Introduction ................................................................................................................... 1

1.1 – Opioid analgesics ............................................................................................................... 1

1.1.1 General introduction ....................................................................................................... 1

1.1.2 Current clinical use and treatment indications ............................................................... 1

1.1.3 Pharmacology ................................................................................................................. 2

1.1.3.1 Opioid receptors ....................................................................................................... 2

1.1.3.2 Opioid metabolism ................................................................................................... 4

1.1.4 Opioid neuropharmacology ............................................................................................ 5

1.1.4.1 Analgesia.................................................................................................................. 5

1.1.4.2 Tolerance.................................................................................................................. 7

1.1.4.3 Dependence .............................................................................................................. 9

1.1.5 Opioid epidemic; misuse and abuse ............................................................................. 10

1.2 – CYPs................................................................................................................................. 14

1.2.1 General introduction ..................................................................................................... 14

1.2.2 CYP2D6 enzyme .......................................................................................................... 15

1.2.2.1 Substrates and clinical drugs of interest ................................................................. 15

1.2.2.2 Variation in CYP2D6 ............................................................................................. 17

1.2.2.2.1 Genetic sources of variation ............................................................................ 17

1.2.2.2.1.1. Interethnic variability .............................................................................. 19

1.2.2.2.1.2 Functional impact of CYP2D6 polymorphism ......................................... 21

1.2.2.2.2 Non-genetic sources of variation and drug-drug interactions ......................... 22

1.2.2.3 Species differences in CYP2D6 enzyme ............................................................... 23

1.3 – CYP2D6 and opioids ........................................................................................................ 24

Acknowledgements ........................................................................................................................ iii

Page 6: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

vi

1.3.1 CYP2D6 metabolism of opioids ................................................................................... 24

1.3.2 Impact of CYP2D6 phenotype on opioid analgesia ..................................................... 25

1.3.3 Impact of CYP2D6 phenotype on opioid abuse liability .............................................. 28

1.3.4 Other sources of variability in opioid response ............................................................ 29

1.4 – Brain CYPs ....................................................................................................................... 30

1.4.1 Brain CYP expression .................................................................................................. 30

1.4.2 Brain CYP activity........................................................................................................ 31

1.4.3 Regulation of brain CYP activity ................................................................................. 33

1.4.4 Brain CYP2D ................................................................................................................ 34

1.4.4.1 Brain CYP2D expression and activity ................................................................... 34

1.4.4.2 Brain CYP2D regulation ........................................................................................ 36

1.4.4.3 Functional impact of variation in brain CYP2D .................................................... 38

1.4.4.3.1 Neurotoxicity ................................................................................................... 38

1.4.4.3.2 Endogenous neurochemicals ........................................................................... 41

1.4.4.4 Brain CYP2D metabolism of opioids .................................................................... 42

1.5 – Statement of Research Hypotheses .................................................................................. 44

2. Chapter 1: Nicotine increases codeine analgesia through the induction of brain CYP2D and

central activation of codeine to morphine ..................................................................................... 45

2.1 Abstract ............................................................................................................................... 46

2.2 Introduction ......................................................................................................................... 47

2.3 Materials and Methods ........................................................................................................ 48

2.4 Results ................................................................................................................................. 51

2.5 Discussion ........................................................................................................................... 59

2.6 Acknowledgements ............................................................................................................. 62

2.7 Supplementary Information................................................................................................. 63

2.8 Significance to Thesis ......................................................................................................... 64

3. Chapter 2: Inducing rat brain CYP2D with nicotine increases the rate of codeine tolerance;

predicting the rate of tolerance from acute analgesic response .................................................... 65

3.2 Introduction ......................................................................................................................... 67

3.3 Methods and Materials ........................................................................................................ 69

3.4 Results ................................................................................................................................. 73

Page 7: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

vii

3.5 Discussion ........................................................................................................................... 82

3.6 Acknowledgements ............................................................................................................. 87

3.7 Significance to Thesis ......................................................................................................... 88

4. Chapter 3: Rat brain CYP2D activity alters in vivo central oxycodone metabolism, levels, and

resulting analgesia ......................................................................................................................... 89

4.1 Abstract ............................................................................................................................... 90

4.2 Introduction ......................................................................................................................... 91

4.3 Materials and Methods ........................................................................................................ 92

4.4 Results ................................................................................................................................. 96

4.5 Discussion ......................................................................................................................... 104

4.6 Acknowledgements ........................................................................................................... 108

4.7 Supplementary Information............................................................................................... 108

4.8 Significance to Thesis ....................................................................................................... 114

5. General Discussion ................................................................................................................. 115

5.1 Summary of findings ......................................................................................................... 115

5.1.1 Brain CYP2D and codeine ......................................................................................... 115

5.1.2 Brain CYP2D and oxycodone .................................................................................... 116

5.2 Predicting the contribution of opioid parent and CYP2D metabolite to drug effect ........ 118

5.2.1 Pharmacokinetics, pharmacodynamics and blood-brain barrier transport ................. 118

5.2.1.1 Codeine ................................................................................................................ 118

5.2.1.2 Oxycodone ........................................................................................................... 120

5.2.2 The potential impact of brain CYP2D on other opioids ............................................. 124

5.3 Brain CYP2D and chronic opioid administration ............................................................. 126

5.3.1 Absolute vs. proportional decrease in codeine analgesia ........................................... 126

5.3.2 Implications of brain CYP2D in opioid abuse liability .............................................. 131

5.4 Functional impact of brain CYP2D6 metabolism of opioids in humans .......................... 133

5.4.1 Brain CYP2D6 and the opioid epidemic .................................................................... 133

5.4.2 Potential sex differences in the brain CYP2D6 metabolism of opioids ..................... 135

5.4.3 Endogenous opioid pain thresholds ............................................................................ 137

5.5 Extensions of research ....................................................................................................... 139

5.5.1 Drug-drug interactions with brain CYP2D ................................................................. 139

Page 8: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

viii

5.5.2 Imaging brain CYP2D6 in humans ............................................................................ 140

5.5.3 Therapeutic targeting of brain CYP2D6 in humans ................................................... 142

5.5.4 The mechanism of inducible brain-specific CYP2D .................................................. 144

5.6 Conclusions ....................................................................................................................... 145

6. References ............................................................................................................................... 147

7. Appendices .............................................................................................................................. 185

Page 9: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

ix

List of Publications

Research Articles in Thesis

McMillan, D.M., Tyndale, R.F. Nicotine increases codeine analgesia through the induction of

brain CYP2D and central activation of codeine to morphine. Neuropsychopharmacology. 2015;

40(7): 1804-1812.

McMillan, D.M., Tyndale, R.F. Inducing rat brain CYP2D with nicotine increases the rate of

codeine tolerance; predicting the rate of tolerance from acute analgesic response. Biochemical

Pharmacology. 2017; 145: 158-168.

McMillan, D.M., Miksys, S., Tyndale, R.F. Rat brain CYP2D activity alters in vivo central

oxycodone metabolism, levels, and resulting analgesia. Addictions Biology. 2017; Epub ahead

of print.

Additional Manuscript from Doctoral Work Attached in Appendix

McMillan, D.M., Tyndale, R.F. CYP-mediated drug metabolism in the brain impacts drug

response. Pharmacology and Therapeutics. 2018; 184: 189-200.

Submitted Abstracts for Oral and Poster Presentations

McMillan, D.M., Tyndale, R.F. Poster: Variation in rat brain CYP2D alters oxycodone

metabolism and analgesia. May 2017 – UofT Department of Pharmacology Visions in

Pharmacology; Toronto, ON, Canada. October 2016 – Annual symposium on Microsomes and

Drug Oxidations; Davis, CA, USA.

McMillan, D.M., Tyndale, R.F. Poster: Induction of brain CYP2D-mediated activation of codeine

to morphine increases the rate of codeine analgesic tolerance. October 2016 – Annual

symposium on Microsomes and Drug Oxidations; Davis, CA, USA. July 2016 – Annual

International Narcotics Research Conference; Bath, UK. June 2016 – UofT Department of

Pharmacology Visions in Pharmacology; Toronto, ON, Canada.

McMillan, D.M., Tyndale, R.F. Poster: The impact of variation in rat brain CYP2D activity on

codeine analgesia is similar across multiple routes of codeine administration and analgesia

assays. June 2015 – Annual meeting of the Canadian Society of Pharmacology and

Therapeutics; Toronto, ON, Canada. May 2015 – UofT Department of Pharmacology Visions

in Pharmacology; Toronto, ON, Canada.

Page 10: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

x

McMillan, D.M., Tyndale, R.F. Oral: Nicotine increases codeine analgesia through the induction

of brain CYP2D and central activation of codeine to morphine. April 2015 – Campbell Family

Research Institute, Center for Addiction and Mental Health, Trainee Seminar Series; Toronto,

ON, Canada.

McMillan, D.M., Tyndale, R.F. Poster: Nicotine increases codeine analgesia through induction of

brain CYP2D and central activation of codeine to morphine. March 2015 – Annual Meeting for

The American Society for Pharmacology and Experimental Therapeutics (ASPET) in

conjunction with Experimental Biology (EB); Boston, MA, USA.

McMillan, D.M., Zhao, B., Tyndale, R.F. Poster: Chronic nicotine increases codeine analgesia

through induction of brain CYP2D metabolic activation of codeine to morphine. May 2014 –

Department of Pharmacology Visions in Pharmacology (VIP); Toronto, ON, Canada.

Page 11: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xi

List of Tables

Table 1: Examples of CYP2D6 substrates and the reactions catalyzed by CYP2D6 ................... 17

Table 2: Examples of competitive and mechanism-based CYP2D6 inhibitors ............................ 24

Page 12: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xii

List of Figures

Figure 1: Neural pathway of nociception and descending modulation by opioids ......................... 7

Figure 2: Neural pathway of opioid dependence .......................................................................... 10

Figure 3: Region-specific percentage of daily opioid consumption in 2001-2003, compared to

2011-2013 ..................................................................................................................................... 12

Figure 4: Country-specific opioid consumption per capita, and the prevalence of opioid misuse,

in 2016 .......................................................................................................................................... 13

Figure 5: Total opioid use and reported misuse in individuals above the age of 12 in the US in

2016, split by primary opioid compound ...................................................................................... 13

Figure 6: Genomic impact of select CYP2D6 variant alleles ....................................................... 18

Figure 7: Select CYP2D6 allele frequencies across ethnic groups ............................................... 20

Figure 8: Structures and properties of opioid parent compounds and CYP2D6-mediated

metabolites .................................................................................................................................... 26

Figure 9: Fold change in brain CYP2D protein in human smokers, compared to non-smokers,

and short-term nicotine pretreated rats, compared to saline pretreated ........................................ 37

Figure 10: Modifying rat brain CYP2D activity alters catalepsy after acute haloperidol

administration ............................................................................................................................... 40

Figure 11: Modifying rat brain CYP2D activity alters vacuous chewing movements after chronic

haloperidol administration ............................................................................................................ 41

Figure 12: The analgesic time course following codeine and morphine suggests peripheral

morphine levels do not predict central morphine levels or analgesia ........................................... 52

Figure 13: Inhibiting brain CYP2D reduced the first 30 min of codeine-induced analgesia but had

no effect on morphine-induced analgesia ..................................................................................... 53

Figure 14: Nicotine-mediated induction of brain CYP2D increased codeine analgesia and had no

effect on morphine-induced analgesia .......................................................................................... 55

Figure 15: Nicotine induction of brain CYP2D increased the first 30 min of codeine-induced

analgesia, an effect that was blocked by inhibiting brain CYP2D ............................................... 57

Figure 16: Codeine, but not morphine, levels correlate between plasma and brain; brain

morphine levels reflect codeine analgesia .................................................................................... 58

Page 13: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xiii

Figure 17: Nicotine mediated induction of brain CYP2D increased codeine analgesia, and had no

effect on morphine-induced analgesia when assessed via the hot plate assay .............................. 63

Figure 18: Experimental study design and timeline for pretreatment and treatment phases of

tolerance experiment ..................................................................................................................... 71

Figure 19: Nicotine pretreatment increased acute codeine-induced analgesia; repeated codeine

injections resulted in analgesic tolerance, the rate of which was increased by nicotine

pretreatment .................................................................................................................................. 75

Figure 20: Control animals showed no change in analgesia after a one-week washout between

codeine injections; analgesia was not altered by daily p.o. (i.e. codeine vehicle) injections or

behavioural testing ........................................................................................................................ 77

Figure 21: Nicotine pretreatment did not alter morphine-induced analgesia; repeated morphine

injections resulted in analgesic tolerance, the rate of which was not altered by nicotine

pretreatment .................................................................................................................................. 78

Figure 22: Control animals showed no change in analgesia after a one-week washout between

morphine injections; analgesia was not altered by daily p.o. (i.e. morphine vehicle) injections and

behavioural testing ........................................................................................................................ 79

Figure 23: Initial analgesia correlated to rate of tolerance in animals given higher doses of

codeine and morphine ................................................................................................................... 81

Figure 24: Initial analgesia predicts the rate of onset of tolerance across multiple codeine and

morphine doses ............................................................................................................................. 82

Figure 25: Oxycodone and oxymorphone doses were chosen from dose–response curves ......... 94

Figure 26: Inhibiting brain CYP2D increased, and inducing brain CYP2D decreased, oxycodone

analgesia ........................................................................................................................................ 98

Figure 27: Inhibiting induced brain CYP2D reversed the changes in oxycodone analgesia;

variation in brain CYP2D activity did not affect oxymorphone analgesia ................................. 100

Figure 28: Manipulating brain CYP2D further suggests that brain oxycodone levels, not

oxymorphone levels, are responsible for oxycodone analgesia .................................................. 102

Figure 29: Manipulating brain CYP2D altered the CYP2D metabolic ratio in the brain;

oxycodone analgesia was inversely correlated with brain CYP2D activity ............................... 104

Figure 30: Study paradigm of pretreatment timing prior to oxycodone or oxymorphone

administration ............................................................................................................................. 111

Page 14: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xiv

Figure 31: Inhibiting brain CYP2D with the competitive inhibitor propafenone increased

oxycodone analgesia ................................................................................................................... 112

Figure 32: Full microdialysis analyte curves for each pretreatment group after oxycodone

administration ............................................................................................................................. 113

Figure 33: Proposed model for the functional role of rat brain CYP2D in the activation of

codeine to morphine in vivo ....................................................................................................... 116

Figure 34: Proposed model for the functional role of rat brain CYP2D in the metabolism of

oxycodone to oxymorphone in vivo ........................................................................................... 117

Figure 35: Initial hypotheses (option A and B, compared to reference in grey) of the influence of

increased acute codeine analgesia on the rate of development of tolerance ............................... 128

Figure 36: Dose-escalation of morphine and oxycodone after 21 days of opioid treatment in

individuals receiving treatment for chronic cancer pain ............................................................. 129

Figure 37: Modified abuse quotient (EMAX/TMAX) for codeine and oxycodone after brain

CYP2D inhibitor and/or inducer pretreatment ............................................................................ 132

Figure 38: Pilot data on sex differences in ex vivo rat brain and liver CYP2D activity and central

oxycodone metabolism in vivo ................................................................................................... 137

Page 15: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xv

List of Abbreviations

%MPE Percentage of maximum possible effect

8MOP 8-Methoxypsoralen

ACSF Artificial cerebrospinal fluid

ANOVA Analysis of variance

ATP Adenosine triphosphate

AUC Area under the curve

BBB Blood-brain barrier

cAMP Cyclic adenosine monophosphate

CNS Central nervous system

CYP Cytochrome P450

DAMGO (D-Ala2, N-MePhe4, Gly-ol)-enkephalin

DOR Delta (δ) opioid receptor

EM Extensive metabolizer

fMRI Functional magnetic resonance imaging

GABA Gamma (γ)-aminobutyric acid

GDP Guanosine diphosphate

GPCR G protein-coupled receptor

GRK G protein-coupled receptor kinase

GTP Guanosine triphosphate

HNF4a Hepatocyte nuclear factor 4-alpha (α)

HPLC High-performance liquid chromatography

i.c.v. Intracerebroventricular

IM Intermediate metabolizer

i.p. Intraperitoneal

KOR Kappa (κ) opioid receptor

LCMS Liquid chromatography-mass spectrometry

MBI Mechanism-based inhibitor

MDR1 Multiple drug resistance protein 1

miRNA Micro ribonucleic acid

MOR Mu (µ) opioid receptor

MPP+ 1-Methyl-4-phenylpyridinium

MPTP 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine

mRNA Messenger ribonucleic acid

nAChR Nicotinic acetylcholine receptor

NADPH Nicotinamide adenine dinucleotide phosphate

NMDA N-methyl-D-aspartic acid

O-DSMT O-Desmethyl tramadol

OPRM1 Opioid receptor mu (µ) 1

OR Opioid receptor

PAG Periaqueductal gray

PET Positron emission tomography

P-gp P-Glycoprotein

PM Poor metabolizer

p.o. Per os (oral gavage)

PO Prescription opioid

Page 16: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xvi

RVM Rostral ventromedial medulla

s.c. Subcutaneous

SEM Standard error of the mean

SNP Single nucleotide polymorphism

TFL Tail-flick latency

UGT Uridine 5’-diphospho-glucuronosyltransferase

UM Ultra-rapid metabolizer

VCM Vacuous chewing movement

VTA Ventral tegmental area

Page 17: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xvii

List of Appendices

Appendix A: CYP-mediated drug metabolism in the brain impacts drug response…………185

Page 18: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xviii

Statement of Research Problem

Centrally-acting drug response does not always correlate with circulating drug or

metabolite levels (Michels and Marzuk, 1993). This is due, in part, to permeability of the drug

and/or active drug metabolites across the blood-brain barrier (BBB) and into the brain, but may

also be due to drug metabolism within the brain affecting central nervous system (CNS) drug

and/or metabolite levels. Drug-metabolizing cytochrome P450 (CYP) enzymes are expressed and

active in the brain (Miksys and Tyndale, 2013); region- and cell-specific brain CYP expression

and activity have been characterized in numerous experimental animals including rats (Miksys and

Tyndale, 2002). The CYP2 family, including CYP2D, is expressed in both liver and brain, and

metabolizes roughly 20% of all clinically used drugs, including a large proportion of CNS-acting

drugs (e.g. antidepressants, antipsychotics), drugs of abuse (e.g. psychostimulants), and

neurotransmitters (Bromek et al, 2010; Haduch et al, 2013; Zanger et al, 2004; Zanger and

Schwab, 2013).

While hepatic CYP2D levels are primarily regulated by genetics, brain CYP2D levels are

influenced by genetics, age, sex, and xenobiotic inducers (Mann et al, 2012; Miksys and Tyndale,

2013). Higher brain (but not liver) CYP2D is found in smokers, and seven days of nicotine

administration induces rat brain (but not liver) CYP2D (Mann et al, 2008; Miksys et al, 2002; Yue

et al, 2008; Zanger and Schwab, 2013). Variation (genetic and/or environmental) in brain CYP2D

activity may contribute to clinically relevant differences in individual response to centrally acting

CYP2D substrates that are not reflected in peripheral, hepatic CYP-influenced, drug and

metabolite levels.

Opioid analgesics are an effective class of analgesic compounds, used widely in the

treatment of pain associated with numerous indications (e.g. diabetic neuropathy [neuropathic],

osteoarthritis [visceral], and myocardial ischaemia [somatic]) (Smith, 2011). Due to their

rewarding and reinforcing properties, extended opioid treatment can lead to misuse, dependence,

and mortality. The overall prevalence of opioid abuse and misuse among adult opioid users in the

United States was 12.5% in 2016, relatively stable from 12.8% in 2015 (NSDUH, 2016, 2017).

However, abuse prevalence ranges by opioid compound, from 10.4% of adult codeine users

reporting abuse, 14.1% of oxycodone users, and 27.6% of oxymorphone users (NSDUH, 2017).

Interindividual differences in opioid pain relief and in risk for dependence are not fully

understood. Most opioids, like codeine and oxycodone, are metabolically activated by CYP2D to

Page 19: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xix

compounds that are more potent opioid receptor ligands, and thereby elicitors of analgesia. CYP2D

metabolizes codeine to morphine, a step required for codeine analgesia; CYP2D poor metabolizers

and individuals given CYP2D inhibitors produce little morphine from codeine and exhibit little

codeine analgesia (Chen et al, 1991b; Sindrup et al, 1992). Oxycodone is also metabolized to the

more potent analgesic compound oxymorphone; however, while giving individuals CYP2D

inhibitors decreased the production of oxymorphone from oxycodone, oxycodone analgesia was

unaffected (Gronlund et al, 2010). Investigation into opioid metabolism and analgesia in CYP2D

poor metabolizers, as well as individuals given peripheral CYP2D enzyme inhibitors, indicate that

depending on the opioid, CYP2D metabolism may promote, mitigate, or have no effect on, opioid

response (Smith, 2011). Little is known about the role of brain CYP2D metabolism in opioid

response.

Brain CYP metabolism may play a role in neuroprotection through metabolic inactivation

of neurotoxins. Brain CYP2D inactivates toxins that can cause parkinsonism-symptoms such as

pesticides; genetically rapid CYP2D6 metabolizers are at a lower risk for Parkinson disease,

potentially due to enhanced brain CYP2D6-mediated inactivation of parkinsonism-inducing toxins

(Elbaz et al, 2004; Zanger et al, 2004). Smokers also have a lower risk of Parkinson’s disease

which may be due in part to greater CYP2D6 expression in the brain (Alves et al, 2004). In support

of this, nicotine protects against the parkinsonism-inducing compound 1-methyl-4-

phenylpyridinium (MPP+)-induced nigrostriatal damage in an animal model of Parkinson’s

disease (Quik et al, 2009). Also, inhibiting CYP2D exacerbates MPP+ (neurotoxin)-induced cell

death in a human neuronal cell line, further suggesting a role for brain CYP2D in protection against

neurotoxicity (Mann and Tyndale, 2010). Manipulating brain, but not liver, CYP2D metabolism

of haloperidol in rats alters neurotoxicity in both acute and chronic haloperidol administration

paradigms (Miksys et al, 2017).

The studies included in the thesis aim to provide insight into the impact of brain CYP2D

metabolism of opioids on the functional outcome of analgesia through altering brain CYP2D

metabolism during acute or repeated opioid administration. This line of research stems from the

finding that selectively inhibiting rat brain, but not liver, CYP2D decreased brain morphine levels

and analgesia after acute subcutaneous codeine administration (Zhou et al, 2013). This thesis aims

to extend these findings to models of variable (increased and decreased) brain CYP2D activity,

routes of opioid administration more closely mirroring human use, multiple opioid compounds for

Page 20: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xx

assessing opioid-specific influence of brain CYP2D metabolism, and multiple functional outcomes

of opioid response. Investigation into this relatively new field of CYP function in the brain may

provide a mechanistic understanding of a novel source of variation in opioid response, specifically

codeine and oxycodone, and may have clinical implications in long-term opioid-use, escalation,

and transition to abuse.

Page 21: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xxi

Main Research Objectives

Objective 1: CYP2D activates codeine to morphine, a step required for codeine analgesia.

Differences in CNS permeability and efflux suggest that initial morphine in the brain after codeine

dosing may be derived from brain CYP2D metabolism. While CYP2D in the liver is primarily

regulated by genetics, a combination of genetics, environmental inducers, and age, may contribute

to a wide range of brain CYP2D activity. Inhibiting rat brain CYP2D reduced brain morphine

formation, and resulting analgesia, after subcutaneous codeine administration. However, it is

unknown whether selectively inducing brain CYP2D changes brain CYP2D sufficiently to alter

drug response in vivo, in the presence of substantial first-pass metabolism by the liver using

intraperitoneal drug administration. Therefore, the first objective was to assess the effect of

inducing (with and without concurrent inhibition) rat brain CYP2D activity, without

altering liver CYP2D, on brain codeine and morphine levels, and analgesia, following

intraperitoneal codeine administration.

Objective 2: Repeated exposure to opioids results in tolerance, a loss of analgesic potency, which

may lead to dose escalation, a greater likelihood of opioid misuse, and transition to dependence.

Inducing rat brain CYP2D increases the central activation of codeine to morphine, resulting in

increased onset and magnitude of codeine analgesia (objective 1). It is unknown whether this

difference in initial analgesia can affect the rate of development of tolerance. Higher initial codeine

analgesia could result in an equivalent relative drop (equal slope of tolerance development), or a

more rapid proportional drop (equal time to tolerance), in analgesic potency. Therefore, the second

objective was to assess the effect of inducing brain CYP2D, and thereby the central activation

of codeine to morphine, on the rate of development of tolerance to codeine’s analgesic effects

across seven days of repeated oral codeine administration.

Objective 3: Oxycodone, similar to codeine, is metabolized by CYP2D to a more potent opioid

receptor activating compound, oxymorphone. However, unlike codeine, oxycodone itself is a

potent µ-opioid receptor agonist. Compared to its CYP2D metabolite oxymorphone, oxycodone

has a greater influx across the BBB and is present at higher concentrations in the CNS after

peripheral administration. While inhibiting human hepatic CYP2D does not alter oxycodone

response, brain CYP2D metabolism may increase oxycodone analgesia through the formation of

Page 22: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

xxii

oxymorphone directly within the CNS, may reduce oxycodone analgesia through local oxycodone

metabolism, or may have no impact. The third and final objective was to assess the impact of

rat brain CYP2D on the metabolism of oxycodone to oxymorphone, using in vivo brain

microdialysis, and resulting analgesia after oral oxycodone administration.

Page 23: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

1

1. General Introduction

1.1 – Opioid analgesics

1.1.1 General introduction

Opioid analgesics are among the most effective treatments available for pain. The term ‘opioid’

includes all compounds that bind to opioid receptors, while ‘opiate’ conventionally refers to opioid

alkaloids derived from the opium poppy (Papaver somniferum) (Rosenblum et al, 2008). Opioids

include semi-synthetic compounds derived from naturally occurring opiates (e.g. heroin from

morphine). The history of human use of opioids dates back thousands of years, from records of

exudation of opium from the opium poppy in the sixth millennium BCE, to the chemical isolation

of the active alkaloid morphine from opium in the 19th century, and the clinical use of synthetic

opioid analgesics today (Brook et al, 2017). While opioids are often regarded as the standard of

care in treating acute and chronic pain, the associated side effects tolerance, and risks of abuse,

addiction, and withdrawal have caused major concern about their long-term usefulness

(Rosenblum et al, 2008). For this reason, the use of opioids for less severe indications, such as

chronic non-malignant pain, remains controversial (discussed further in 1.1.5) (Manchikanti,

2008).

1.1.2 Current clinical use and treatment indications

Opioid analgesics are used to treat moderate to severe chronic pain, presenting in numerous disease

states. For example, oxycodone has been indicated for neuropathic (e.g. diabetic neuropathy, post-

herpetic neuralgia), somatic (e.g. osteoarthritis-related and rheumatic joint pain, low back pain),

visceral (e.g. post-operative pain, myocardial ischaemia), and cancer pain (Riley et al, 2008). A

meta-analysis of 41 randomized controlled trials up to 2006, found that opioids were effective in

treating pain and improving quality of life in patients with nociceptive (benign, acute damage) and

neuropathic (chronic nerve damage) pain (Furlan et al, 2006). While guidelines on formulations,

dosing, and prescribing have been developed to better instruct opioid use in pain management,

controversy still exists surrounding the type of conditions treated, and the safety in treating, with

opioids (Dowell et al, 2016). This is due, in part, to the high prevalence of chronic pain in the

general population; estimates vary, but up to 40% of Canadians (2011) and 30% of Americans

(2010) reported experiencing some form of chronic pain in the previous year due to illness, an

accident, or an existing medical condition (Johannes et al, 2010; Schopflocher et al, 2011).

Page 24: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

2

Consequentially, due to the high prevalence of chronic pain and the effectiveness of opioids in

treating pain, opioids are prescribed regularly; in 2016, 92 million people above the age of 12 in

the US reported using any prescription opioid, while US retail pharmacies dispensed

approximately 250 million opioid prescriptions (NSDUH, 2017; Pezalla et al, 2017).

In Canada and the US, numerous opioid formulations have been commercialized for oral and

transdermal administration, used primarily in ambulatory care settings, and for intravenous

administration, used primarily in surgical settings. These include combination products of an anti-

inflammatory (Acetaminophen, Acetylsalicylic acid (aspirin), Ibuprofen) with an opioid

compound, such as codeine (Tylenol 1-4®), oxycodone (Percocet®, Percodan®), hydrocodone

(Vicodin®, Vicoprofen®, Lorset®), or tramadol (Ultracet®) (Rosenblum et al, 2008). There are

also single entity opioid formulations containing morphine (Avinza®, Kadian®, MS Contin®,

MSIR®), codeine (Codeine Contin®), oxycodone (OxyContin®), fentanyl (Duragesic®, Actiq®,

Fentora®), hydromorphone (Dilaudid®), oxymorphone (Opana®), and methadone (Dolophine®),

to name a few (Rosenblum et al, 2008).

1.1.3 Pharmacology

1.1.3.1 Opioid receptors

There are three major families of opioid receptors (OR): µ-opioid receptors (MOR), δ-opioid

receptors (DOR), and κ-opioid receptors (KOR). Beyond analgesia, opioid receptors are involved

in other physiological functions, such as membrane ionic homeostasis, cell proliferation, emotional

response, epileptic seizures, immune function, feeding and obesity, respiratory and cardiovascular

control, and some neurodegenerative diseases (Feng et al, 2012). ORs are highly expressed in the

CNS, though they are also found in many peripheral organs (e.g. heart, lungs, gastrointestinal tract)

(Feng et al, 2012; Wittert et al, 1996).

All families of ORs are seven transmembrane G protein-coupled receptors (GPCRs) and show

high sequence homology (>50%) between subtypes (Katritch et al, 2013). Upon ligand binding,

receptor conformational changes result in intracellular binding of Gi/o protein, guanosine

triphosphate (GTP) replacement of guanosine diphosphate (GDP), and dissociation of the trimeric

Gi/o complex into Gα and Gβγ proteins (Stein, 2016). Gα inhibits adenylyl cyclase and cyclic-

Page 25: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

3

adenosine monophosphate (cAMP) production, whereas Gβγ interacts directly with membrane ion

channels (Stein, 2016). All three families of ORs modulate pre- and post-synaptic calcium (Ca2+)

channels, suppressing Ca2+ influx, thereby decreasing neuronal excitability and reducing the

release of pro-nociceptive neuropeptides (Wang et al, 2010). Furthermore, OR activation leads to

the opening of G protein-coupled inwardly rectifying potassium and sodium (Na+) channels,

among others, summing to an overall decrease of neuronal excitation by neuron hyperpolarization,

the prevention of action potential firing, and decreased transmission of stimulus (Luscher and

Slesinger, 2010). Activation of each of the three OR subtypes produces analgesia, but with

different side-effects, likely due to variable regional expression, plasticity, and activity of receptors

in different parts of the central and peripheral systems (Stein, 2016). For example, respiratory

depression and euphoria are associated with activation of MORs, convulsions and anxiolysis with

DORs, and anti-inflammatory effects with KORs (Stein and Machelska, 2011; Zollner and Stein,

2007).

Endogenous opioid receptor ligands are derived from precursors proopiomelanocortin (β-

endorphin), proenkephalin (met- and leu-enkephalin), and prodynorphin (dynorphins). In general,

these peptides are not selective to one receptor subtype, due in part to 1) similarity in peptide

structure, as most endogenous opioids contain an N-terminal tyrosine residue required for ligand-

binding to ORs; 2) similarity in OR subtype structure and resultant intracellular signal transduction

pathways; and 3) the formation of homo and heteromeric complexes between opioid receptors that

modify their response to ligand binding (Ananthan, 2006; Feng et al, 2012; Law and Loh, 1999;

Waldhoer et al, 2004). β-endorphin and the enkephalins act primarily at MORs and DORs to

produce antinociception, while dynorphins can elicit both pro and antinociceptive effects by N-

methyl-D-aspartate (NMDA) and KOR receptor activation, respectively (Stein, 2016).

Endogenous opioids should be considered as adjuvants to opioid treatment, as preventing

extracellular degradation of endogenous opioids by inhibiting peptidases (e.g. aminopeptidase N

and neutral endopeptidase) in the CNS and periphery has been shown to produce analgesia in

animal models (Roques et al, 2012).

The regional distribution of OR subtypes in the brain and in peripheral tissue have been

documented across multiple species (including human and rat) using techniques including

Page 26: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

4

immunohistochemistry, in situ hybridization, and radiolabeled ligand binding (Mansour et al,

1995; Peng et al, 2012). In general, these studies show high correlation between OR mRNA

expression, protein expression, and ligand binding (Brodsky et al, 1995). Investigating region-

specific mRNA levels in human post mortem brains, MOR was found to be widely distributed

throughout the CNS with the highest transcript levels in the cerebellum; DOR transcript levels

were greatest in the cerebral cortex, putamen, temporal lobe, and hippocampus; and KOR

transcript levels were highest in the putamen (Peng et al, 2012). All three OR subtypes have high

mRNA expression in the nucleus accumbens and caudate, consistent with their role in regulating

pain perception (Peng et al, 2012). Differences in OR protein expression between species is also

observed; for example, while all three OR subtypes have been identified and cloned in the rat brain,

the rat cerebellum has abundant DOR but little-to-moderate MOR expression, while the human

cerebellum has abundant MOR but little DOR protein (Mrkusich et al, 2004).

1.1.3.2 Opioid metabolism

Opioid compounds differ widely with respect to how they are metabolized, to what extent, and by

which enzymes; however, there are several general patterns that can be discerned. Enzymatic

metabolism of opioids produces more hydrophilic compounds (by dealkylation, hydrolysis, and

oxidation) capable of undergoing renal excretion (Smith, 2009, 2011). Hepatic metabolism of

opioids primarily involves structural modifications via cytochrome P450 (CYPs; phase I

metabolism) and conjugation via uridine 5’-diphospho-glucuronosyl-transferase (UGT) enzymes

(phase II metabolism) (Smith, 2011). These metabolites are often active and may interact to

varying degrees with the same or different OR subtypes as the parent compound (Smith, 2011).

Most opioids undergo first-pass metabolism in the liver, which may reduce or enhance their

bioavailability (Somogyi et al, 2007). Phase I metabolism of opioids primarily involves CYP3A4

and CYP2D6 to different degrees depending on the opioid. For example, CYP3A4 is the primary

metabolic enzyme for oxycodone in humans, producing noroxycodone by N-demethylation (45%

of urinary recovery), while CYP2D6 facilitates O-demethylation of a smaller proportion of

oxycodone to oxymorphone (11% of urinary recovery) (Klimas et al, 2013). CYP3A4 metabolizes

more than 50% of all clinically used drugs, therefore drug-drug interactions can occur with co-use

of CYP3A4 inducers and/or inhibitors and opioids, that are extensively metabolized by this

enzyme (Smith, 2009). Individuals who ingested grapefruit juice, which contains bergamottin a

Page 27: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

5

strong inhibitor of CYP3A4, for 5 days, had a 1.5-fold greater peak plasma oxycodone

concentration and 1.7-fold greater overall oxycodone exposure (area under the concentration-time

curve [0 – ∞]) after oral oxycodone administration (Nieminen et al, 2010). CYP2D6 also plays a

prominent role in the metabolism of opioids, for example in the O-demethylation of codeine to

morphine (15% by urinary recovery, compared to 10% metabolized through CYP3A4 to

norcodeine) (Thorn et al, 2009). In general, CYP2D6 metabolically activates opioids to

compounds with greater MOR affinity than the parent compound (e.g. morphine relative to

codeine, and oxymorphone relative to oxycodone), whereas CYP3A4 metabolism of opioids

produces compounds with less activity than the parent compound (e.g. norcodeine relative to

codeine, and noroxycodone relative to oxycodone) (Somogyi et al, 2007). Consequently, research

into the inter-individual variability in opioid response has primarily focused on variability in

CYP2D6 activity (further discussed in 1.2, 1.3, and 1.4 inclusive).

Phase II metabolism of opioids represents another major metabolic pathway. Glucuronidation of

opioids generally takes place on free hydroxyl groups, and preferentially on aromatic hydroxyl

groups (e.g. C-3 of morphine) rather than alicyclic hydroxyl groups (e.g. C-6 of morphine) (Smith,

2011). Metabolism of morphine occurs primarily by UGT2B7 and UGT1A1 glucuronidation to

morphine-3-glucuronide (60% of urinary recovery) and morphine-6-glucuronide (10% of urinary

recovery) (Christrup, 1997); thus, genetic variation in UGT (e.g. UGT2B7) enzymatic

glucuronidation of opioids may influence opioid excretion and response (Innocenti et al, 2008).

Liver samples containing a UGT2B7 variant responsible for increased enzymatic activity showed

increased morphine glucuronidation in vitro (45% versus samples without the variant) (Innocenti

et al, 2008).

1.1.4 Opioid neuropharmacology

1.1.4.1 Analgesia

The neural pathway of pain starts at the dorsal horn of the spinal cord and ascends through the

contralateral spinothalamic tract to the brain where pain processing occurs (Ossipov et al, 2010).

Pain is processed as nociception, which refers to all thermal, mechanical, or chemical stimuli

detected by nociceptors, whose cell bodies are located in the dorsal root ganglia (Basbaum et al,

2009). When noxious stimuli occur, nociceptors release glutamate, generating excitatory

Page 28: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

6

postsynaptic currents in second order dorsal horns neurons; glutamate receptor activation

stimulates action potential firing and transmission of the noxious stimulus to higher order neurons

(Basbaum et al, 2009). Ascending projections target the thalamus, which sends projections to the

lateral and basolateral amygdala, while collateral projections target the mesencephalic nuclei

(rostral ventral medulla and periaqueductal grey) (Ossipov et al, 2010). Initial opioid analgesia can

occur by direct activation of ORs expressed in the dorsal root ganglia, reducing pain transmission

at this first synapse (Stein, 1995).

The primary mechanism behind opioid analgesia is descending modulation (supraspinal) with

action at multiple brain regions that have high OR concentration, such as the periaqueductal grey

(PAG), locus coeruleus, and rostral ventral medulla (RVM) (Ossipov et al, 2010; Pathan and

Williams, 2012). The PAG-RVM-spinal cord pathway is an essential neural circuit for opioid-

based antinociception or analgesia; the PAG receives input from the hypothalamus and amygdala,

then synapses with the RVM, which in turn terminates in the dorsal horn (Figure 1) (Basbaum and

Fields, 1984; Loyd and Murphy, 2009). Administration of morphine directly into the PAG

produces MOR-dependent analgesia (reversible by naloxone, a MOR antagonist), as well as

naloxone-reversible excitation of RVM neurons (Behbehani and Fields, 1979; Loyd and Murphy,

2009; Yaksh and Rudy, 1978). Similarly, lesioning the PAG, or administrating MOR antagonists,

attenuates the antinociceptive effects of morphine administered systemically (Dostrovsky and

Deakin, 1977; Loyd and Murphy, 2009; Ma and Han, 1991). Opioids are believed to increase

descending inhibitory control (through the PAG-RVM-spinal cord circuit) of spinal nociceptive

output by an indirect action (Kanjhan, 1995; Ossipov et al, 2010). Activation of ORs in the RVM

involves excitation (or disinhibition) of off-cells, neurons that decrease action potential firing, and

direct inhibition of on-cells, neurons that increase action potential firing (Kanjhan, 1995; Ossipov

et al, 2010). Disinhibition of off-cells may involve direct OR-mediated presynaptic inhibition of

GABAergic input, while on-cells are directly hyperpolarized by the electrophysiological

consequences of OR activation (discussed previously in 1.1.3.1) (Li et al, 2015b).

Page 29: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

7

Figure 1: Neural pathway of nociception and descending modulation by opioids

Figure 1: Neural pathway of nociception and descending modulation by opioids. Ascending

pain pathways are activated by noxious stimuli (red arrows). Opioid analgesics (blue circles) act

on opioid receptors located in descending pain modulatory pathways (blue arrows) in the

periaqueductal gray (PAG), rostroventral medulla (RVM), dorsal horn, and dorsal root ganglion

(DRG) to downregulate ascending nociception (red arrows). Adapted from Ossipov et al. 2010.

Opioids may also act in the anterior cingulate cortex and insula, which are two components of the

central limbic (emotional) system involved in encoding the emotional and motivational aspects of

pain (Bushnell et al, 2013). Imaging studies show a relationship between emotional aspects of pain

perception and neural activation of the anterior cingulate cortex and insula, as well as with

endogenous activation of MORs in these regions, while lesioning these regions results in altered

emotional responses to pain (Bushnell et al, 2013; Starr et al, 2009; Zubieta et al, 2001).

1.1.4.2 Tolerance

Repeated opioid administration results in tolerance, a reduction in opioid effect or drug potency,

indicated by a rightward shift in the opioid dose response curve (Cahill et al, 2016); the rate of

tolerance varies by opioid compound and by opioid pharmacological effect. For example, altered

gut motility (constipation) exhibits minimal tolerance over time compared to other side-effects of

repeated opioid use such as respiratory-depression, sedation, and nausea (Cahill et al, 2016). While

Page 30: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

8

tolerance to opioids per se has not been demonstrated to be a reliable predictor of abuse liability,

it presents a challenge for opioid treatment of chronic pain and relapse in opioid-dependent

individuals (Cahill et al, 2016). Decreased analgesic potency and inadequate pain relief is a

predictor of dose-escalation in pain patients. Tolerance to opioid-induced respiratory depression

at higher opioid doses may cause mortality in opioid-dependent individuals who relapse after

prolonged abstinence, due to loss of tolerance to opioid-induced respiratory side effects (Cahill et

al, 2016; Siegel et al, 1982).

Numerous mechanisms contribute to opioid tolerance at a neurobiological level, including opioid

receptor desensitization, phosphorylation, internalization, and down-regulation and the initiation

of compensatory processes (like adenylyl cyclase superactivation) (Williams et al, 2013). OR

phosphorylation is mediated by protein kinases (e.g. GPCR kinases, protein kinase C) and is a key

initiating event for OR desensitization, i.e. the progressive loss of OR function after repeated

exposure to an opioid (Bailey et al, 2009). Phosphorylation induces an OR conformation change

that increases the affinity for β-arrestin proteins. Binding of β-arrestin to ORs uncouples G-protein

signalling and recruits endocytotic machinery, leading to receptor internalization and degradation

(Ferguson et al, 1996). Downregulation is a long-term cellular adaptation to repeated opioid

administration, which may result from repeated degradation of internalized receptors and/or from

a decrease in receptor synthesis (Koch and Hollt, 2008). Compensatory mechanisms that

counteract OR function further contribute to opioid tolerance; for example, counterregulatory

increases in adenylyl cyclase activity (adenylyl cyclase superactivation) are seen after OR

withdrawal and are thought to contribute to the development of opioid tolerance and subsequently

to dependence (Chakrabarti et al, 1998a; Chakrabarti et al, 1998b).

Neural outcomes of prolonged opioid use (e.g. OR phosphorylation and desensitization) contribute

to tolerance in a complex way and are agonist-dependent. For example, protein kinase C is

involved in MOR phosphorylation (leading to desensitization) during morphine- but not DAMGO

(MOR agonist)-mediated receptor activation, while MOR endocytosis mediated by β-arrestin

occurs during DAMGO, but not morphine, activation (Celver et al, 2004; Johnson et al, 2006;

Koch and Hollt, 2008). Agonists that do not induce endocytosis (like morphine) tend to

Page 31: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

9

preferentially activate adaptive processes (e.g. adenylyl cyclase superactivation), leading to

tolerance and subsequent dependence (Koch and Hollt, 2008).

1.1.4.3 Dependence

The biological mechanisms underlying opioid dependence involve OR activation (primarily MOR)

in brain regions associated with reward and reinforcement, including the ventral tegmental area

(VTA) (Le Merrer et al, 2009; Ting-A-Kee and van der Kooy, 2012). The VTA consists primarily

of dopamine-producing cell bodies that project to the nucleus accumbens and further to the cortex,

which mediate natural and drug-motivated behaviours; dopamine is the primary neurotransmitter

implicated in positive reinforcement (Wise and Rompre, 1989). Rats and mice readily self-

administer MOR-agonists into the VTA, and conditioned place preference produced by MOR

agonists given systemically in rats is blocked by intra-VTA MOR-antagonist administration

(Bozarth and Wise, 1981; Britt and Wise, 1983; Fields and Margolis, 2015). While VTA dopamine

release has been regarded as the predominant theory of opioid-seeking motivation, dopamine-

independent reinforcement may occur at the brainstem tegmental pedunculopontine nucleus (Ting-

A-Kee and van der Kooy, 2012). However, relatively less is known about this non-dopaminergic

VTA circuitry and its comparative impact is unclear (Ting-A-Kee and van der Kooy, 2012).

Regulation of VTA dopamine release is complex and multifactorial, involving dopaminergic, γ-

aminobutyric acid (GABA)-ergic, and glutamatergic regulation (Figure 2). OR activation produces

inhibition, either directly through hyperpolarization of neurons via activation of G protein-coupled

inwardly rectifying potassium and Na+ channels, or indirectly through inhibiting neurotransmitter

release (Williams et al, 2001). The majority of MORs in the VTA are expressed on GABAergic

cells (Fields and Margolis, 2015). MOR agonists promote the release of dopamine in the VTA via

disinhibition, i.e. removal of tonic GABAergic inhibition of dopamine neurons (Fields and

Margolis, 2015; Kelley et al, 1980). This ‘two neuron’ model of opioid reward has been assessed

in vitro; in rat VTA tissue, half of the immunocytochemically identified GABA neurons were

inhibited (hyperpolarized) by DAMGO (Margolis et al, 2012). However, MOR activation in the

VTA also inhibits GABA release onto non-dopaminergic neurons (Xia et al, 2011), and inhibits

pre-synaptic glutamate release from terminals synapsing onto VTA neurons (Margolis et al, 2005).

Despite this, glutamate signaling appears to be required in opioid reward; disrupting glutamate

Page 32: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

10

neurotransmission in rat VTA blocked morphine-induced VTA dopamine release, and the

acquisition of morphine CPP (Harris et al, 2004). Therefore, all physiological input contributing

to opioid-induced VTA dopaminergic transmission and ultimately opioid reward and

reinforcement is yet to be fully understood.

Figure 2: Neural pathway of opioid dependence

Figure 2: Neural pathway of opioid dependence. Opioid analgesics (blue circle) increase

dopamine transmission (red arrows) from the ventral tegmental area (VTA) to the nucleus

accumbens (NAc), and further the prefrontal cortex, by disinhibition (blue arrow) of GABA cells

acting on dopaminergic projections from the VTA. Adapted from Ting-A-Kee and van der Kooy

2012.

1.1.5 Opioid epidemic; misuse and abuse

Non-medicinal prescription opioid (PO) use and abuse in Canada and the United States has

increased rapidly over the past three decades, along with the associated personal, economic, and

healthcare burden from opioid morbidity and mortality, leading to what has been termed the

“opioid epidemic” (Florence et al, 2016; Han et al, 2015). In 2016, more than 11 million people

in the United States (12.5 % of PO users) reported using POs non-medicinally, defined as use of a

medicine for a purpose other than as directed or indicated, whether willfully or unintentionally

(NSDUH, 2017; Tetrault and Butner, 2015). Similarly, in Canada, POs are the second most

prevalent drugs used illicitly (Fischer and Rehm, 2017). In 2014, US retail pharmacies dispensed

245 million opioid prescriptions, with between 9 and 11 million people receiving longer-term

opioid therapy (Volkow and McLellan, 2016). Emergency department visits involving misuse or

abuse of POs increased 1.5-fold between 2004 and 2011, while admissions to substance-abuse

Page 33: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

11

treatment programs due to PO use increased more than 4-fold between 2002 and 2012 (Compton

et al, 2016). Between 2000-2014, the US death rate from PO overdose increased from 1.5 to 5.9

per 100,000 persons, with a similar increase in death rate from heroin overdose alone, from 0.7 to

3.4 per 100,000 persons (Compton et al, 2016). In 2015, drug overdoses accounted for over 52

thousand deaths in the United States, 63% of which involved an opioid (Rudd et al, 2016); among

these opioid-related deaths, 45% (approximately 15 thousand) involved a prescription opioid (Guy

et al, 2017).

Effectively managing the opioid epidemic is difficult due to the utility of opioids in managing

chronic pain; at any given time, between 30 – 40% of the US population reports experiencing acute

or chronic pain (Wilson-Poe and Moron, 2017). When asked for the main reason for most recent

opioid use, the majority of respondents (62.3%) report using POs to relieve physical pain, while

significantly fewer (12.9%) reported use to feel good or get high (NSDUH, 2017). The source of

obtaining POs also ranges; while a proportion of individuals in the US who misused POs in 2016

had them prescribed by a doctor (35.4%), more than half obtained POs from a friend or relative

(53%), and often for free (40%) (NSDUH, 2017). Additionally, due to various factors in PO supply

and price, dangers linked to the seeking of alternative opioids (e.g. heroin) have emerged. PO users

are 40-fold more likely to become heroin users, and in both Canada and the US, spikes in heroin

use have been observed when PO availability was curtailed by selective PO intervention (e.g. de-

scheduling of specific POs, restricting prescribing guidelines, etc.) (Compton et al, 2016; Dart et

al, 2015; Fischer and Rehm, 2017; Jones, 2013; Lake et al, 2016). In 2016, roughly 950 thousand

individuals in the US had used heroin, while approximately 67% of those used POs and heroin

concomitantly (NSDUH, 2017). More recently, the trajectory of PO abuse has led to seeking illicit

PO analogs, such as fentanyl, as well as the lacing of POs with these illicit analogs, creating an

additional aspect to the opioid epidemic (Fischer and Rehm, 2017). Over a six-month period in

2014 in one Florida Medical Examiner District, 97% of the overdose deaths reported involved

accidental drug intoxication with fentanyl as a sole or contributing factor (Lee et al, 2016).

The increase in opioid abuse over the last two decades appears to predominate in higher socio-

economic countries like Canada and the US (Fischer and Rehm, 2017; Fischer et al, 2006). Canada

and the US account for roughly 80% of world PO consumption (Figures 3 and 4) (NSDUH, 2017).

Page 34: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

12

This is due in part to POs being unavailable in most less-resourced world countries. However, this

may also be due to the high availability of, and ease of access to, POs in North America, in which

the medical system may rely more heavily on pharmaceutical treatments (Fischer and Rehm, 2017;

Lohman et al, 2010). For example, in looking at commonly prescribed opioid compounds (often

in combination with anti-inflammatories; see section 1.1.2), 10.4% of 26.5 million codeine users

and 14.1% of 27.6 million oxycodone users reported misuse within the last year (Figure 5). There

is a strong correlation between the amount of medical dispensing of POs and harmful PO-related

outcomes (e.g. morbidity and mortality) (Wisniewski et al, 2008).

Figure 3: Region-specific percentage of daily opioid consumption in 2001-2003, compared to 2011-2013

Figure 3: Region-specific percentage of daily opioid consumption in 2001-2003, compared to

2011-2013. Adapted from the 2016 International Narcotics Control Board (INCB) availability

report.

Page 35: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

13

Figure 4: Country-specific opioid consumption per capita, and the prevalence of opioid misuse, in 2016

Figure 4: Country-specific opioid consumption per capita, and the prevalence of opioid

misuse, in 2016. Adapted from the 2016 International Narcotics Control Board (INCB)

availability report.

Figure 5: Total opioid use and reported misuse in individuals above the age of 12 in the US in 2016, split by primary opioid compound

Figure 5: Total opioid use and reported misuse in individuals above the age of 12 in the US

in 2016, divided by primary opioid compound. Axis labels: OC, Oxycodone; OM,

Oxymorphone; COD, Codeine; MOR, Morphine: HC, Hydrocodone; HM, Hydromorphone; FEN,

Fentanyl; TRAM, Tramadol; DEM, Demerol; MD, Methadone. Adapted from the National Survey

on Drug Use History, detailed tables from 2016.

To

tal

use

rs (

Age

12

or

abo

ve,

in t

ho

usa

nd

s)

Page 36: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

14

One consideration in addressing the epidemic, and balancing the care and needs of pain patients,

is the spread of economic burden resulting from PO overdose, abuse, and dependence. In 2013,

the aggregate economic burden of PO abuse and dependence in the US was estimated to be

between 70 to 87 billion dollars (Florence et al, 2016). Within this estimate, non-fatal cases

accounted for almost two thirds of the costs in health care ($26 billion, 33%), substance abuse

treatment ($3 billion, 4%), and lost productivity ($20 billion, 26%) (Florence et al, 2016). Fatal

cases accounted for a little more than one quarter of the costs (specifically in lost productivity and

health care costs; $21 billion, 27%), while criminal justice (police protection, correctional

facilities, legal fees) accounted for the last 10% of economic burden ($8 billion) (Florence et al,

2016). Quantifying some of the adverse health and economic impacts associated with POs may

help decision makers select where to focus for mitigating future, and treating current, opioid

dependence, to reduce the productive life-years lost to PO morbidity and mortality.

1.2 – CYPs

1.2.1 General introduction

Cytochrome P450 (CYP) enzymes are a superfamily of membrane-bound hemoproteins,

responsible for metabolizing xenobiotic and endogenous compounds (Cook et al, 2016; Miksys

and Tyndale, 2013). CYP-mediated metabolism is a complex, multistep process involving

oxidation of a substrate after binding to the active site in the heme domain (Cook et al, 2016).

Molecular oxygen is activated and incorporated into the CYP substrate, forming water as a by-

product; this mechanism of oxidation is the basis for numerous reactions including hydroxylation,

epoxidation, deamination, and dealkylation (Hannemann et al, 2007). CYP-mediated metabolism

generally produces a hydrophilic metabolite, which is more easily excreted from the human body

(Zanger and Schwab, 2013). These metabolites may have decreased (inactivation), increased

(activation), or un-altered, pharmacologic activity compared to the parent compound. CYP

metabolism of drugs occurs predominantly in the liver, where CYPs are highly expressed and are

largely responsible for systemic drug and metabolite levels.

CYPs are divided into families, based on amino acid sequence homology (Nelson, 2006). While

the majority of human CYPs function to maintain endogenous compound homeostasis (e.g.

biosynthesis of steroid hormones, prostaglandins, bile acids, etc.), enzymes belonging to CYP

Page 37: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

15

families 1-3 metabolize a large proportion of clinically indicated drugs and xenobiotics (Zanger

and Schwab, 2013). For example, over 30% of clinically used drugs are metabolized by CYP3A4

(e.g. atorvastatin), while 20% are metabolized by CYP2D6 (e.g. dextromethorphan), 12.8% by

CYP2C9 (e.g. diclofenac), 8.9% by CYP1A2 (e.g. caffeine), and 7.2% by CYP2B6 (e.g.

bupropion) (Zanger and Schwab, 2013).

While CYP metabolism primarily occurs in the liver, extrahepatic (e.g. in the brain) CYP

expression may alter drug activation/inactivation within a tissue/organ and thereby drug response.

A large proportion of CNS-acting drugs are metabolized by CYP enzymes, particularly by the

CYP2 family, to active and/or inactive metabolites (Zanger et al, 2004). Therefore, target-tissue

drug metabolism in the brain may impact CNS-acting drug response contributing, in part, to why

peripheral drug and/or metabolite levels do not always correlate to CNS-acting drug response

(discussed further in 1.4) (Michels and Marzuk, 1993; Miksys and Tyndale, 2013).

1.2.2 CYP2D6 enzyme

1.2.2.1 Substrates and clinical drugs of interest

CYP2D6 is an important enzyme, as it metabolizes approximately 20% of clinically used drugs

(Zanger et al, 2004). CYP2D6 shows a high degree of interindividual variability, primarily due to

genetic polymorphisms, resulting in altered therapeutic consequences for CYP2D6 substrates

(more in section 1.2.2.2). The enzyme was first characterized in humans as a consequence of

interindividual variability in drug response. An inherited monogenic trait resulting in differing

interindividual response to sparteine (an antiarrhythmic) and debrisoquine (an antihypertensive),

referred to as the sparteine/debrisoquine polymorphism, was subsequently demonstrated to be due

to a protein of the human CYP2D subfamily (Eichelbaum et al, 1987; Gonzalez et al, 1988). The

CYP2D gene locus spans 45 kilobases on chromosome 22 (22q13.1) and contains three distinct

genes: CYP2D6, CYP2D7 (shares sequence homology with CYP2D6 except for a base insertion in

exon 1), and CYP2D8 (a pseudogene with sequence variations that prevent the formation of

functional protein) (Gaedigk, 2013).

Substrates of CYP2D6 show structural similarities, including a planar hydrophobic aromatic ring

and a nitrogen atom at a distance of about 5-7 Å from the site of oxidation, therefore initial

Page 38: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

16

pharmacophore models using these arrangements were developed to predict compounds likely to

be substrates (Zanger et al, 2004). Crystal structures of CYP2D6 with and without substrate bound

have been developed for improving understanding of the active site architecture of the enzyme and

have employed more accurate computational approaches to examine substrate binding (Rowland

et al, 2006; Wang et al, 2012).

CYP2D6 metabolizes numerous xenobiotic compounds, including drugs that target the CNS and

cardiovascular systems, as well as endogenous compounds. This includes antidepressants (e.g.

fluoxetine), antipsychotics (e.g. haloperidol), analgesics (e.g. codeine), antiarrhythmics (e.g.

propafenone), β-blockers (e.g. propranolol), amphetamines (e.g. 3,4-

methylenedioxymethamphetamine), plant alkaloids (e.g. ibogaine), neurotoxins (e.g. 1-methyl-4-

phenyl-1,2,3,6-tetrahydropyridine [MPTP]), and endogenous neurochemicals (e.g. tyramine, 5-

methoxytryptamine) (Table 1) (Miksys and Tyndale, 2013; Mo et al, 2009).

Page 39: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

17

Table 1: Examples of CYP2D6 substrates and the reactions catalyzed by CYP2D6.

Table 1: Examples of CYP2D6 substrates and the reactions catalyzed by CYP2D6

Adapted from Zanger et al. 2004 and Miksys and Tyndale 2013.

1.2.2.2 Variation in CYP2D6

1.2.2.2.1 Genetic sources of variation

The majority of CYP2D6 allelic variants are characterized by the presence of one or more

nucleotide variations, including single nucleotide polymorphisms (SNPs), insertions, and deletions

(Gaedigk, 2013). To date, over 80 sequence variants have been identified in CYP2D6 resulting in

completely inactive function, reduced function, increased function (via additional functional gene

copies), or no influence on the function of CYP2D6 enzyme activity

(https://www.pharmvar.org/gene/CYP2D6). For example, the CYP2D6*10 variant allele contains

Page 40: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

18

a mutation in a sequence necessary for folding of the CYP2D6 protein, therefore results in reduced

protein stability and function (Ingelman-Sundberg, 2005). Gene duplications occur when

individuals express multiple CYP2D6 copies on the same allele, with the majority being

CYP2D6*2 (2850C>T and 4180G>C SNPs) deemed CYP2D6*2xN (with N representing the

number of additional gene copies) (Aklillu et al, 1996; Dahl et al, 1995; Lundqvist et al, 1999).

CYP2D6*1, *2, and *4 are the most common alleles which are duplicated, but duplication events

occur in numerous other functional, non-functional, and reduced function alleles (Figure 6)

(Gaedigk, 2013; Lundqvist et al, 1999).

Figure 6: Genomic impact of select CYP2D6 variant alleles

Figure 6: Genomic impact of select CYP2D6 variant alleles. Predicted structures and functional

impacts of multiple CYP2D6 alleles; the DNA impacts of mutations (green circles) are shown

above the alleles and the amino acid impacts below, while open reading frames are indicated by

grey shaded boxes. DNA abbreviations: A, adenosine; C, cytidine; G, guanosine; T, thymidine.

Amino acid abbreviations: H, histidine; L, leucine; M, methionine; P, proline; R, arginine; S,

serine; T, threonine. Adapted from Zanger et al. 2004.

Page 41: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

19

CYP2D6 substrates primarily (>80%) metabolized by CYP2D6, such as debrisoquine or

dextromethorphan, have been used for in vivo phenotyping of individuals into distinct CYP2D6

activity groups (Zanger et al, 2004). These phenotype groups, in order of ascending activity, are

poor metabolizers (PM), intermediate metabolizers (IM), extensive metabolizers (EM), and ultra-

rapid metabolizers (UM) (Zanger et al, 2004). Translating genotypes into clinically useful

information is difficult, owing to the potentially hundred(s) of genotype combinations (diplotypes)

that could exist in a population and due to overlap in phenotypic grouping. One cohort of 672

individuals was genotyped to have 95 different CYP2D6 diplotypes (Gaedigk et al, 2008), while

over 100 unique diplotypes were identified in the first 800 individuals enrolled in the PG4KDS

study (Hoffman et al, 2014). Attempts have been made to better categorize CYP2D6 phenotypes

from different genotypes, such as assigning an activity score to each allele (null: 0, reduced: 0.5,

normal: 1, or increased: 2) and using the sum diplotype activity score as a metric of CYP2D6

activity (Gaedigk and Coetsee, 2008; Gaedigk et al, 2008).

1.2.2.2.1.1. Interethnic variability

The frequencies of CYP2D6 polymorphisms and resulting phenotype groups vary across

ethnicities as specific CYP2D6 variant alleles are found at higher and lower frequencies in different

populations. White European populations have a high incidence of PMs at 3-10% of the total

population, while Black Africans and African Americans range between 2-7% and East Asian

populations between 0-2% (Teh and Bertilsson, 2012). This is due, in part, to the higher frequency

of null-function CYP2D6*3, *4, *5, *6, and reduced-function *10 and *41 alleles in White

European populations, with low frequencies of CYP2D6*3 and *4 alleles in East Asian and

Oceania populations (Gaedigk, 2013; Teh and Bertilsson, 2012). CYP2D6*36 contributes

substantially to the PM status in Asian (3% allele frequency) and African American (0.5%)

populations (Gaedigk et al, 2006). The frequencies of common CYP2D6 variant alleles are

displayed for select ethnicities in Figure 7.

Page 42: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

20

Figure 7: Select CYP2D6 allele frequencies across ethnic groups

Figure 7: Select CYP2D6 allele frequencies across ethnic groups. Combined results of 172

independent studies. Black: Black Africans and African Americans; Asian: South Asians and East

Asians; Native: Native Oceanians, Native Americans, and Circumpolar Populations; White:

Caucasian American, Scandinavian, European, Mediterranean, and Slavs. Adapted from Llerena

et al. 2014.

The reduced function CYP2D6*10 allele is the most common variant allele in Asians; in Chinese,

CYP2D6*10 comprises 51-70% of all CYP2D6 alleles (Johansson et al, 1994; Johansson et al,

1991). This may account for the lower overall mean CYP2D6 enzyme activity in Asians compared

to Caucasians, as well as an overall rightward shift in the median CYP2D6 metabolic ratio in

Chinese EMs (Johansson et al, 1994; Johansson et al, 1991). Europeans have the lowest frequency

of CYP2D6*10 at 1-2%, while the overall frequency in other populations ranges from 3-7%

(Gaedigk, 2013). Black African and African American populations have a high frequency of the

CYP2D6*17 allele (14-26%), which contains two missense mutations (850C>T SNP and

4180G>C) resulting in an altered CYP2D6 active site (Gaedigk, 2013; Teh and Bertilsson, 2012).

The CYP2D6*17 allele is also found among other populations with African founders such as

Brazilians (Kohlrausch et al, 2009), Cubans (Llerena et al, 2012), Trinidadians (Montane Jaime et

al, 2013), and South Africans (Dodgen et al, 2013; Wright et al, 2010).

The percentage of CYP2D6 gene duplications or multiplications, resulting in CYP2D6 UMs, also

varies across populations. The allele frequency of CYP2D6*1xN or *2xN in European populations

Page 43: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

21

is around 2% (e.g. 1-2% in Swedish Caucasians) which is comparative to Japanese, Chinese, and

Indian populations (Aklillu et al, 1996; Kubota et al, 1996; Qin et al, 2008). Frequencies of

CYP2D6 duplications are higher in Mediterranean (e.g. 7-10% in Spanish) and North African (e.g.

29% in black Ethiopians) populations (Aklillu et al, 1996; Teh and Bertilsson, 2012).

1.2.2.2.1.2 Functional impact of CYP2D6 polymorphism

CYP2D6 polymorphism results in a range of CYP2D6 enzyme activity which alters drug

metabolism and response, drug efficacy (e.g. activation or inactivation), and adverse drug effects

(e.g. toxicity). There is extensive evidence of individuals with different CYP2D6 phenotypes

having altered response to psychotropic drugs (e.g. haloperidol), cardiovascular drugs (e.g.

metoprolol), antineoplastics (e.g. tamoxifen), and analgesics (e.g. codeine, discussed further in

1.3) (Teh and Bertilsson, 2012). CYP2D6 genotype has also been associated with disease risk

including for Alzheimer’s disease and Parkinson’s disease (Golab-Janowska et al, 2007).

Most antipsychotics, including haloperidol, are metabolized by CYP2D6. In a random sample of

schizophrenic inpatients treated with haloperidol an increased number of active CYP2D6 genes

correlated with increased haloperidol clearance and trended towards lower haloperidol therapeutic

efficacy (Brockmoller et al, 2002; Teh and Bertilsson, 2012). The occurrence of pseudo-

parkinsonism (a haloperidol-induced adverse reaction), among other extrapyramidal side effects,

was higher in CYP2D6 PMs, likely due to the reduced metabolic clearance of haloperidol in these

individuals (Brockmoller et al, 2002).

Metoprolol is a cardio-selective β-blocker used in the treatment of hypertension, angina pectoris,

cardiac arrhythmia, and chronic heart failure; CYP2D6 is responsible for roughly 75% of the total

metabolism of metoprolol through α-hydroxylation (10% of total dose) and O-demethylation

(65%) (Borg et al, 1975; Prakash and Markham, 2000). Total plasma exposure to metoprolol is 4-

to 6-fold higher in CYP2D6 PMs than EMs after a single dose, while there is a 10-fold difference

in metoprolol clearance between PMs and UMs (Hamelin et al, 2000; Kirchheiner et al, 2004).

PMs have a 2-fold greater metoprolol-induced cardiovascular effect (reduced exercise-induced

heart rate) than UMs, and they have a 5-fold higher risk of developing adverse effects to metoprolol

(Kirchheiner et al, 2004; Wuttke et al, 2002).

Page 44: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

22

Tamoxifen is a selective estrogen receptor modulator used as a treatment for breast cancer

(Schroth et al, 2009) Tamoxifen is metabolized into an active metabolite endoxifen by N-

demethylation and 4-hydroxylation carried out by CYP2D6 (Schroth et al, 2009). Individuals

homozygous for the null-function CYP2D6*4 and taking tamoxifen to treat estrogen receptor-

positive breast cancer have significantly decreased relapse-free survival compared to those

heterozygous or homozygous for CYP2D6*1 (wild-type) (Schroth et al, 2009). Similarly, Japanese

women with breast cancer homozygous for the reduced-function CYP2D6*10 had lower steady

state plasma levels of endoxifen from tamoxifen, shorter median time to disease, and higher

incidence of breast cancer recurrence within 10 years (Kiyotani et al, 2008; Lim et al, 2007; Lim

et al, 2011; Xu et al, 2008). While routine testing of women on tamoxifen has not been approved

for risk stratification, numerous reports support CYP2D6 genotype analysis being used as a

predictive biomarker for treatment success (Gaedigk and Coetsee, 2008).

1.2.2.2.2 Non-genetic sources of variation and drug-drug interactions

Beyond genetic polymorphism, non-genetic variation in CYP2D6 protein level can also occur.

Hepatic CYP2D6 displays constitutive expression during adulthood and is therefore relatively

unaltered by common CYP inducers like phenobarbital (Edwards et al, 2003; Stevens et al, 2008).

However, the CYP2D6 gene can be regulated by binding of transcription factors to the CYP2D6

promoter region, such as hepatocyte nuclear factor (HNF) 4α (Pan et al, 2017). In a human liver

cell line (HepG2), binding of HNF4α enhanced promotor activity of CYP2D6; HNF4α mRNA

expression correlated with microsomal CYP2D6 enzyme activity (Cairns et al, 1996; Yang et al,

2010). HNF4α knockdown decreased CYP2D6 expression in human hepatocytes, indicating that

HNF4α is required for constitutive CYP2D6 activity (Jover et al, 2001). Similarly, in a HepG2

cell line, overexpression of the transcription factor CAAT/enhancer-binding protein α increased

CYP2D6 mRNA (Jover et al, 1998; Pan et al, 2017).

Since CYP2D6 metabolizes a large portion of clinically used drugs, drug-drug interactions can

occur through competition or inhibition by a CYP2D6 substrate or inhibitor (Table 2). Inhibition

can occur by competitive inhibition (binding of an inhibitor in or near the enzyme active site

blocking substrate accessibility), irreversible inhibition (metabolism of a mechanism-based

Page 45: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

23

inhibitor (MBI) by CYP2D6 to a reactive intermediate that binds irreversibly to the enzyme

rendering it inactive), non-competitive inhibition (binding of an inhibitor to the enzyme away from

the active site, altering the conformation of the enzyme), or uncompetitive inhibition (binding of

an inhibitor to the enzyme after substrate has bound) (Bertelsen et al, 2003; Lin and Lu, 1998).

For example, propranolol, a non-selective β-adrenergic receptor blocker, is a MBI of CYP2D6;

propranolol is hydroxylated by CYP2D6 to 4-hydroxypropranolol which forms a reactive

intermediate that binds to, and inactivates, CYP2D6 (Rowland et al, 1994). Paroxetine, a selective

serotonin reuptake inhibitor, is also a potent MBI of CYP2D6 (Bertelsen et al, 2003). In

schizophrenic patients receiving risperidone (anti-psychotic and CYP2D substrate), paroxetine

dose-dependently increased plasma risperidone concentrations and associated extra-pyramidal

side effects (Saito et al, 2005). By comparison, quinidine is a potent and selective competitive

inhibitor of CYP2D6, but not a substrate itself (Branch et al, 2000; McLaughlin et al, 2005).

1.2.2.3 Species differences in CYP2D6 enzyme

CYP2D enzymes have been identified in numerous species including humans, rat, mouse, and

monkey. While humans have one functional isoform of CYP2D (CYP2D6) rats have six

(CYP2D1, 2, 3, 4, 5, and 18) with unique substrate specificity, metabolism, expression, and

inhibition profiles (Hiroi et al, 2002). For example, while rat CYP2D1 and CYP2D2 are the most

abundant isoforms in the liver, CYP2D4 is primarily expressed in rat brain (Wyss et al, 1995).

However, rat CYP2D enzymes collectively, are a useful model of human CYP2D, due to relatively

similar amino acid sequence identity (>70% for all isoforms) and the ability to perform metabolic

reactions in vitro similar to human CYP2D6 (Venhorst et al, 2003). Human CYP2D6 and rat

CYP2D2 exhibit high levels of 4-hydroxylation of debrisoquine and propranolol (Hiroi et al,

2002). Similarly, human CYP2D6 and rat CYP2D2 (along with rat CYP2D1) O-demethylate

codeine to morphine in vitro (Grobe et al, 2012).

Page 46: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

24

Table 2: Examples of competitive and mechanism-based CYP2D6 inhibitors.

Table 2: Examples of competitive and mechanism-based CYP2D6 inhibitors

Adapted from Ingelman-Sundberg 2005, Zanger et al. 2004, and Wang et al. 2009.

1.3 – CYP2D6 and opioids

1.3.1 CYP2D6 metabolism of opioids

CYP2D6 plays an important role in metabolizing opioids. This is due to 1) CYP2D6 metabolism

of opioids producing metabolites that are more potent in activating ORs, 2) CYP2D6

polymorphisms altering the formation of these metabolites and thereby altering therapeutic effect

or toxicity, and 3) other CYP2D6 substrates or inhibitors interacting with opioid metabolism

(Smith, 2009, 2011). For these reasons, genetic and non-genetic variability in CYP2D6 activity

have been studied in relation to inter-individual variation in opioid response, side effects,

tolerance, and abuse liability.

Page 47: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

25

While usually a minor metabolic pathway, the O-demethylation of opioids by CYP2D6 generally

produces a metabolite with greater OR affinity (Figure 8). This may result in a CYP2D6 metabolite

that modestly contributes to (e.g. hydrocodone to hydromorphone), or is entirely responsible for

(e.g. codeine to morphine), analgesia (Chen et al, 1991a; Grond and Sablotzki, 2004; Smith, 2009).

While the CYP2D6-mediated metabolism of codeine to morphine represents a relatively minor

pathway of codeine metabolism (0-15% of urinary recovery) morphine has a 3000-fold greater

affinity for the MOR than codeine; therefore, codeine analgesia depends on this metabolic

activation (Adler et al, 1955; Chen et al, 1991b; Pert and Snyder, 1973). While a similar percentage

of oxycodone is metabolized by CYP2D6 (11% of urinary recovery) to oxymorphone, a 44-fold

more potent MOR activating metabolite (9-fold greater than morphine), oxycodone itself also has

a relatively high affinity to MOR (only 5-fold less than morphine) and therefore, unlike codeine,

oxycodone does not require CYP2D6 activation to elicit analgesia (Klimas et al, 2013; Lalovic et

al, 2006).

1.3.2 Impact of CYP2D6 phenotype on opioid analgesia

The wide range of CYP2D6 metabolism phenotypes across populations makes it imperative to

understand the relative consequences on opioid metabolism and response. For active parent opioid

compounds primarily metabolized by CYP2D6, a lower clearance in PMs may produce higher

plasma concentrations of the parent compound and thereby more side-effects (Zahari and Ismail,

2014). Conversely, UMs may experience no or less pain relief leading to therapeutic failure (Zahari

and Ismail, 2014). For pro-drug opioids that require bioactivation by CYP2D6 to active

metabolites to elicit therapeutic effect (e.g. codeine to morphine) PMs may produce no CYP2D6-

mediated metabolite and therefore experience no analgesia, while UMs may form metabolite in

excess and therefore experience more pronounced analgesia and a greater likelihood of opioid-

related side effects (Zahari and Ismail, 2014).

Page 48: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

26

Figure 8: Structures and properties of opioid parent compounds and CYP2D6-mediated metabolites

Figure 8: Structures and properties of opioid parent compounds and CYP2D6-mediated

metabolites. Codeine, oxycodone, and hydrocodone (A) structures differ by defined functional

groups (R1 and R2), while tramadol (B) has a unique structure.

Codeine analgesia is solely mediated by the CYP2D6 formation of morphine and is therefore often

used as a prototypical example of the impact of CYP2D6 genotype on opioid response. In general,

CYP2D6 PMs produce little-to-no morphine from codeine, with extremely low plasma morphine

concentrations, and therefore little-to-no analgesia while UMs may experience more severe side-

effects, likely mediated by greater morphine concentrations (Kirchheiner et al, 2007). In healthy

volunteers given codeine, CYP2D6 PMs had 29-fold lower maximum plasma morphine

concentration than EMs and had no analgesic effect, tested as a response to selective

transcutaneous nerve stimulation (Desmeules et al, 1991). Similarly in another set of healthy

volunteers given codeine, CYP2D6 PMs had a 15-fold lower plasma morphine AUC and 11-fold

lower total excreted morphine than EMs, and reduced effects of codeine on gastrointestinal

motility (Mikus et al, 1997). Inhibiting CYP2D6 with quinidine in EMs resulted in undetectable

plasma morphine after codeine and no change in pin-prick pain thresholds compared to placebo,

thus inhibition abolished morphine formation and codeine effect (Sindrup et al, 1992).

Page 49: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

27

Tramadol is metabolized by CYP2D6 to a 300-fold more pharmacologically active metabolite O-

desmethyltramadol (Figure 8) (Abdel-Rahman et al, 2002). In patients receiving tramadol for post-

operative analgesia, the mean O-desmethyltramadol plasma concentrations were lower in

individuals with a CYP2D6 PM phenotype (0 ng*h/ml AUC) compared to UM (149.7 ng*h/ml),

and were lower in individuals co-medicated with CYP2D6 inhibitors (Stamer et al, 2007).

Consequently, more CYP2D6 PMs (81%) were categorized as tramadol non-responders compared

to patients with all other phenotypes (10-20%); PMs also required more tramadol analgesics

(calculated by total consumed mg of tramadol) over 48 h after surgery (Stamer et al, 2007). In a

Chinese population receiving tramadol post elective nephrectomy, individuals homozygous for the

reduced function CYP2D6*10 allele reported higher post-operative pain (VAS scores) and

required more tramadol than individuals with heterozygous and wild-type (*1/*1) genotypes

(Dong et al, 2015).

In some cases, although CYP2D6 metabolizes an opioid to a more pharmacologically active

metabolite, CYP2D6 genotype does not affect response. In post-operative patients receiving

oxycodone, plasma oxymorphone concentrations and the CYP2D6-metabolic ratio

(oxymorphone/oxycodone) differed across CYP2D6 phenotypes and were greater in CYP2D6

UMs than PMs; however pain scores did not change (Stamer et al, 2013). When CYP2D6 was

inhibited by quinidine in healthy CYP2D6 EMs, oxymorphone levels were undetectable in 80% of

the subjects however the subjective effects of oxycodone were unaltered (Heiskanen et al, 1998).

Similarly, inhibiting CYP2D6 in healthy subjects with paroxetine reduced the oxymorphone AUC

by 44%, and increased the CYP3A4-mediated metabolite noroxycodone AUC by 68%, but did not

alter subjective effects of oxycodone (Gronlund et al, 2010). The analgesic response to

hydrocodone, a 6-keto opioid with similar structure to oxycodone, appears to be similarly

unaffected by CYP2D6 phenotype (Yiannakopoulou, 2015). In a large prospective observational

study, individuals taking at least one CYP2D6 substrate or inhibitor experienced less hydrocodone

analgesia than those without (1.7-fold lower change in VAS score) (Monte et al, 2014). However,

a separate study showed that CYP2D6 EMs with active CYP2D6, EMs with inhibited CYP2D6

(via quinidine), and CYP2D6 PMs have no difference in subjective response to hydrocodone,

suggesting minimal role of CYP2D6 metabolism in hydrocodone analgesia (Kaplan et al, 1997).

Page 50: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

28

1.3.3 Impact of CYP2D6 phenotype on opioid abuse liability

There are numerous pharmacologic characteristics of opioid response that contribute to abuse

potential including, but not exclusive to, speed of onset of drug response, duration of response,

and net positive (e.g. analgesia and euphoria) and negative (e.g. adverse events) effects (Wightman

et al, 2012). Like analgesia, the subjective effects (e.g. euphoria and elation) of opioids are

mediated by action at MORs. Therefore, interindividual differences in CYP2D6 phenotype,

resulting in differences in the formation of metabolites that influence positive and negative

subjective effects, may be useful to predict opioid-specific abuse potential. In support of this,

CYP2D6 PMs were under-represented in individuals dependent on oral opioids (0% of 83

individuals), compared to those with multi-drug dependencies (4% of 93) and with no drug

dependencies (6.5% of 93) (Tyndale et al, 1997).

Codeine exhibits pronounced interindividual variation in analgesia, due to wide variation in

CYP2D6 phenotype resulting in altered formation of the active metabolite morphine (Desmeules

et al, 1991). Similarly, variation in this CYP2D6 activation alters codeine abuse potential. In

healthy non-drug-dependent CYP2D6 EMs inhibiting CYP2D6, with single and short-term

quinidine treatment, decreased morphine formation and positive subjective effects from a preferred

dose of codeine (Kathiramalainathan et al, 2000).

Assessing individual CYP2D6 phenotype may be able to predict oxycodone’s abuse potential,

despite the more active CYP2D6 metabolite of oxycodone, oxymorphone, not contributing

significantly to oxycodone analgesia. Predictive pharmacogenetic/pharmacokinetic models have

been used to predict oxycodone abuse potential from CYP2D6 phenotype (Linares et al, 2014).

However, analgesia models show no significant contribution of the CYP2D6 metabolite of

oxycodone, oxymorphone, to oxycodone analgesia, therefore the validity of these models is

unknown. In healthy CYP2D6 EMs and PMs, there were no significant differences in objective

(e.g. pupil dilation) and subjective (e.g. Addiction Research Center Inventory scale) effects elicited

by hydrocodone (Kaplan et al, 1997). Inhibiting CYP2D6 with quinidine abolished the

pharmacokinetic difference between EMs and PMs, determined by the parent (hydrocodone) to

metabolite (hydromorphone) ratio, but had no impact on hydrocodone subjective effects,

Page 51: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

29

suggesting CYP2D6 activity does not contribute to hydrocodone’s abuse potential (Kaplan et al,

1997).

1.3.4 Other sources of variability in opioid response

Beyond genetic variation in the CYP2D6 metabolism of opioids, there are numerous additional

sources of interindividual variation in opioid response, including variation in OR expression and

function, metabolism by enzymes other than CYP2D6, and extrahepatic metabolism of opioids.

Variants in OPRM1, the gene encoding the MOR, may influence opioid response in humans by

altering receptor function or substrate binding. The 802T>C OPRM1 SNP is located in an

intracellular protein domain and interferes with G-protein coupling and receptor desensitization,

resulting in diminished MOR potency of DAMGO, β-endorphin, and morphine (Lotsch et al,

2004). However, the 802C allele is rare across populations (allele frequency <1%) which has made

study of the functional outcome in humans challenging (Befort et al, 2001; Koch et al, 2000). In

comparison, the 118A>G OPRM1 SNP is located on an extracellular binding region, altering

substrate binding, and has relatively high frequencies (allele frequency of 10 – 50% across

ethnicities) (Mague and Blendy, 2010). Individuals heterozygous or homozygous for the 118G

OPRM1 variant experience reduced analgesic response to morphine and numerous other opioids

(Mague and Blendy, 2010).

CYP3A4 represents a major metabolic pathway for numerous opioids (Lotsch et al, 2004). Due to

a large number of drug compounds metabolized by CYP3A4, clinically significant drug-drug

interactions may occur between opioids and CYP3A4 substrates. The anti-viral medications

ritonavir and nevirapine induce the CYP3A4 metabolism of methadone, therefore decreasing its

pharmacologic effect; HIV-patients receiving ritonavir and nevirapine alongside methadone

treatment experience less methadone effect and therefore heightened opioid-withdrawal symptoms

(Geletko and Erickson, 2000; Heelon and Meade, 1999). Genetic polymorphisms in the human

CYP3A4 gene have been identified, however the clinical influence on opioid metabolism and

response is currently unknown (Lotsch et al, 2004).

While interindividual variation in opioid metabolism has primarily focused on hepatic CYP

metabolism altering circulating plasma opioid parent and/or metabolite levels, CYP-mediated drug

Page 52: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

30

metabolism in extra-hepatic tissues, such as the brain, has emerged as an novel source of variation

in drug response (see Appendix A) (McMillan and Tyndale, 2017a; Miksys and Tyndale, 2013).

As the brain is the primary site of opioid action, local metabolism of opioid compounds may impact

brain parent and metabolite levels and resulting therapeutic effects, independent of peripheral

metabolism and systemic drug levels. This novel source of interindividual variation in opioid

response is the primary focus of this thesis, and further discussed in the following introduction

section 1.4.

1.4 – Brain CYPs

1.4.1 Brain CYP expression

CYPs are expressed in the brains of numerous species, where they may contribute to the local

metabolism of exogenous and endogenous compounds, and to neuroprotection (Meyer et al, 2007).

The majority of the types of human CYP mRNA transcripts found in the liver have been found in

the brain, however only a small number of these isoforms have been studied at the level of protein

and/or activity (Dutheil et al, 2009; Miksys and Tyndale, 2013). This is due, in part, to uncertainty

as to whether an adequate environment exists (e.g. required cofactors and coenzymes) to allow

brain CYP metabolism in vivo, and to total brain CYP content being low relative to liver (1–2%)

(Hedlund et al, 2001; Miksys and Tyndale, 2013). The relative expression of CYPs in the brain,

compared to the liver, varies across isoforms. CYP46A1 transcripts are virtually absent in the liver

while highly expressed in the brain; CYP1B1 and CYP2U1 transcripts are expressed in the brain

at twice the level of liver, and CYP2J2 levels in the brain are 10% that of liver (Agundez et al,

2014). Other isoforms such as CYP1A1, CYP3A4, and CYP2E1, while detected in human brain,

are expressed at low transcript levels in normal conditions (Agundez et al, 2014). However, the

cell- and region-specific expression of CYPs may produce microenvironments in the brain that

may influence CNS-acting drug effects in those cells and/or regions (Britto and Wedlund, 1992).

Hepatic CYPs are generally expressed in the endoplasmic reticulum, while brain CYPs are found

on mitochondrial membranes, the plasma membrane, and other cell compartment membranes, and

display large variability in species and regional expression (Walther et al, 1986). Human CYP1A

mRNA and protein have been identified in the cortex, thalamus, and numerous brainstem nuclei

(e.g. substantia nigra, median raphe, and locus coeruleus) (Farin and Omiecinski, 1993; McFadyen

Page 53: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

31

et al, 1998). Rat CYP1A mRNA and protein exhibit similarly high expression in the cortex, but

also in the cerebellum, midbrain, and striatum (Schilter and Omiecinski, 1993). Human and rat

CYP2B mRNA and protein exhibit high expression in the cortex, striatum, hippocampus, basal

ganglia, and cerebellum, while rat brain CYP2B is further expressed in the olfactory bulbs and

hypothalamus (Farin and Omiecinski, 1993; McFadyen et al, 1998; Miksys et al, 2003; Schilter

and Omiecinski, 1993). Species differences in regional expression of brain CYPs is important to

consider when relating the functional outcome of brain CYP metabolism in animal models of

variable brain CYP metabolism to the potential impact in humans (further discussed in 1.4.3).

CYPs are also expressed at the BBB, an important endothelial barrier to compounds entering the

CNS, where CYPs may act in conjunction with transport proteins to provide protection for the

CNS (Agundez et al, 2014; Decleves et al, 2011). CYP1B1 and CYP2U1 mRNA in humans are

highly expressed in cerebral micro-vessels at the BBB, as well as CYP2E1 and CYP2D6 (Dauchy

et al, 2008). CYP2B may also function at the BBB interface as it is highly expressed in astrocytes

with end-feet that envelop cerebral blood vessels (Miksys and Tyndale, 2013). The precise

function of CYPs expressed at the BBB is currently unknown; however, their localization at this

important juncture of xenobiotic permeation to the CNS suggests a role for protection against the

entry of drugs or environmental toxins.

1.4.2 Brain CYP activity

After the discovery of brain CYP expression, it was unknown whether brain CYP enzymes were

functional in vivo, or able to influence the levels and resulting effect of exogenous or endogenous

compounds. Since parent and/or metabolite levels in the periphery, influenced by liver CYPs, can

often cross the BBB it is difficult to separate the relative contribution of brain and liver CYPs to

the levels of a specific metabolite in the brain (Miksys and Tyndale, 2013). Furthermore, in vitro

study of brain CYP membranes is difficult, as their activity is sensitive to storage; 80% of rat brain

CYP2D membrane activity is lost when stored in Tris buffer rather than artificial cerebrospinal

fluid while 40% of rat brain CYP2D membrane activity is lost when kept at -30°C for one week

(Tyndale et al, 1999). Nevertheless, in vitro studies in rat brain membranes confirmed the

metabolism of a variety of CYP-substrates, such as the CYP2D-mediated hydroxylation of the

Page 54: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

32

bufuralol or the CYP2B-mediated oxidation of parathion (Albores et al, 2001; Coleman et al,

2000).

Initial evidence of brain CYP activity in situ was investigated through using two mechanism-based

inhibitors of rat brain CYP2B. [3H]-8-methoxypsoralen (8MOP), a radiolabelled CYP2B

mechanism-based inhibitor, was injected into rat frontal cortex whereby, upon metabolism by

CYP2B enzyme to a radiolabelled metabolite, 8MOP would inhibit and label CYP2B (Koenigs

and Trager, 1998). CYP2B would only become radiolabelled if 8MOP were metabolized (i.e. if

CYP2B was enzymatically functional); a monoclonal antibody was used to immunoprecipitate

CYP2B and radiolabelled enzyme was detected (Miksys and Tyndale, 2009). The specificity of

mechanism-based inactivation of CYP2B by 8MOP was assessed by pre-treating the frontal cortex

unilaterally with another mechanism-based inhibitor of CYP2B, C8-Xanthate, prior to bilateral

8MOP treatment (Miksys and Tyndale, 2009; Yanev et al, 2000). The side of the frontal cortex

pre-treated with C8-Xanthate had a reduced amount of 8MOP-radiolabelled immunoprotein

compared to the contralateral side, providing further evidence that brain CYP2B was functional in

vivo (Miksys and Tyndale, 2009). Similar techniques have been used to demonstrate a functional

impact of brain CYP2B metabolism on CNS-acting CYP2B substrate drug response in vivo.

Inhibiting brain-specific CYP2B in rat with a single intracerebroventricular injection of C8-

Xanthate increased brain levels of the sedative-anesthetic propofol (inactivated by CYP2B) and

increased propofol drug effect, measured by sleep time (Khokhar and Tyndale, 2011). There was

no difference in ex vivo hepatic CYP2B metabolism between animals given intracerebroventricular

vehicle or inhibitors (C8-Xanthate or 8MOP), indicating no effect on brain-administered CYP2B

inhibitors on liver CYP2B activity (Khokhar and Tyndale, 2011). Evidence of the functional

impact of brain CYP metabolism on drug response in experimental animals has now been

demonstrated across different CYP isoforms (e.g. CYP2B and CYP2D) and CNS-acting drug

classes (e.g. antipsychotics and neurotoxins), through novel techniques of brain-specific CYP

inhibition and induction (more examples in 1.4.4.3 and 1.4.4.4) (Ferguson and Tyndale, 2011;

Miksys and Tyndale, 2013). While brain CYP metabolism may not influence drug

pharmacokinetics by way of peripheral metabolism and clearance, local metabolism in the brain

may influence drug levels at the target site of action and have significant consequences on drug

response.

Page 55: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

33

1.4.3 Regulation of brain CYP activity

Brain CYPs differ from their hepatic CYP counterparts in their sensitivity to xenobiotic inducers

in an isoform, brain-region, and mechanism specific manner (Miksys and Tyndale, 2013).

Phenobarbital, a potent inducer of numerous hepatic CYPs, increases the expression and activity

of brain and liver CYP2B1 and CYP2B2 in both rats and monkeys (Lee et al, 2006c; Schilter et

al, 2000). Phenobarbital also increases brain CYP2E1 protein levels in rats and monkeys, but liver

CYP2E1 protein and mRNA levels in rats but not monkeys (Caron et al, 2005a; Lee et al, 2006a).

Nicotine and ethanol independently, and together, increase brain and liver CYP2E1 in monkeys

and rats (Miksys and Tyndale, 2013). This is consistent with higher brain CYP2E1 levels observed

in alcoholics and smokers compared to non-alcoholics and non-smokers (Ferguson et al, 2013;

Howard et al, 2003; Joshi and Tyndale, 2006b; Warner and Gustafsson, 1994). However, nicotine

and ethanol may regulate brain CYP2E1 via different mechanisms; for example, ethanol treatment

induces rat brain CYP2E1 mRNA levels, suggesting a transcriptional mechanism of regulation,

while nicotine induction of rat brain CYP2E1 does not, suggesting increased translational

efficiency or protein stability (Howard et al, 2003; Joshi and Tyndale, 2006a; Zhong et al, 2012).

The species and isoform specific effects of inducers on brain CYP expression, together or

independent of an effect on liver CYP expression, is hallmark to brain CYP metabolism and

important for understanding the translation of in vivo studies on their functional significance.

Short term nicotine pretreatment (seven days, subcutaneously) induces rat brain CYP2B, with no

change in liver CYP2B protein and mRNA levels (Khokhar et al, 2010; Miksys et al, 2000a).

Similarly, monkey brain CYP2B6 protein is induced by nicotine with no increase in mRNA or

liver protein levels (Ferguson et al, 2013). Smokers have higher levels of CYP2B6 in the brain

compared to non-smokers, as assessed in post-mortem brain samples, while liver CYP2B6

expression does not differ (Hesse et al, 2004; Miksys et al, 2003). This suggests that similar to the

effects seen in rats and monkeys, nicotine, the primary psychoactive component in cigarettes, may

induce CYP2B in human brain but not liver.

Brain CYP expression is further regulated by endogenous factors including hormonal and other

factors associated with development (Miksys and Tyndale, 2013). Human liver CYP2D is

constitutively expressed across an adult individual’s lifetime; however, brain CYP2D6 levels

Page 56: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

34

(assessed in the frontal cortex) increase with age (Mann et al, 2012; Treluyer et al, 1991). Sex-

based differences may also exist in brain CYP expression; exogenously administered estrogen and

testosterone increases, while progesterone decreases, brain CYP2D expression and mRNA levels

in female rats (Baum and Strobel, 1997; Bergh and Strobel, 1996). Transcripts for nuclear

receptors have also been detected in the brain (e.g. peroxisome proliferator-activated receptor and

retinoid X receptor), that may regulate the expression of brain CYPs in a similar, or different,

manner to liver CYPs (Nishimura et al, 2004; Petersen et al, 2000). The overall extent of

endogenous regulation of brain CYP expression, and impact in vivo, is still relatively unknown

and poorly understood.

1.4.4 Brain CYP2D

1.4.4.1 Brain CYP2D expression and activity

CYP2D enzyme has been identified in the brains of numerous species, including rat, mouse, dog,

monkey, and human (Miksys et al, 2002; Miksys et al, 2005; Siegle et al, 2001; Tyndale et al,

1999). CYP2D6, in humans, is expressed in the brain at levels that are a fraction of liver; however,

is one of the most prominent drug metabolizing CYPs expressed in the brain (Dutheil et al, 2009).

Human CYP2D6 is expressed heterogeneously across most brain regions, such as in the cortex,

nucleus accumbens, hippocampus, and thalamus, with highest expression in the caudate, putamen,

cortex, and cerebellum (Miksys et al, 2002). Brain CYP2D6 expression is cell-specific; for

example, CYP2D6 immunoreactivity was detected in neurons of the substantia nigra, pyramidal

cells in the frontal cortex, Purkinje and glial cells in the cerebellum, and endothelial cells at the

BBB (Dutheil et al, 2009; Dutheil et al, 2010; Miksys et al, 2002). The region and cell specific

expression of brain CYP2D is relatively consistent across species, with marked similarities in

regions with highest brain CYP2D expression. Rat brain CYP2D is expressed heterogeneously

with highest levels in the cerebellum, frontal cortex, parietal cortex, pons, medulla, and spinal cord

(Miksys et al, 2000b). Similar to human brain CYP2D6, rat brain CYP2D showed strong

immunoreactivity in neurons of the substantia nigra, pyramidal neurons in the neocortex, Purkinje

cells in the cerebellum, and glial cells, but also in neurons in the olfactory bulb and pons, and

endothelial cells in the choroid plexus (Miksys et al, 2000b; Miksys et al, 2005; Norris et al, 1996).

Rat CYP2D4 and CYP2D18 are highly expressed in the brain; however, of the six rat CYP2D

Page 57: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

35

isozymes, the relative region- and cell-specific expression of each enzyme is unknown and difficult

to discern (Komori, 1993; Miksys et al, 2000b; Wyss et al, 1995).

The activity of brain CYP2D was first demonstrated in vitro by the ability of rat brain microsomes

to perform the same enzymatic conversions as human or rat liver CYP2D; for example, rat brain

membranes catalyzed the demethylation of codeine and dextromethorphan, the demethylenation

of 3,4-methylenedioxymethamphetamine, and the hydroxylation of bufuralol (Chen et al, 1990;

Coleman et al, 2000; Jolivalt et al, 1995; Lin et al, 1992). Different rat brain regions metabolized

dextromethorphan to varying degrees, with the highest catalytic activity observed in cerebellar,

olfactory, and hippocampal membranes, notably, regions of highest CYP2D mRNA and protein

expression (Miksys et al, 2000b; Tyndale et al, 1999). Brain membranes also biosynthesized the

monoaminergic neurotransmitters dopamine and serotonin in vitro (Hiroi et al, 1998b; Yu et al,

2003). Formation of dopamine from tyramine and serotonin from 5-methyltryptamine was

demonstrated in rat brain membranes, with highest formation observed, of regions tested, in the

cerebellum; this was inhibited in vitro by the CYP2D inhibitor quinine and anti-CYP2D4

antibodies (Bromek et al, 2010; Haduch et al, 2013). The activity of brain CYP2D in situ was later

tested by assessing CYP2D-mediated metabolite levels directly in the brain. When rats were given

an intraperitoneal dose of codeine and morphine that resulted in similar plasma morphine

concentrations at 30 min post-injection, ex vivo brain morphine levels were only detectable after

codeine, suggesting local formation from codeine by brain CYP2D (discussed further in 1.4.4.4)

(Chen et al, 1990).

Recently, the in vivo activity of brain CYP2D was demonstrated using a CYP2D mechanism-based

inhibitor propranolol, similar to the demonstration of brain CYP2B activity in vivo. Propranolol is

hydroxylated by CYP2D to a reactive metabolite that binds the active site of the enzyme, thereby

inactivating it (Narimatsu et al, 2001). Administering propranolol intracerebroventricularly to rats,

24 hours prior to testing, yielded decreased ex vivo oxidation of codeine to morphine by brain

membranes (Zhou et al, 2013). The formation of dopamine and serotonin by CYP2D in brain has

also been assessed in vivo, through in vivo brain microdialysis measurement of neurotransmitter

levels after exogenous administration of CYP2D-substrate precursors. Rats administered tyramine

and 5-methyltryptamine had higher brain levels of dopamine and serotonin, respectively,

Page 58: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

36

suggesting in vivo brain CYP2D formation of neurotransmitters (Bromek et al, 2011; Haduch et

al, 2015).

1.4.4.2 Brain CYP2D regulation

While hepatic CYP2D levels are primarily regulated by genetics and are considered constitutively

expressed, brain CYP2D activity can be altered by both genetics and by exposure to xenobiotic

inducers. Single doses of the solvent toluene and the antipsychotic clozapine given to rats increased

immunohistochemical staining of CYP2D4 in the olfactory bulbs, cerebellum, ventral tegmental

area, and substantia nigra (Hedlund et al, 1996). There were no changes in brain CYP2D mRNA

transcript levels suggesting the increase in CYP2D protein levels was due to a non-transcriptional

mechanism (Hedlund et al, 1996). Additionally, while toluene produced a robust increase in liver

CYP2D4 protein and mRNA levels, clozapine did not, indicating a unique brain-specific induction

of CYP2D (Hedlund et al, 1996). Antipsychotics and antidepressants influence brain and liver

CYP2D expression in drug-specific manners. For example, treatment for two-weeks with the

antipsychotic thioridazine, shown to inhibit rat liver CYP2D, increased rat CYP2D protein levels

in the substantia nigra and cerebellum (Daniel et al, 2005; Haduch et al, 2011). Single injections

of the antidepressants fluoxetine and imipramine decreased brain and liver CYP2D expression

(Haduch et al, 2011). However, after chronic (two-week) treatment, fluoxetine paradoxically

produced region-specific increases (cerebellum) and decreases (striatum and nucleus accumbens)

in brain CYP2D levels, while imipramine did not alter brain CYP2D (Haduch et al, 2011).

CYP2D6 levels are higher in the post-mortem brains of smokers and alcoholics, compared to non-

smokers and non-alcoholics, while liver CYP2D6 levels do not differ (Mann et al, 2008; Miksys

et al, 2002; Miksys and Tyndale, 2004). Consistent with this, short term nicotine pretreatment

(seven days, subcutaneously), compared to saline, increased rat brain CYP2D enzyme levels in the

cerebellum, hippocampus, and striatum (Figure 9) (Yue et al, 2008). The change in brain CYP2D

expression was maximal at 8 hours after the last nicotine administration, a time where there was

no detectable nicotine in plasma, and returned to baseline at 12 hr (Micu et al, 2003; Yue et al,

2008). There was no change in liver CYP2D expression, suggesting the induction occurred in brain

alone (Yue et al, 2008). Furthermore, while brain CYP2D protein was increased there was no

change in brain CYP2D mRNA levels, indicating the induction occurred by a non-transcriptional

Page 59: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

37

mechanism (Yue et al, 2008). The precise mechanism behind the induction of brain CYP2D by

nicotine is currently unknown, but may involve increased protein stability or decreased protein

degradation; fourteen-day nicotine treatment downregulates enzymes involved in the ubiquitin-

proteasome pathway of protein degradation in rat hypothalamus, suggesting nicotine may regulate

this degradation pathway for CYP2D (Kane et al, 2004).

Figure 9: Fold change in brain CYP2D protein in human smokers, compared to non-smokers, and short-term nicotine pretreated rats, compared to saline pretreated

Figure 9: Fold change in brain CYP2D protein in human smokers, compared to non-

smokers, and 7-day short-term nicotine pretreated rats, compared to saline pretreated.

*p<0.05, **p<0.01, and ***p<0.001 according to respective analyses. (A) FC, frontal cortex; TC,

temporal cortex; CG, cingulate gyrus; OC, occipital cortex; HC, hippocampus; EC, entorhinal

cortex; CD, caudate nucleus; PT, putamen; NA, nucleus accumbens; GP, globus pallidus; SN,

substantia nigra; Cv, cerebellar vermis; Ch, cerebellar hemisphere. (B) BS, brain stem; FC, frontal

cortex; HC, hippocampus; TH, thalamus; ST, striatum; CB, cerebellum. Adapted from Mann et al.

2008 and Yue et al. 2008.

Brain CYP2D may also undergo endogenous regulation. Liver CYP2D6 levels in humans increase

immediately after birth to adult levels and remain constant with age (Treluyer et al, 1991);

however, CYP2D6 enzyme levels assessed in frontal cortex increased with age (tested from fetal

to 80 years of age), an effect that may occur in other brain regions as well (Mann et al, 2012).

Brain CYP2D expression and mRNA levels in female rats were increased after short term (seven

days, subcutaneous) estrogen and testosterone treatment, and decreased after progesterone (Baum

Page 60: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

38

and Strobel, 1997; Bergh and Strobel, 1996). In contrast, short term testosterone treatment

decreased brain CYP2D expression and mRNA levels in orchiectomized male rats, with no effect

on liver CYP2D (Li et al, 2015a). In culture, testosterone increased levels of brain-specific

microRNA (miR-101 and miR-128-2) in human brain-derived cell lines (SH-SY5Y and U251),

while overexpression of microRNA decreased CYP2D mRNA levels in these cells (Li et al,

2015a). Sex hormone induced changes in these microRNA (short non-coding RNA segments

involved in silencing gene expression), may be one mechanism for the sex-specific regulation of

human brain CYP2D6.

1.4.4.3 Functional impact of variation in brain CYP2D

1.4.4.3.1 Neurotoxicity

A large number of centrally acting neurotoxins (e.g. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

[MPTP], tetrahydroisoquinoline, paraquat, and harmaline) are metabolized by CYP2D (Buratti et

al, 2003; Miksys and Tyndale, 2006). Brain CYP2D inactivation of neurotoxins to less neurotoxic

metabolites locally, in regions affected by neurotoxin action, may contribute to individual

susceptibility to neurotoxicity. Parkinson’s disease is a neurodegenerative disorder with both

familial and sporadic etiology; environmental exposure to neurotoxins likely contributes to

Parkinson’s disease (Elbaz et al, 2004; Vance et al, 2010). There is a well-established association

between genetic variation in CYP2D6 and susceptibility to Parkinson’s disease. In a case-control

study, CYP2D6 PMs were at a greater overall risk for developing Parkinson’s disease (Odds Ratio

= 1.41, 95% Confidence Interval [CI] = 0.81-2.44) which was increased with exposure to

pesticides (Odds Ratio = 1.55, 95% CI = 1.00-2.55) (Elbaz et al, 2004). Moreover, Parkinson’s

disease patients were found to have 40% lower CYP2D6 levels in the frontal cortex and cerebellum

than age-matched genotype-matched controls, suggesting that low brain CYP2D6 levels may be a

risk factor for Parkinson’s disease (Mann et al, 2012). The neurotoxin MPTP and its toxic

metabolite MPP+ are metabolized to inactive metabolites by CYP2D6; both are used in

experimental animal models of Parkinson’s disease (Mann and Tyndale, 2010). Overexpressing

CYP2D6 in rat adrenal medulla cells (PC12) protected against MPP+ neurotoxicity, while

inhibiting CYP2D6 (with quinidine, propranolol, metoprolol, or timolol) in human neuroblastoma

cells (SH-SY5Y) increased MPTP and MPP+ neurotoxicity (Mann and Tyndale, 2010; Matoh et

al, 2003). Interindividual variation in the activity or expression of CYP2D6, specifically in the

Page 61: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

39

brain, may contribute to differences in neurotoxin metabolic inactivation and subsequent

neurotoxicity, like that seen in Parkinson’s disease.

While CYP2D6 phenotypes have been used as predictors of neurotoxicity risk, variable brain

CYP2D activity through environmental exposure to xenobiotic inducers may be a novel source of

variation in individual susceptibility to neurotoxicity. Smokers are at a lower risk for Parkinson’s

disease, and nicotine has been found to be neuroprotective in several neurotoxin-induced animal

models of Parkinson’s disease (Alves et al, 2004; Quik et al, 2009; Quik et al, 2006). Short-term

nicotine treatment (two weeks via minipump) in mice protected against MPTP-induced damage in

the neostriatum and substantia nigra, two regions largely affected in Parkinson’s disease (Janson

et al, 1992). Mice exposed to cigarette smoke (for one week) were also protected against MPTP-

induced neuronal degeneration in the substantia nigra (Parain et al, 2003). Increased brain CYP2D

activity in animals given nicotine, consistent with increased brain CYP2D6 levels in smokers, may

contribute to protection against Parkinson’s disease by increased CYP2D metabolic inactivation

of neurotoxins (Miksys and Tyndale, 2006). Brain CYP2D levels are higher in alcoholics,

compared to non-alcoholics, and are increased in rats given a single dose of intraperitoneal ethanol

(Miksys et al, 2002; Warner and Gustafsson, 1994). Case-control studies have found that alcohol

consumption is weakly associated with protection against Parkinson’s disease; similar to with

smoking or nicotine exposure in rats, increased brain CYP2D levels observed in alcoholics may

play a neuroprotective role against Parkinson’s disease (Bettiol et al, 2015).

Evidence of a functional role for brain CYP2D metabolism in drug-induced neurotoxicity has

recently been demonstrated in vivo. Haloperidol is an antipsychotic drug and CYP2D substrate,

associated with extra-pyramidal side-effects; catalepsy, measured as the amount of time a rodent

spends immobilized with its paws on a raised surface, is used as a model of acute haloperidol

induced extra-pyramidal symptoms (Castagne et al, 2009; Creed and Nobrega, 2013). Inhibiting

CYP2D specifically in rat brain, by intracerebroventricular pretreatment with the mechanism-

based inhibitor propranolol, decreased catalepsy; inducing CYP2D specifically in rat brain with

short-term nicotine pretreatment increased catalepsy, an effect that was blocked by inhibiting brain

CYP2D (Figure 10) (Miksys et al, 2017). This suggested that brain CYP2D metabolized

haloperidol to a neurotoxic metabolite, responsible for producing catalepsy, and that reducing

Page 62: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

40

CYP2D activity in the brain via direct administration of an inhibitor was neuroprotective for acute

haloperidol induced toxicity. In support of this, haloperidol catalepsy correlated with brain, but

not liver, CYP2D activity (Miksys et al, 2017). There is some evidence that antipsychotic side-

effects are more prevalent in smokers (Wagner et al, 1988); increased brain CYP2D levels in

smokers may contribute to a greater risk for these acute extrapyramidal side-effects.

Figure 10: Modifying rat brain CYP2D activity alters catalepsy after acute haloperidol administration

Figure 10: Modifying rat brain CYP2D activity alters catalepsy after acute haloperidol

administration. Compared to vehicle (V), inhibiting brain CYP2D (with propranolol, PL)

decreased, inducing brain CYP2D (with nicotine, NIC) increased, and inhibiting the induced brain

CYP2D (NIC + PL) did not alter, catalepsy after acute haloperidol (A). Catalepsy correlated with

brain (B) but not liver (C) ex vivo CYP2D activity. *p<0.01 and **p<0.001 vs. vehicle and #p<0.01

and ##p<0.001 vs. inhibitor and inducer, using 2-way ANOVA. Relationships between data sets

were assessed by Spearman’s correlation coefficients. Adapted from Miksys et al. 2017.

While acute haloperidol induces catalepsy, chronic haloperidol exposure produces vacuous

chewing movements (VCM) in rats, a model of the chronic side effect tardive dyskinesia (Castagne

et al, 2009; Creed and Nobrega, 2013). Inhibiting brain specific CYP2D (by

intracerebroventricular propranolol pretreatment) during chronic haloperidol administration

increased vacuous chewing movements (Figure 11) (Miksys et al, 2017). This suggests that

haloperidol itself, or an alternative non-CYP2D metabolite, may be responsible for the chronic

side effect of tardive dyskinesia and this may be exacerbated by inhibiting brain CYP2D; VCMs

correlated negatively with brain, but not liver, CYP2D activity (Miksys et al, 2017). Elucidating

the role of brain CYP2D in haloperidol metabolism introduces important considerations for the

underlying neurochemistry of acute (catalepsy, likely mediated by a CYP2D-formed metabolite)

and chronic (VCMs, likely mediated by the parent drug haloperidol or a non-CYP2D-mediated

Page 63: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

41

metabolite) haloperidol-induced side effects and adds to the understanding of brain CYP2D in

neurotoxicity.

Figure 11: Modifying rat brain CYP2D activity alters vacuous chewing movements after chronic haloperidol administration

Figure 11: Modifying rat brain CYP2D activity alters vacuous chewing movements after

chronic haloperidol administration. Compared to vehicle (V), inhibiting brain CYP2D (with

propranolol, PL) increased VCMs during weeks 12–16 of chronic haloperidol (A). VCMs

negatively correlated with brain (B) but not liver (C) CYP2D activity. *p<0.01 using independent

samples t-test. Relationships between data sets were assessed by Pearson correlation coefficients.

Adapted from Miksys et al. 2017.

1.4.4.3.2 Endogenous neurochemicals

Speculation about a role for brain CYP2D in the metabolism of endogenous neurochemicals in

vivo was initially derived from associations between CYP2D6 phenotypes and personality (Penas-

Lledo and Llerena, 2014). Resting brain perfusion, measured through magnetic resonance imaging,

is an indicator of cerebral activity and may be associated with personality traits; CYP2D6 PMs,

compared to EMs, exhibit lower resting brain perfusion in the thalamus and hippocampus as well

as more anxious personality traits (Abler et al, 2008; Gonzalez et al, 2008; Kirchheiner et al,

2011). CYP2D6 genotype, ranked by activity score, was associated with cortical brain activation

in cognitive tests of working memory and emotional face matching, further suggesting a

physiological role for brain CYP2D6 in CNS function (Stingl et al, 2012). In vitro and in vivo

studies in experimental animals have shown a potential role for brain CYP2D in neurotransmitter

formation. Compared to wild type CYP2D6 enzyme expressed in recombinant bacteria, reduced

function allelic CYP2D6 variants had lower hydroxylation of progesterone and tyramine (Niwa et

al, 2004). Rat brain membranes formed dopamine from tyramine in vitro, a reaction reduced by

the CYP2D inhibitor quinine and anti-CYP2D4 antibodies (Bromek et al, 2010). Similarly, rat

Page 64: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

42

brain membranes, as well as cDNA-expressed rat CYP2D and human CYP2D6, formed serotonin

from exogenous 5-methyltryptamine, a reaction reduced by CYP2D inhibitors quinine and

fluoxetine (Haduch et al, 2013). Rat brain CYP2D formation of dopamine and serotonin in vivo

has been assessed using brain microdialysis (Bromek et al, 2011; Haduch et al, 2015). After

inhibiting the primary pathways of dopamine and serotonin production, dopamine and serotonin

levels were increased in vivo after exogenous administration of CYP2D-substrate precursors

(tyramine for dopamine, and 5-methyltryptamine and melatonin for serotonin) (Bromek et al,

2011; Haduch et al, 2015; Haduch et al, 2016). Humanized transgenic mice (with human CYP2D6

protein expressed in the liver and brain along with normal mice CYP2D protein) have higher basal

brain serotonin levels relative to wild type mice (Cheng et al, 2013). The absolute functional role

of brain CYP2D in neurotransmitter homeostasis in the normal physiologically functioning CNS

is currently unknown.

1.4.4.4 Brain CYP2D metabolism of opioids

There is a well-established influence of CYP2D6 phenotype on opioid metabolism and response

(discussed in introduction section 1.3). This is due, largely, to CYP2D6-mediated metabolites of

opioids generally displaying a much greater potency at MORs compared to the parent compounds

(e.g. CYP2D-formed metabolites morphine and oxymorphone have a 3000-fold and 44-fold

greater MOR affinity than the parent compounds codeine and oxycodone, respectively) (Lalovic

et al, 2006; Pert and Snyder, 1973). In general, CYP2D6 metabolism occurs in the liver, after

which opioid parent and/or metabolite compounds can cross the BBB to interact with MORs in

the CNS and elicit analgesia. However, the expression of CYP2D6 enzyme in human brain, and

demonstration of brain CYP2D activity in vivo using experimental animal models, suggests that

opioids may be metabolized locally at their target site of action in the CNS.

Initial evidence of potential brain CYP2D metabolism of opioids in situ was derived from

peripheral and central drug levels of codeine and its CYP2D metabolite morphine in rats (briefly

discussed in 1.4.4.1). Rats were given an intraperitoneal dose of codeine or morphine that produced

similar plasma morphine concentrations at 30 min post-injection, but with brain morphine levels

detectable only in animals given codeine (Chen et al, 1990). This suggested that the central

morphine levels at this time point after injection of codeine may not be due to peripheral morphine

Page 65: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

43

levels crossing the BBB, but instead to codeine metabolism within the brain (Chen et al, 1990).

Morphine is much less permeable across the BBB than codeine; using radiolabelled compounds

given intra-arterially, measured as a percentage compared to freely diffusible tritiated water, rat

brain morphine uptake after 15 seconds was negligible, while codeine uptake was 24% (Bouw et

al, 2000; Oldendorf et al, 1972). Similarly, using pharmacokinetic-pharmacodynamic modelling

of the BBB transport of oxycodone and its CYP2D metabolite oxymorphone in rats, oxymorphone

brain-to-plasma partitioning was 1.6-fold lower than oxycodone (Bostrom et al, 2006; Sadiq et al,

2013). Comparing CNS influx and efflux clearances, the net uptake of oxymorphone into the CNS

was 16.2-fold less than oxycodone (Bostrom et al, 2006; Sadiq et al, 2013). Oxycodone, like

codeine, appears to cross the BBB much more readily than oxymorphone, like morphine, and

therefore may also be impacted by brain CYP2D metabolism.

Since parent opioid compounds cross the BBB more rapidly compared to their peripherally formed

CYP2D metabolites, central CYP2D metabolism of opioids may contribute to the pharmacologic

effect of opioids. Suggestive of this, selectively inhibiting brain CYP2D by pretreatment with the

mechanism-based inhibitor propranolol, and separately with the competitive inhibitor

propafenone, decreased codeine analgesia over the first 60 min after subcutaneous codeine

administration (Zhou et al, 2013). The two brain CYP2D inhibitor pretreatments decreased

codeine metabolism in brain, but not liver (measured by morphine levels and morphine-to-codeine

ratios), and decreased ex vivo codeine metabolism in brain membranes, but not liver microsomes

(Zhou et al, 2013). Morphine analgesia was assessed as a control for the effect of brain CYP2D

inhibitor and/or inducer pretreatments on opioid-mediated analgesia; inhibiting brain CYP2D did

not alter analgesia across 120 min after subcutaneous morphine administration (Zhou et al, 2013).

This proof of concept study showed, for the first time, the contribution of brain CYP2D to opioid

metabolism and analgesia in vivo. However, it is unknown whether brain CYP2D can play a

functional role in opioid analgesia across a range of brain CYP2D activity (increased and

decreased), routes of opioid administration mirroring human use, multiple functional outcomes of

opioid response, and multiple opioid compounds with differing relative contributions of CYP2D-

formed metabolites to opioid analgesia. Investigation into the potential interindividual variability

in brain opioid metabolism and response may meaningfully contribute to knowledge of variation

in individual response to opioids clinically.

Page 66: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

44

1.5 – Statement of Research Hypotheses

In Chapter 1, “Nicotine increases codeine analgesia through the induction of brain CYP2D

and central activation of codeine to morphine”, we hypothesized that initial brain morphine

levels from codeine administration were due to brain CYP2D metabolism. We further

hypothesized that inducing brain CYP2D with seven days of nicotine pretreatment would

increase brain morphine levels, and analgesia, despite significant first pass metabolism from

intraperitoneal codeine administration, and that the impact of inducing brain CYP2D could be

reversed by inhibiting brain CYP2D. Due to our pretreatment paradigms being utilized to alter

brain CYP2D specifically, we hypothesized that inducing brain CYP2D would not alter plasma

morphine levels from codeine. We also hypothesized that inducing brain CYP2D with nicotine

would not alter baseline nociception or morphine-induced analgesia (not a CYP2D substrate).

In Chapter 2, “Inducing rat brain CYP2D with nicotine increases the rate of codeine

tolerance; predicting the rate of tolerance from acute analgesic response”, we hypothesized

that codeine tolerance would develop as a function of central morphine formation by brain

CYP2D; inducing rat brain, but not hepatic, CYP2D with seven days of nicotine pretreatment

would result in a greater proportional drop in codeine analgesia (greater rate of tolerance) during

repeated daily oral codeine exposure. We further hypothesized that inducing brain CYP2D with

nicotine would not alter the rate of morphine tolerance (not a CYP2D substrate) during repeated

daily oral morphine exposure. Since we hypothesized a proportional drop in codeine analgesia

across repeated administration, we speculated that the rate of tolerance may be a function of

acute analgesic response, with nicotine pretreatment altering the rate of codeine tolerance simply

by augmenting acute codeine analgesia by inducing the brain CYP2D activation of codeine.

In Chapter 3, “Rat brain CYP2D activity alters in vivo central oxycodone metabolism,

levels, and resulting analgesia”, we hypothesized that oxycodone, rather than the more potent

opioid-receptor activating CYP2D metabolite oxymorphone, was responsible for oxycodone

analgesia, and that increasing and decreasing brain CYP2D activity would decrease and increase,

respectively, central oxycodone levels, thereby oxycodone analgesia. We further hypothesized

that pretreatments used to modify brain CYP2D would not affect oxymorphone analgesia (not a

CYP2D substrate), baseline nociception, or peripheral oxycodone metabolism.

Page 67: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

45

2. Chapter 1: Nicotine increases codeine analgesia through the induction of brain CYP2D

and central activation of codeine to morphine

Douglas M. McMillan and Rachel F. Tyndale

Reprinted with permission from Nature Publishing Group: Neuropsychopharmacology

40(7): 1804-1812. © 2015 Springer Nature. All rights reserved.

Dr. Rachel F. Tyndale and Douglas M. McMillan designed the overall study. DMM conducted all

animal experiments, performed the data analysis, prepared plasma and brain samples for HPLC

detection, and wrote the manuscript. Fariba Baghai Wadji assisted in animal cannulation surgeries.

Dr. Bin Zhao performed all HPLC procedures for codeine and morphine quantification. RFT

guided the manuscript writing and edited the manuscript.

Page 68: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

46

2.1 Abstract

CYP2D metabolically activates codeine to morphine, which is required for codeine analgesia.

Permeability across the blood–brain barrier, and active efflux, suggests that initial morphine in the

brain after codeine is due to brain CYP2D metabolism. Human CYP2D is higher in the brains, but

not in the livers, of smokers and 7-day nicotine treatment induces rat brain, but not hepatic,

CYP2D. The role of nicotine-induced rat brain CYP2D in the central metabolic activation of

peripherally administered codeine and resulting analgesia was investigated. Rats received 7-day

nicotine (1 mg/kg subcutaneously) and/or a single propranolol (CYP2D mechanism-based

inhibitor; 20 μg intracerebroventricularly) pretreatment, and then were tested for analgesia and

drug levels following codeine (20 mg/kg intraperitoneally) or morphine (3.5 mg/kg

intraperitoneally), matched for peak analgesia. Nicotine increased codeine analgesia (1.59X

AUC0–30 min vs vehicle; p<0.001), while propranolol decreased analgesia (0.56X; p<0.05); co-

pretreatment was similar to vehicle controls (1.23X; p>0.1). Nicotine increased, while propranolol

decreased, brain, but not plasma, morphine levels, and analgesia correlated with brain (p<0.02),

but not plasma (p>0.4), morphine levels after codeine. Pretreatments did not alter baseline or

morphine analgesia. Here we show that brain CYP2D alters drug response despite the presence of

substantial first-pass metabolism of codeine and further that nicotine induction of brain CYP2D

increases codeine response in vivo. Thus, variation in brain CYP2D activity, due to genetics or

environment, may contribute to individual differences in response to centrally acting substrates.

Exposure to nicotine may increase central drug metabolism, not detected peripherally, contributing

to altered drug efficacy, onset time, and/or abuse liability.

Page 69: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

47

2.2 Introduction

The majority of cytochrome P450 (CYP)-mediated drug metabolism occurs in the liver; however,

resulting plasma drug and/or metabolite levels do not always correlate with therapeutic effect

(Ding and Kaminsky, 2003). Extra-hepatic CYP activity, specifically in the brain, may influence

target-tissue drug concentration and effect (Krishna and Klotz, 1994; Michels and Marzuk, 1993).

Experimentally, this has been explored, for example, by inhibiting rat brain CYP2B, which

decreased the inactivation of the sedative-hypnotic propofol, resulting in a prolonged propofol

effect and decreased the activation of the organophosphate chlorpyrifos reducing neurotoxicity

(Khokhar and Tyndale, 2011, 2012).

CYP2D6 (herein referred to as CYP2D for all species) metabolizes a large proportion of drugs

acting in the central nervous system (CNS) (e.g., amphetamine, haloperidol, and antidepressants),

as well as endogenous neurochemicals (Zanger et al, 2004). The human CYP2D gene is genetically

polymorphic, resulting in a wide range of metabolism phenotypes (Gaedigk et al, 2008), while

brain CYP2D levels are altered by common compounds and increase with age (Mann et al, 2012;

Miksys and Tyndale, 2004; Warner and Gustafsson, 1994; Yue et al, 2008). Rat and monkey brain,

but not liver, CYP2D is induced by nicotine pretreatment, consistent with higher levels of human

brain, but not liver, CYP2D in smokers (Miksys and Tyndale, 2004; Miller et al, 2014; Yue et al,

2008). Thus, the combination of genetics, environmental inducers, and age contribute to a wide

range of brain CYP2D activity, which may alter the metabolism and efficacy of CNS-acting

substrate drugs.

CYP2D metabolizes codeine to its primary analgesic metabolite morphine, a step required for

codeine analgesia (Adler et al, 1955; Chen et al, 1991b; Pert and Snyder, 1973; Sindrup et al,

1992; Sindrup et al, 1990). This occurs in the liver, after which morphine crosses the blood–brain

barrier (BBB) to interact with opioid receptors in the CNS and elicit analgesia. However, morphine

is less permeable across the BBB than codeine and is actively effluxed (Bouw et al, 2000;

Oldendorf et al, 1972). Coupled with this, brain concentrations of morphine after codeine

administration do not appear to be solely attributable to transfer of morphine across the BBB (Chen

et al, 1990; Zhou et al, 2013). Thus, following codeine administration, the initial morphine present

Page 70: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

48

in the brain, and initial analgesia1, may be due to brain CYP2D-mediated metabolism, rather than

hepatic metabolism.

Our objectives were to model the role of human brain CYP2D in response to centrally acting drugs,

through investigating rat brain CYP2D metabolic activation of codeine to morphine, in the

presence of hepatic first-pass metabolism, in a rat model of codeine analgesia. Rat brain CYP2D

has the ability to metabolize human CYP2D probe drugs and can be inhibited and induced in vivo,

independently of hepatic CYP2D, allowing the modelling of a wide range of CYP2D activity

within the brain, as postulated to exist in a heterogeneous population (Hiroi et al, 2002; Zhou et

al, 2013). Whether the nicotine induction of brain CYP2D is sufficient to alter drug response in

vivo, using a peripheral route of drug administration with substantial first-pass metabolism by the

liver, is unknown. This study aims to demonstrate that variable brain CYP2D alters substrate drug

metabolism and efficacy and provides further understanding to the functional role of CYP enzymes

in the brain.

2.3 Materials and Methods

Animals

Adult male Wistar rats (250–500 g; Charles River, St-Constant, QC, Canada) were housed in pairs

or triplets with ad libitum access to food and water. Rats were maintained under a 12-h artificial

light/dark cycle with experimentation occurring during the light cycle. Stress was reduced through

acclimation to experimenters and apparatuses. All procedures were approved by the Animal Care

Committee at the University of Toronto.

Drug Treatment

Codeine phosphate and morphine sulfate (PCCA, London, Canada) were dissolved in saline (0.9%

NaCl; pH 7) and distilled water, respectively, and injected intraperitoneally (i.p.). The doses of

codeine (20 mg/kg) and morphine (3.5 mg/kg) were chosen to match peak analgesia after testing

multiple doses of codeine (20, 25 and 30 mg/kg) and morphine (0.5, 1, 2, and 3.5 mg/kg).

Propranolol hydrochloride (Sigma-Aldrich, Oakville, Canada) was dissolved in artificial

1 “Initial analgesia”, as described throughout this chapter, is equivalent to “acute analgesia” as referred to in

chapters 1 and 3

Page 71: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

49

cerebrospinal fluid to deliver 20 μg base in 4 μl via intracerebroventricular (i.c.v.) injection 24 h

prior to codeine or morphine administration. Propranolol is a CYP2D mechanism-based inhibitor,

metabolized by CYP2D; the metabolite covalently binds and inactivates CYP2D (Narimatsu et al,

2001). Propafenone hydrochloride (Sigma-Aldrich), a CYP2D competitive inhibitor (Xu et al,

1995), was dissolved in a 20% (w/v) solution of 2-hydroxypropyl-β-cyclodextrin (Sigma-Aldrich)

in water to deliver 40 μg base in 4 μl i.c.v., 5 min prior to codeine or morphine administration. Two

mechanistically and structurally distinct CYP2D inhibitors were used to reduce the possibility of

off-target effects other than inhibition of codeine metabolism. Nicotine bitartrate (Sigma-Aldrich)

was dissolved in saline and given subcutaneously (s.c.) at 1 mg base/kg daily for 7 days, which

results in elevated rat brain, but not liver, CYP2D levels as previously described (Yue et al, 2008).

Cannulation Surgeries for Multiple i.c.v. Injections

Rats utilized for brain CYP2D-inhibition experiments were surgically implanted with i.c.v.

cannulas into their right lateral ventricle (Zhou et al, 2013). Screw anchors (BASi, West Lafayette,

USA) were implanted into the skull to serve as an anchor to the DuraLay inlay pattern resin

(Reliance Dental Mfg. Co., Worth, USA) used as an adhesive for the implanted cannula.

Nociceptive Testing of Codeine and Morphine Analgesia

Analgesia was measured by the tail-flick reflex test through the use of a tail-flick meter (Columbus

Instruments, Columbus, OH, USA) (D'Amour and Smith, 1941). A thermal stimulus is applied to

the rat's tail, −5 cm from the distal end, and the time of onset of stimulus to a vigorous tail flick is

recorded as the tail-flick latency (TFL). Prolongation of the TFL is an indication of both spinally

and supra-spinally mediated analgesia (Le Bars et al, 2001). The thermal strength of the tail-flick

meter was adjusted to obtain baseline TFLs around 3 s; a cutoff of 10 s was used to avoid damage.

TFLs were measured twice in each rat 24 h prior to experimentation, and the mean was used as the

individual rat's baseline TFL. Analgesia was expressed as a percentage of the maximal possible

effect (%MPE) (Dewey and Harris, 1971).

Page 72: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

50

Analgesia Experimental Design

For pretreatment experiments, codeine or morphine was given 24 h after i.c.v. pretreatment

injection (propranolol or vehicle) and/or 8 h after the last s.c. pretreatment injection (nicotine or

saline). After opioid administration, analgesia was assessed by TFL every 5 min for 30 min and

then every 10 min for the next 30 min. Within-animal designs were used where possible in which,

after a 1-week washout period, rats were crossed over to a comparator pretreatment group and

retested or killed for pharmacokinetic analysis.

Plasma and Brain Sample Preparation

For analysis of codeine and morphine from the plasma and brain, rats were pretreated, then

administered codeine, tested for TFL at 15 min postinjection, and killed by decapitation. Trunk

blood was centrifuged at 5000 g for 10 min. Half-brains were homogenized in 1: 3 (w/v) 0.01 N

HCl and centrifuged at 5000 g for 10 min. Nicotine plasma samples were obtained on the seventh

day of nicotine pretreatment via saphenous-vein blood draws.

HPLC Measurement of Plasma and Brain Drug/Metabolite Levels

Plasma and brain homogenate samples were analyzed as previously described (Zhou et al, 2013).

In brief, 100 μl of solid phase extract was analyzed by HPLC with ultraviolet detection at 214 nm

(Agilent 1200 Separation Module, Palo Alto, CA, USA). Morphine, codeine, and internal standard

(1 μg 2-benzoxazolinone) were separated on an Agilent ZORBAX SB-C18 Column (250 mm ×

4.6 mm I.D.; particle size, 5 μm), with the retention times of 4.1, 9.0 and 16.7 min, respectively.

The mobile phase used was methanol: phosphate buffer (29.3: 70.7 (v/v); pH 5.8) with a flow rate

of 1 ml/min. The limits of quantification were 5 and 10 ng/ml (5 and 50 ng/g for brain tissue) for

morphine and codeine, with an extraction efficiency of 76.9 and 90.1%, respectively. Nicotine and

cotinine were quantified by HPLC (Siu et al, 2006).

Statistical Analyses

Data were analyzed through one-way or two-way repeated-measures ANOVA with Bonferroni

corrections or unpaired (between animal) and paired (within animal) one-tailed t-tests, as

indicated.

Page 73: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

51

2.4 Results

Analgesic Profiles and Morphine Concentrations Suggest Involvement of Brain CYP2D in

Codeine Metabolism and Initial Codeine Analgesia

Codeine (20 mg/kg) resulted in a peak of analgesia around 15 min (n=22; Figure 12A) with

morphine metabolite levels detectable in both plasma and brain (n=5; Figure 12B and 12C). This

codeine dose used throughout provided analgesia below maximal %MPE, allowing for

measurement of both increases and decreases in analgesia. Morphine (3.5 mg/kg) matched the peak

analgesia level obtained from codeine, however, later at 30 min (n=8; Figure 12A). At 15 min, the

morphine dose produced similar plasma morphine levels to those from codeine

(p>0.3; n=10; Figure 12B), but lower brain morphine levels (p<0.02; Figure 12C), consistent with

lower analgesia at this time point. This suggests that the early analgesia peak at 15 min following

codeine, along with the high levels of brain morphine present, was due to morphine formed by

central (brain) metabolism. The delayed peak in analgesia after morphine suggests that peripheral

morphine, as compared with the more lipophilic codeine, crosses the BBB more slowly, therefore

eliciting analgesia at a later time.

Page 74: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

52

Figure 12: The analgesic time course following codeine and morphine suggests peripheral morphine levels do not predict central morphine levels or analgesia

Figure 12: The analgesic time course following codeine and morphine suggests peripheral

morphine levels do not predict central morphine levels or analgesia. Codeine (20 mg/kg)

produced an analgesic peak around 15 min postinjection (TMAX=13+1.8 min, mean+SEM; n=22;

a). A morphine dose (3.5 mg/kg), chosen to match peak analgesia from codeine, resulted in an

equivalent albeit later analgesia peak around 30 min postinjection (TMAX=27+4.9 min; n=8). At

15 min postinjection, the time of the codeine analgesia peak, the morphine dose (n=10) produced

similar plasma morphine levels to the codeine dose (n=5; b), and lower brain morphine levels (c)

consistent with the lower analgesia. A between-animal study design was used; *p<0.05 compared

with codeine (20 mg/kg) using an unpaired t-test.

Pretreatment with Two Distinct CYP2D Inhibitors (i.c.v.) Decreased the First 30 min of

Codeine-Induced Analgesia but Did Not Affect Morphine-Induced Analgesia

With a focus on the time frame (0–30 min postinjection) hypothesized to be associated with brain

metabolism of codeine to morphine and the resulting peak in codeine-induced analgesia, the effect

of inhibiting brain CYP2D was assessed using two structurally and mechanistically distinct

CYP2D inhibitors. Compared with vehicle pretreatment, 20 μg propranolol (mechanism-based

inhibitor) pretreatment resulted in significantly lower %MPE at numerous time points

(n=16/group; Figure 13A) after codeine (20 mg/kg) administration, producing a significant

reduction of the analgesic AUC0–30 min (p<0.01; Figure 13B). Likewise, 40 μg propafenone

Page 75: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

53

(competitive inhibitor) pretreatment resulted in significantly lower %MPE (n=6/group; Figure

13D, and analgesic AUC0–30 min after codeine administration (p<0.05; Figure 13E).

Figure 13: Inhibiting brain CYP2D reduced the first 30 min of codeine-induced analgesia but had no effect on morphine-induced analgesia

Figure 13: Inhibiting brain CYP2D reduced the first 30 min of codeine-induced analgesia

but had no effect on morphine-induced analgesia. Compared with vehicle pretreatment,

propranolol pretreatment (darker bar) resulted in significantly lower analgesia (%MPE; a) and area

under the analgesia–time curve (AUC; b) for 0–30 min after codeine administration (n=16/group).

Propranolol pretreatment (darker bars) did not result in a difference in analgesic AUC0–30 min or

AUC30–60 min (c) after morphine administration (n=5/group; hatched bars). Compared with vehicle

pretreatment, propafenone pretreatment (darker bar) resulted in significantly lower %MPE (d) and

analgesic AUC0–30 min (e) after codeine administration (n=6/group). Propafenone pretreatment

(darker bars) did not result in a difference in analgesic AUC0–30 min or AUC30–60 min (f) after

morphine administration (n=5/group; hatched bars). Error bars indicate SEM. A within-animal

study design was used; for analgesia–time curves, *p<0.05, **p<0.01, and ***p<0.001 using

repeated-measures ANOVA with Bonferroni post hoc. For AUCs, *p<0.05, **p<0.01 using

paired t-tests.

Page 76: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

54

As morphine is the active analgesic metabolite of codeine and not further metabolized by CYP2D,

its analgesia should not be altered by changes in CYP2D activity. Compared with vehicle

pretreatment, neither propranolol nor propafenone pretreatment altered %MPE (p>0.1 at all time

points) or analgesic AUC (p>0.1) across any time frame after morphine administration (n=5/group;

illustrated for AUC0–30 min and AUC30–60 min in Figure 13C and 13F).

Pretreatment with CYP2D inhibitors also had no effect on baseline nociception; there was no

difference between TFL prior to (3.52+0.21, mean+SEM) and 24 h after vehicle (3.64+0.16 s) or

(3.72+0.20 s) propranolol pretreatment (n=16; p>0.2), or TFL prior to (3.03+0.14 s) and 5 min

after vehicle (3.13+0.14 s) or (3.18+0.16 s) propafenone pretreatment (n=6; p>0.2). Together, this

indicated that CYP2D inhibitor pretreatments had no effect on analgesia that was unrelated to their

effects on codeine metabolism.

7-Day Pretreatment with Nicotine, to Induce Brain CYP2D, Increased the First 30 min of

Codeine-Induced Analgesia but Did Not Affect Morphine-Induced Analgesia

Nicotine administration induces rat brain CYP2D without altering hepatic CYP2D; rats were

administered nicotine (1 mg/kg; s.c.) or vehicle (saline, 1 ml/kg; s.c.) and tested at 8 h after the

seventh and final injection (Figure 14A), when brain CYP2D is significantly induced (Yue et al,

2008). On the final day of nicotine pretreatment, plasma nicotine levels peaked within the first

30 min and were undetectable at 8 h postinjection, the time of codeine or morphine administration

and analgesia testing (n=16; Figure 14B). Compared with vehicle pretreatment, 7 days of nicotine

pretreatment resulted in significantly higher %MPE (n=20/group; Figure 14A) after codeine

administration, producing a significantly higher AUC0–30 min (p<0.03; Figure 14B).

The effect of nicotine pretreatment on morphine analgesia (3.5 mg/kg) was also tested; if nicotine's

actions on increasing codeine-induced analgesia were via induced brain CYP2D, no change in

morphine-induced analgesia would be expected. Compared with vehicle pretreatment, nicotine

pretreatment did not result in significantly altered %MPE (p>0.3 at all time points) or analgesic

AUC (p>0.3) across any time frame after morphine administration (n=16/group; illustrated for

AUC0–30 min and AUC30–60 min in Figure 14E).

Page 77: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

55

Figure 14: Nicotine-mediated induction of brain CYP2D increased codeine analgesia and had no effect on morphine-induced analgesia

Figure 14: Nicotine-mediated induction of brain CYP2D increased codeine analgesia and

had no effect on morphine-induced analgesia. Rats were administered nicotine (1 mg/kg; s.c.)

or saline vehicle (s.c.) once daily for 7 days as outlined (a). As measured on the final day of nicotine

pretreatment, plasma nicotine levels peaked within the first 30 min and were undetectable by 8 h,

the time of codeine or morphine administration (n=16; b). Mean plasma nicotine levels averaged

over 8 h was approximately 70 ng/ml and over 24 h was approximately 25 ng/ml. Compared with

vehicle pretreatment, nicotine pretreatment (darker bar) resulted in significantly higher %MPE (c)

and analgesic AUC0–30 min (d) after codeine administration (n=20/group). Seven days of nicotine

pretreatment (darker bars) did not result in a difference in analgesic AUC0–30 min or AUC30–60 min (e)

after a morphine injection (n=16/group; hatched bars). Error bars indicate SEM. A within-animal

study design was used; for analgesia–time curves, **p<0.01 using repeated-measures ANOVA

with Bonferroni post hoc. For AUCs, *p<0.05 using paired t-tests.

The effect of 7-day nicotine pretreatment on codeine and morphine analgesia from the tail-flick

assay was also tested using the hot-plate assay (Eddy and Leimbach, 1953; Rowland et al, 1994).

Compared with vehicle, nicotine pretreatment resulted in significantly higher %MPE

(Supplementary, Figure 17A) and a 2.46-fold greater AUC0-30 min (p<0.01; n=8/group;

Supplementary, Figure 17B) after codeine administration, with no change in %MPE (p>0.2 at all

Page 78: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

56

time points) or AUC (p>0.3) across any time frame after morphine administration

(n=5/group; Supplementary, Figure 17C).

TFLs were assessed prior to daily nicotine injections and at 8 h postinjection (the time at which

codeine and morphine were tested). There was no change in preinjection TFL (Day 1: 5.4+0.5 s,

mean+SEM; Day 7: 5.3+0.4 s; p=0.39) or 8 h postinjection TFL (Day 1: 5.1+0.5 s; Day 7:

4.7+0.3 s; p=0.20) and no change in TFL between preinjection and 8 h postinjection (p>0.1) across

each of the 7 days of nicotine injections. Together, with the lack of effect on morphine analgesia,

this suggests that nicotine (as tested here) did not alter nociception during codeine and morphine

testing, suggesting, instead, a direct effect on codeine analgesia via induction of brain CYP2D.

Pretreatment with a Mechanism-Based Brain CYP2D Inhibitor Blocked the Effect of

Nicotine Induction of Brain CYP2D on Codeine Analgesia

Compared with vehicle pretreatment, when animals were pretreated with 7 days of nicotine, there

was a significant increase in %MPE (n=12/group; Figure 15A) and resulting increase in analgesic

AUC0–30 min after codeine (p<0.001; Figure 15B). Conversely, when animals were pretreated with

propranolol, there was a significant decrease in %MPE and analgesic AUC0-30 min after codeine

administration (p<0.05). Co-pretreatment of propranolol blocked the nicotine-induced increase in

codeine analgesia, likely through inhibiting the induced levels of brain CYP2D, and resulted in

similar codeine to the vehicle pretreatment (p=0.8). There was no significant difference in baseline

TFL between the four pretreatment groups (p>0.3). Thus, the effect of propranolol and nicotine

pretreatment together resulted in no net change in codeine-induced analgesia, suggesting that they

were acting in opposing directions via the same mechanism, presumably through modification of

brain CYP2D activity.

Page 79: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

57

Figure 15: Nicotine induction of brain CYP2D increased the first 30 min of codeine-induced analgesia, an effect that was blocked by inhibiting brain CYP2D

Figure 15: Nicotine induction of brain CYP2D increased the first 30 min of codeine-induced

analgesia, an effect that was blocked by inhibiting brain CYP2D. Compared with vehicle

pretreatment, 7 days of nicotine pretreatment or propranolol pretreatment alone resulted in

significantly higher and lower, respectively, %MPE (a) and analgesic AUC0–30 min (b) after codeine

administration. Compared with vehicle pretreatment, co-pretreatment of nicotine and propranolol

together resulted in a significantly different %MPE and AUC0–30 min than nicotine or propranolol

pretreatment alone and no change in %MPE or AUC0–30 min from vehicle pretreatment

(n=12/group). Error bars indicate SEM. A within-animal study design was used; *p<0.05,

**p<0.01, ***p<0.001, compared with vehicle, and #p<0.05, ##p<0.01, compared with the

combination pretreatment, using repeated-measures ANOVA with a Bonferroni post hoc test.

Variation in Brain CYP2D Altered Brain Metabolism of Codeine Without Affecting Hepatic

CYP2D

To investigate whether the alterations in codeine analgesia correlated with changes in codeine

metabolism in vivo, brain and plasma codeine and morphine levels were assessed after

administration of the combination of pretreatments used previously (Figure 15). There was a

significant correlation between plasma and brain codeine levels (r=0.60; p<0.001; Figure 16A) but

not between morphine levels (r=0.14; p=0.26; Figure 16B) at 15 min after codeine administration

(n=26). This suggests that at 15 min after codeine administration plasma morphine levels did not

predict brain morphine levels, indicating the presence of barriers to morphine permeability across

the BBB. In the same analysis, analgesia at 15 min correlated with brain morphine levels

(r=0.41; p<0.02; Figure 16C) but not with plasma morphine (r=0.01; p=0.48; Figure 16D) at

15 min after codeine administration (n=26), consistent with brain CYP2D-mediated codeine

metabolism and resulting brain morphine levels, being responsible for codeine analgesia over the

first 30 min after codeine administration.

Page 80: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

58

Figure 16: Codeine, but not morphine, levels correlate between plasma and brain; brain morphine levels reflect codeine analgesia

Figure 16: Codeine, but not morphine, levels correlate between plasma and brain; brain

morphine levels reflect codeine analgesia. Plasma and brain codeine levels correlated

significantly (a) at 15 min, suggesting that codeine equilibrated quickly between the plasma and

brain compartments. There was no correlation between plasma and brain morphine levels

following codeine administration (b). Analgesia correlated with brain morphine levels (c) but not

with plasma morphine (d) at 15 min after codeine administration (n=26). Pearson correlations were

used.

Compared with vehicle pretreated rats (48.5+2.0 ng/g, mean+SEM), nicotine pretreated rats had

significantly higher morphine levels in the brain (54.7+2.4 ng/g; p<0.02), propranolol pretreated

trended towards lower morphine levels in the brain (44.5+2.6 ng/g; p=0.2), and rats co-pretreated

with nicotine and propranolol had no difference in the brain morphine levels

(51.6+2.2 ng/g; p=0.8) after codeine administration, all consistent with brain CYP2D-specific

changes (n=8/group). The three active pretreatment paradigms did not alter plasma morphine

levels (p>0.7, for all comparisons), suggesting that these pretreatments modified brain metabolism

of codeine, without affecting hepatic metabolism.

Page 81: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

59

2.5 Discussion

This is the first study to demonstrate that brain CYP2D has a role in drug response even in the

presence of substantial first-pass metabolism of codeine and further that nicotine induction of brain

CYP2D can increase central drug metabolism and response in vivo. Previously, it was unknown

whether the nicotine-induced levels of brain CYP2D were sufficient and possessed adequate

cofactors in situ to alter CNS-acting drug metabolism and resulting drug effect in vivo. This is also

the first study to show that this nicotine induction of brain CYP2D alters drug response and can be

blocked with brain CYP2D inhibitors, suggesting the importance of the interplay of compounds

that alter brain activity on centrally acting drug effect, including for orally administered drugs.

Codeine and morphine, given at doses resulting in equivalent plasma morphine levels at 15 min

postinjection and peak analgesia, resulted in significantly different brain morphine levels and

analgesia time lines, suggesting that hepatic metabolism of codeine to morphine was not solely

responsible for the levels of brain morphine and centrally mediated analgesia after a codeine

injection. Our findings suggest a significant role for variable brain metabolism in codeine

analgesia; inhibiting brain CYP2D with two mechanistically distinct inhibitors decreased, while

inducing brain CYP2D with nicotine increased, codeine analgesia and resulting brain morphine

levels, effects that were selectively blocked by co-pretreatment. Furthermore, neither inhibiting

nor inducing brain CYP2D altered baseline response, morphine, analgesia, or peripheral

metabolism of codeine, suggesting that the pretreatments used to modify brain CYP2D did not

have off-target effects on codeine's downstream mechanism of action.

The 7-day nicotine pretreatment paradigm resulted in average daily nicotine plasma levels of

25 ng/ml (Figure 14B), similar to those seen in human smokers (20–50 ng/ml) (Benowitz, 1999);

similar to nicotine-treated animals, smoking is associated with higher human brain CYP2D (Mann

et al, 2008; Miksys and Tyndale, 2004; Miller et al, 2014; Yue et al, 2008). Peripheral

pharmacokinetics of codeine are similar in smokers and non-smokers, consistent with our current

animal studies, and with a lack of effect of smoking or nicotine treatment on hepatic CYP2D

(Miller, 1990). Approximately 20% of the US population continues to smoke cigarettes; smokers

have a greater risk of multi-drug use and dependency (Berg et al, 2013). Furthermore, the use of

electronic cigarettes that deliver nicotine mimicking conventional cigarettes is increasing,

Page 82: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

60

particularly in adolescents (Dutra and Glantz, 2014). Opioid analgesics such as codeine, while

widely accepted for the treatment of acute to chronic pain, are abused for their euphoric mood-

altering properties, with the incidence of non-medicinal prescription opioid use increasing (Fischer

et al, 2006). The reinforcing qualities of codeine come from its morphine metabolite, therefore for

individuals with higher levels of brain CYP2D (e.g. smokers) a more rapid exposure of the brain

to morphine, through faster centrally mediated metabolic activation, could speed the onset time of

codeine's reinforcing effects and increase its abuse liability (Kathiramalainathan et al, 2000;

Tyndale et al, 1997). Increased central opiate activation may be one mechanism increasing the risk

of smoking and opioid co-dependency (Richter and Ahluwalia, 2000). In support of this, the

prevalence of repeated prescription frequency, and general non-medicinal prescription opioid use,

is higher in both men and women with smoking dependence (Skurtveit et al, 2010). Although

alternative mechanistic explanations exist, such as receptor cross-sensitization in the enhanced

rewarding and psychomotor effects of morphine in rodents following nicotine treatment, nicotine's

ability to alter drug metabolism within the brain may also contribute (Vihavainen et al, 2008).

In animals pretreated with chronic nicotine, there was a trend for a faster onset of analgesia;

the TMAX with brain CYP2D induction (14.6+1.7 min, mean+SEM) was shorter compared with

vehicle pretreatment (18.8+1.4 min; p>0.1), brain CYP2D inhibition (17.1+1.8 min; p>0.4) and

co-pretreatment (17.1+1.8 min; n=12/group; p>0.5 using a two-way ANOVA against brain

CYP2D induction). A shift in codeine analgesic TMAX, due to enhancement of brain CYP2D

metabolism, could contribute to a faster onset of codeine's clinical actions but may also increase

its abuse liability. Individuals exposed to compounds that affect brain CYP2D levels and/or

activity could present with varied profiles of both clinical effect and abuse potential to drugs that

are activated (i.e., codeine), or inactivated (i.e., amphetamine) by CYP2D (Wu et al, 1997).

The absolute magnitude of the change in morphine levels, analgesia and TMAX was likely blunted,

due to the brain CYP2D inhibition paradigm employed, resulting from incomplete inhibition of

CYP2D. A larger dose of propranolol (40 μg base in 4 μl), which reduced analgesia further,

inhibited hepatic CYP2D (determined by ex vivo metabolic assays) suggesting that propranolol

had crossed the BBB into the periphery and obscured the ability to focus solely on brain

metabolism (Zhou et al, 2013). An active search is underway for alternative molecular-inhibition

Page 83: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

61

options for future experiments. In addition, brain CYPs are highly localized and respond to

inducers and inhibitors with complex patterns of regulation that are both tissue and cell-type

specific (Yue et al, 2008). For this reason, the direct correlation between brain regions associated

with antinociception and with CYP2D protein affected by induction and inhibition is unclear.

Thus, global brain inhibition and homogenates were used as a way of capturing a more general

picture, however, likely resulted in a muting of the propranolol effect. Further studies into brain

regions corresponding to high opioid receptor and CYP2D protein density are warranted to

elucidate the importance of region-specific brain CYP2D alterations on codeine metabolism and

analgesia.

Nicotine has an important role in modulating pain transmission. Activation of the α4β2 and α7

nicotinic acetylcholine receptor subtypes elicits analgesia and is purported to involve opioidergic

neurons and endogenous opioid peptides; nicotine analgesia is decreased in μ-opioid receptor

knockout mice (Berrendero et al, 2002; Carstens et al, 2001; Damaj et al, 2000; Dhatt et al, 1995).

Furthermore, nicotine dependence and withdrawal can affect both opioid- and noxious stimulus-

induced analgesia involving these receptors in the nucleus accumbens (Schmidt et al, 2001). For

these reasons, it was important to control for nicotine's own potential effects on nociception and

opioid action. There were no detectable levels of nicotine at 8 h postinjection, the time of

codeine/morphine administration (Figure 14B). Furthermore, there was no change in baseline

nociception between prenicotine and 8 h postnicotine administration, consistent with the absence

of nicotine or nicotine-induced nociception at this time point. Chronic nicotine administration also

did not affect morphine analgesia (Figure 14E), indicating a lack of direct effect of nicotine on

opioid-induced analgesia per se, including on the BBB permeability of morphine. Together, this

substantially reduces the likelihood of nicotine pretreatment effects on general nociception,

nicotine-mediated analgesia, or opioid-mediated analgesia in this chronic administration

paradigm, suggesting instead an effect of nicotine on brain CYP2D levels.

Overall, these results suggest a role of brain CYP2D on codeine metabolism and analgesia.

Following codeine administration, analgesia is initially mediated by morphine formed centrally by

brain CYP2D, due to the delay in hepatically formed morphine entering the CNS. Thus lower (or

higher) brain CYP2D activity and the consequent decrease (or increase) in brain morphine levels

Page 84: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

62

results in lesser (or greater) analgesia during this initial 30 min after codeine injection. Over time,

morphine crosses into the brain, suggesting that analgesia at this later time period is mediated by

morphine formed by hepatic CYP2D crossing into the CNS (seen from the shift in analgesic peak

from morphine (3.5 mg/kg) compared with codeine (20 mg/kg)). The clinical implications of our

findings are that individuals with higher brain CYP2D activity, such as genetically

ultrarapid CYP2D metabolizers or smokers, may experience a quicker and greater onset of

analgesia (and potentially abuse liability) from codeine, whereas those with lower brain CYP2D

activity, such as non-smokers or those taking CYP2D inhibitors, may experience a delayed and

lower analgesia. Accordingly, our results suggest that a translational study assessing the time

course and peak of codeine's analgesic effects in smokers vs non-smokers, or within subject

prenicotine/postnicotine treatment, would be warranted. Furthermore, it is suggested that drug

metabolism within the brain may cause interindividual differences in drug response, which are not

reflected in plasma drug levels.

2.6 Acknowledgements

This work was supported by the Endowed Chair in Addiction for the Department of Psychiatry (to

RFT), Canadian Institutes of Health Research (TMD 132557, MOP 97751, and MOP 136937),

Centre for Addiction and Mental Health and the CAMH Foundation, the Canada Foundation for

Innovation (grant numbers 20289 and 16014), and the Ontario Ministry of Research and

Innovation. Dr. Rachel F Tyndale has consulted for McNeil and Apotex. Douglas M McMillan

has no conflicts of interest to declare. We would like to thank Dr. Sharon Miksys and Dr. Bin Zhao

for their invaluable technical support and scientific guidance and Fariba Baghai Wadji for her

expert assistance with animal procedures.

Page 85: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

63

2.7 Supplementary Information

Supplementary Material: Effect of brain CYP2D induction on codeine hot-plate analgesia

Figure 17: Nicotine mediated induction of brain CYP2D increased codeine analgesia, and had no effect on morphine-induced analgesia when assessed via the hot plate assay

Figure 17: Nicotine mediated induction of brain CYP2D increased codeine analgesia, and

had no effect on morphine-induced analgesia when assessed via the hot plate assay. Compared

to vehicle pretreatment, nicotine pretreatment resulted in significantly higher %MPE (a) and

analgesic AUC0-30 min (b) after codeine administration (n=8/group), with no difference in analgesia

after morphine administration (c; n=5/group; hatched bars). Error bars indicate SEM. For

analgesia-time curves, **p<0.01 using one-way ANOVA with Bonferroni post-hoc. For AUCs,

**p<0.01 using paired t-tests.

Materials and Methods for the Hot-Plate Assay

A secondary technique for assessment of analgesia was employed in the hot-plate test, through the

use of a hot plate analgesia meter (IITC Life Science, Woodland Hills, CA, USA). In the hot-plate

test, rats are placed on a warm plate enclosed with Plexiglas walls. The latency time to either lick

a hind-paw or jump was used as an endpoint measure (Eddy and Leimbach, 1953). The hot-plate

temperature was set at 52.5⁰C, and rats were removed from the hot-plate immediately upon

meeting endpoint measure, or by the cut-off period of 30 s, to avoid tissue damage. The hot-plate

test employs a more complex behaviour as an endpoint, utilizing a primarily supra-spinal

mechanism (Rowland et al, 1994). Similar to the tail-flick reflex assay, hot plate latencies were

measured two times in each rat 24 h prior to experimentation, and the mean was used as the

individual rat’s baseline latency.

Page 86: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

64

2.8 Significance to Thesis

This chapter contributes to the literature by showing that altering rat brain CYP2D metabolism

impacts acute, systemically administered, opioid response in vivo. This study showed that initial

brain morphine levels and early analgesia after intraperitoneal codeine administration is due, in

part, to brain CYP2D metabolism. Inhibiting rat brain CYP2D decreased brain morphine levels

from codeine and resulting analgesia, while inducing rat brain CYP2D increased brain morphine

levels from codeine and resulting analgesia. Prior to this work, it was unknown if the induction of

rat brain CYP2D by short term nicotine pretreatment would be sufficient to impact drug

metabolism or response in vivo. We showed that nicotine administration induced brain CYP2D

activity in vivo, without affecting hepatic CYP2D activity; inducing brain CYP2D increased brain,

but not plasma, morphine levels from codeine and increased ex vivo dextromethorphan (CYP2D

probe substrate) metabolism in brain membranes, but not liver microsomes. We demonstrated the

specificity of our induction paradigm by co-administering the brain CYP2D inhibitor propranolol;

inhibiting the induced brain CYP2D reversed the effects of brain CYP2D induction on ex vivo

brain CYP2D activity, and brain morphine levels and analgesia from codeine. Additional controls

ensured that there was no effect of nicotine pretreatment on codeine analgesia independent of brain

CYP2D induction. Nicotine pretreatment did not alter baseline nociception, assessed by tail-flick

latency at 8 hr after the last nicotine pretreatment (the time of maximal brain CYP2D induction

and therefore opioid administration). In addition, we found no impact of inducing or inhibiting

brain CYP2D on morphine-induced analgesia (not a CYP2D-substrate) indicating our

pretreatments were affecting codeine analgesia by altering brain CYP2D metabolism.

Findings from this chapter suggest that variation in brain CYP2D activity may contribute to some

of the interindividual variability in opioid response. Individuals with high brain CYP2D activity

(e.g. smokers, individuals taking nicotine) may respond greater to codeine, including analgesia,

subjective effects, and adverse events, due to greater central metabolic activation of codeine to

morphine. Comparatively, individuals with decreased brain CYP2D activity (e.g. individuals

taking CYP2D inhibitors which enter the brain, genetically poor CYP2D6 metabolizers) may

respond less to codeine.

Page 87: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

65

3. Chapter 2: Inducing rat brain CYP2D with nicotine increases the rate of codeine

tolerance; predicting the rate of tolerance from acute analgesic response

Douglas M. McMillan and Rachel F. Tyndale

Reprinted with permission from Elsevier: Biochemical Pharmacology

145: 158-168. © 2017 Elsevier. All rights reserved.

Dr. Rachel F. Tyndale and Douglas M. McMillan designed the overall study. DMM conducted all

analgesia experiments and ex vivo metabolism assays, performed the data analysis, prepared

plasma and brain samples for HPLC detection, and wrote the manuscript. Dr. Bin Zhao performed

all HPLC procedures for codeine, morphine, dextromethorphan, and dextrorphan quantification.

RFT guided the manuscript writing and edited the manuscript.

Page 88: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

66

3.1 Abstract

Repeated opioid administration produces analgesic tolerance, which may lead to dose escalation.

Brain CYP2D metabolizes codeine to morphine, a bioactivation step required for

codeine analgesia. Higher brain, but not liver, CYP2D is found in smokers and nicotine induces

rat brain, but not liver, CYP2D expression and activity. Nicotine induction of rat brain CYP2D

increases acute codeine conversion to morphine, and analgesia, however the role of brain CYP2D

on the effects of repeated codeine exposure and tolerance is unknown. Rats were pretreated with

nicotine (brain CYP2D inducer; 1 mg/kg subcutaneously) or vehicle (saline; 1 ml/kg

subcutaneously). Codeine (40–60 mg/kg oral-gavage) or morphine (20–30 mg/kg oral-gavage)

was administered daily and analgesia was assessed daily using the tail-flick reflex assay. Nicotine

(versus saline) pretreatment increased acute codeine analgesia (1.32-fold change in AUC0-60 min;

p < 0.05) and the rate of loss of peak analgesia (11.42%/day versus 4.20%; p < 0.006) across the

first four days of codeine administration (time to negligible analgesia). Inducing brain CYP2D

with nicotine did not alter acute morphine analgesia (1.03-fold; p > 0.8), or the rate of morphine

tolerance (8.1%/day versus 7.6%; p > 0.9). The rate of both codeine and morphine tolerance (loss

in peak analgesia from day 1 to day 4) correlated with initial analgesic response on day 1 (R = 0.97,

p < 001). Increasing brain CYP2D altered initial analgesia and subsequent rate of tolerance.

Variation in an individual’s initial response to analgesic (e.g. high initial dose, smoking) may affect

the rate of tolerance, and thereby the risk for dose escalation and/or opioid dependence.

Page 89: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

67

3.2 Introduction

Codeine is metabolically activated to morphine by cytochrome P450 2D6 (referred to as CYP2D

for all species), a step required for codeine analgesia (Chen et al, 1991b; Sindrup et al, 1992;

Sindrup et al, 1990). After codeine administration, initial morphine in the brain, and resulting

analgesia are due in part to central nervous system (CNS)-expressed CYP2D-mediated formation

of morphine within the brain (Chen et al, 1990; Pert and Snyder, 1973). This may be due to lower

morphine entry/permeability across the blood–brain barrier, as well as morphine efflux (Bouw et

al, 2000; Oldendorf et al, 1972). CYP-mediated drug metabolism primarily occurs in the liver;

plasma drug and/or metabolite levels of drugs do not always correlate with response to CNS-acting

drugs (Ding and Kaminsky, 2003). CYP-mediated metabolism within the brain can influence

target tissue drug concentration and effect, potentially explaining some of this discrepancy, as well

as contributing to inter-individual variability of drug response (Ferguson and Tyndale, 2011;

Krishna and Klotz, 1994; Miksys and Tyndale, 2009, 2013).

Opioids are effective analgesics for acute (e.g. post-operative) and chronic (e.g. neuropathic) pain,

however opioid drug-related emergency department visits, admissions to substance abuse

treatment, and overdose deaths have increased dramatically over the past 15 years (Cai et al, 2010;

Frenk et al, 2015). The United States is currently the world’s highest per-capita consumer of

opioids, including those used therapeutically (e.g. codeine, oxycodone) and illicitly (e.g.

heroin) (Frenk et al, 2015; Jones et al, 2015; Miech et al, 2015). One contributor to this increase

in opioid use is repeated-exposure to opioids resulting in tolerance, or a loss of analgesic potency,

leading to increased dose-requirements over time (Trang et al, 2015). Tolerance is a result of

neuroadaptation to chronic opioid exposure and mu-opioid receptor activation, however the exact

mechanisms are unknown (Williams et al, 2013). Animal studies suggest that gliosis, changes in

the immune system, and central oxidative stress can play important roles, but little is known of the

upstream role of variation in opioid metabolism, centrally or peripherally, in development of

opioid tolerance (Fan et al, 2015; Grace et al, 2015; Johnson et al, 2014).

We have recently shown that brain CYP2D plays a role in acute codeine analgesia (McMillan and

Tyndale, 2015). Inhibiting rat CYP2D selectively in the brain with a central

(intracerebroventricular, i.c.v.) injection of a mechanism-based inhibitor propranolol (given 24 h

Page 90: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

68

ahead of codeine) decreased brain, but not plasma, morphine levels and analgesia indicative of the

inhibition of brain CYP2D’s formation of morphine from codeine (McMillan and Tyndale, 2015).

We had previously shown that in vivo i.c.v. injections of propranolol inhibited brain, but not

hepatic, ex vivo codeine metabolism to morphine (Zhou et al, 2013). Consistent with the action of

i.c.v. propranolol being selective on inhibiting CYP2D, pretreatment had no effect on analgesia

following morphine injection, which is not a substrate of CYP2D (Zhou et al, 2013). We have also

shown that 7-day subcutaneous (s.c.) nicotine pretreatment induces rat brain, but not hepatic,

CYP2D; brain CYP2D is elevated at 8 h after the last nicotine injection, but there is no detectable

nicotine in plasma (McMillan and Tyndale, 2015; Yue et al, 2008). Inducing rat-brain CYP2D

with 7-day nicotine pretreatment (given 8 h ahead of codeine) increased brain morphine levels and

analgesia, after an intraperitoneal codeine injection; this was reversed by pretreatment with i.c.v.

propranolol (CYP2D inhibitor) (McMillan and Tyndale, 2015). Peak codeine analgesia correlated

with brain, but not plasma, morphine levels and there was no effect of brain CYP2D inhibition or

induction pretreatments on baseline nociception, peripheral codeine or morphine levels, or

analgesia following a morphine injection (McMillan and Tyndale, 2015). These results together

indicated that the early analgesic effects of peripherally administered codeine are dependent, in

part, on brain CYP2D activation of codeine to morphine, prior to the entry into the brain of

morphine produced by hepatic metabolism. While nicotine can elicit analgesia via nicotinic

acetylcholine receptor (nAChR) activation, the timing of the pretreatment (ending 8 h before

codeine), the lack of effect of this pretreatment paradigm on baseline nociception, or morphine-

induced analgesia, and the reversibility of the nicotine pretreatment effect by i.c.v. propranolol

argues against a direct effect of nicotine on analgesia in our antinociception paradigm (Tripathi et

al, 1982). Whether the effect of brain CYP2D on codeine analgesia has implications beyond acute

drug response, to the development of analgesic tolerance to codeine, and other opioid drugs

metabolized by CYP2D, is unknown.

Brain CYPs have high sensitivity to environmental inducers compared to their hepatic

forms (Ferguson and Tyndale, 2011; Miksys and Tyndale, 2013). Liver CYP2D is essentially un-

inducible and variation is primarily due to genetics, whereas brain CYP2D levels can be altered

by these same genetics, and also by environmental exposure to compounds such as

nicotine (Miksys and Tyndale, 2004). Rat and monkey brain, but not liver, CYP2D is induced by

Page 91: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

69

repeated exposure to nicotine, consistent with higher levels of human brain, but not liver, CYP2D

in smokers (Mann et al, 2008; Mann et al, 2012; Miksys and Tyndale, 2004; Miller et al, 2014;

Yue et al, 2008). Individuals with higher brain CYP2D may experience a faster onset of, and

increased, analgesia from codeine, through earlier and greater brain morphine formation. This in

turn could alter the rate of tolerance. Here, we sought to examine the effect of inducing brain

CYP2D on repeated codeine exposure and the development of tolerance to codeine’s analgesic

effects; we hypothesized that codeine tolerance would develop as a function of central morphine

formation, in that inducing rat brain, but not hepatic, CYP2D in vivo would increase the rate of

codeine tolerance during repeated daily oral codeine exposure. Rat CYP2D is a useful model of

human CYP2D6-mediated codeine metabolism due to similarities in substrate specificity, and

relative amount of morphine produced from codeine in vivo by rat CYP2D and

human CYP2D6 (Hedenmalm et al, 1997; Hiroi et al, 2002; Oguri et al, 1990). This study aims

to elucidate if variable rat brain CYP2D activity plays a significant part in chronically administered

CNS-acting drug response, specifically codeine analgesic tolerance, thus extending our

understanding of both tolerance, and the role of brain CYP enzymes in drug response.

3.3 Methods and Materials

Animals

Adult male Wistar rats (250–500 g; Charles River, St-Constant, Canada) were housed in triplets

with ad libitum food and water. Food was restricted one hour prior to, and during, experimentation

to decrease variability in oral drug absorption. Rats were maintained under a 12 h artificial

light/dark cycle with experimentation occurring during the light cycle. All procedures were in

accordance with the Canadian Council on Animal Care guidelines for the care and use of

laboratory animals, and approved by the Animal Care Committee at the University of Toronto.

Drug treatment

Codeine phosphate and morphine sulfate (PCCA, London, Canada) were dissolved in saline and

injected via oral-gavage (p.o.), a route of administration that incorporates first-pass

metabolism and closely resembles the oral route of codeine dosing used by humans. The doses of

codeine (40 mg/kg) and morphine (20 mg/kg) used in the brain CYP2D induction paradigm were

chosen from dose–response curves, to obtain sub-maximal peak analgesia, capable of detecting

Page 92: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

70

potential increases in analgesia, occurring due to brain CYP2D induction, or potential decreases

due to tolerance. The subsequent higher doses of codeine (50 and 60 mg/kg) and morphine (25 and

30 mg/kg) were used as they resulted in incremental increases in peak analgesia relative to the

initial doses of codeine and morphine used, extending the findings on initial analgesia and

tolerance. Nicotine bitartrate (MP Biomedicals, Solon, OH) at 1 mg base/kg (in saline, pH 7.4) or

saline vehicle at 1 ml/kg was given subcutaneously (s.c.) daily for seven days, a paradigm which

results in induced rat brain, but not liver, CYP2D levels and activity (McMillan and Tyndale, 2015;

Yue et al, 2008).

Nociceptive testing of codeine and morphine analgesia

Antinociception (denoted throughout as analgesia) was measured as the percentage of maximal

possible effect (%MPE) by tail-flick latency using a tail-flick meter (Columbus Instruments,

Columbus, OH), as previously described (D'Amour and Smith, 1941; McMillan and Tyndale,

2015). The thermal strength was adjusted to obtain baseline tail-flick latencies (TFL) between 3

and 4 s (lamp intensity 8.0, change in temperature (from room temperature) ∼45 °C); a cut-off of

10 s (∼3-times baseline) was used to avoid tissue damage. TFLs were measured twice in each

animal 30 min prior to experimentation and the mean was used as the individual rat’s baseline

TFL. [%MPE = (post-injection latency − baseline latency)/(maximum cut-off − baseline

latency) × 100%].

Tolerance experimental design

The full experimental design is outlined in Figure 18. Nicotine or saline was administered for

seven days prior to (‘Pretreatment’ period), and during (‘Treatment’ period) opioid administration.

All opioid treatments were given at 8 h after the seventh, and every subsequent, nicotine or vehicle

pretreatment. During the treatment period, test animals were given codeine (40 mg/kg; p.o.) or

morphine (20 mg/kg; p.o.) daily; a full time-course of analgesia was assessed by TFL (every 5 min

for 30 min and then every 10 min for the next 30 min) on treatment days 1 and 7, while analgesia

was assessed at 30 min (peak analgesia on day 1) for the five treatment days in between (days 2–

6). Full analgesia time-courses were not performed on days 2–6 to prevent behavioural adaptation

as well as physical damage inflicted by repeated testing with the tail-flick apparatus. In control

animals, codeine or morphine was given on treatment days 1 and 7 only, with vehicle injections

Page 93: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

71

on the treatment days in between (days 2–6). Analgesia was assessed as performed in test animals.

Animals were deemed tolerant when their post-injection 30 min TFL was reduced by 75% or more

compared with the average 30 min TFL on treatment day 1, and was not statistically different from

baseline TFL (Wang et al, 2005c). Codeine tolerance experiments were performed in a group of

rats (n = 25), split by housing group among pretreatment (nicotine vs. vehicle) and treatment (test

vs. control) group. This was reproduced in a subsequent separate group of rats (n = 25); data are

displayed with both groups combined. Morphine tolerance, and additional codeine/morphine dose

experiments, were run in two separate sets of rats (n = 24/experiment), again split by housing

group for pretreatment (nicotine vs. vehicle), treatment (test vs. control), and/or dose.

Figure 18: Experimental study design and timeline for pretreatment and treatment phases of tolerance experiment

Figure 18: Experimental study design and timeline for pretreatment and treatment phases

of tolerance experiment. Rats in the codeine or morphine tolerance studies were randomized to

receive either daily nicotine (brain CYP2D inducer) or vehicle pretreatment and to be run as a

‘Test’ or ‘Control’, as displayed.

Plasma and brain sample preparation for drug level and enzyme activity analyses

To further assess the impact of nicotine pretreatment on brain and liver CYP2D enzyme activity,

as well as brain and peripheral codeine metabolism, a sub-set of rats in the test codeine paradigm

(n = 12) were sacrificed by decapitation on treatment day 7 at 30 min (approximately peak

analgesia). Trunk blood was centrifuged at 5000g for 10 min for plasma isolation. Whole brains

(minus cerebellum) were homogenized in 0.01 N HCl (1:3 w/v) and centrifuged at 5000g for

10 min.

Page 94: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

72

Membrane preparation

Rat brain CYP2D is present and active in microsomal, plasma, mitochondrial, and nuclear

membrane (Miksys et al, 2000b); total brain membrane fractions were prepared as outlined

previously (Tyndale et al, 1999). In brief, fresh cerebellum (approximately 0.4 g) was

homogenized in 6 ml artificial cerebrospinal fluid (ACSF, pH 7.4) and centrifuged at 3000g for

10 min. The supernatant was centrifuged at 110,000g for 60 min and the resulting pellet was re-

suspended in 150 µl ACSF. Liver microsomes were prepared as previously described (Siu et al,

2006). Protein concentrations were measured using the Bradford assay with a Bio-Rad Protein

Assay Kit (Bio-Rad Laboratories, Mississauga, Canada).

Ex-vivo dextromethorphan oxidation

Formation of dextrorphan from dextromethorphan was used to assess CYP2D enzyme activity

using published methods with some modifications (Kerry et al, 1993; Tyndale et al, 1999). For

brain dextromethorphan metabolism, freshly prepared membranes (1.5 mg protein/ml) were

incubated with 50 µM dextromethorphan hydrobromide (Sigma-Aldrich, Oakville, Canada) and

1 mM NADPH in ACSF for 180 min at 37 °C under 95% O2/5% CO2 in a final volume of 1 ml.

For hepatic dextromethorphan metabolism, microsomal membranes (0.5 mg protein/ml) were

incubated with 2.5 µM dextromethorphan, 100 mM K H2PO4∗H2O buffer (pH 7.4) and 1 mM

NADPH in dH2O for 5 min at 37 °C in a final volume of 0.5 ml. The reactions were stopped with

an equal volume of hexane-butanol (95:5 v/v). Controls included conditions without substrate and

also without membrane protein.

Measurement of drugs

Dextromethorphan and dextrorphan were measured following brain membrane or liver microsome

incubations through modified HPLC methods as described previously. Separate standard curves

for dextromethorphan (50–5000 ng/ml) and dextrorphan tartrate (Sigma-Aldrich; 5–50 ng/ml)

were used. In brief, samples were extracted into 5 ml hexane-butanol (Flores-Pérez et al, 2004)

with 50 ng of 2-benzoxazolinone (Sigma-Aldrich; internal standard), the organic layer was

collected and evaporated to dryness at 37 °C under nitrogen steam, and the residue was re-

dissolved in mobile phase. Dextromethorphan (retention time 22.6 min, recovery rate 72.2%) and

dextrorphan (10.5 min, 78.3%) standard curves, internal standard (13.3 min), and samples were

Page 95: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

73

resolved at ambient temperature on an Agilent 1200 HPLC (Palo Alto, CA) equipped with a

ZORBAX Bonus-RP column (250 mm × 4.6 mm I.D.; particle size, 5 µm; Agilent Technologies,

Mississauga, Canada). Gradient elution conditions used solvent A (methanol and 0.05 M

phosphate buffer, 45:55 v/v, pH = 5.8) and solvent B (HPLC grade water), 0–14 min 100–70%

solvent A at 0.8 ml/min, 14–27 min 70–100% solvent A at 0.8–1.2 ml/min, 27–28 min at constant

solvent A at 1.2–0.8 ml/min. Eluent fluorescence was monitored at an excitation wavelength of

230 nm and an emission wavelength of 330 nm (Hendrickson et al, 2003), with a limit of

quantification of 0.5 ng for dextromethorphan and dextrorphan. Codeine and morphine were

quantified by HPLC as performed previously (McMillan and Tyndale, 2015; Zhou et al, 2013).

Statistical analyses

Between-animal study designs were used. Data were analyzed by repeated measures two-way

ANOVAs followed by Bonferroni corrections for multiple comparisons, unpaired two-tailed t-

tests, or Pearson correlations, as indicated in figure legends.

3.4 Results

Inducing brain CYP2D increased acute codeine analgesia and the rate of codeine tolerance;

the rate of tolerance correlated with initial codeine analgesic response

On treatment day 1 after the first codeine administration, compared to vehicle, nicotine

pretreatment resulted in significantly higher %MPE and overall area under the analgesia-time

curve (AUC0-60 min; p < 0.05, n = 12–13/group; Figure 19A). Animals were given codeine daily for

the following 6 days and analgesia was tested throughout; pretreatments were continued at 8 h

prior to codeine administration. On treatment day 7 of codeine administration (day 14 of nicotine

or vehicle pretreatment), animals showed low levels of analgesia (below 20%; Figure 19B). To

assess the effect of brain CYP2D induction on the rate of codeine tolerance, analgesia was assessed

at 30 min after daily codeine administration, the time at which analgesia peaked on treatment day

1. By treatment day 4 in both pretreatment groups, the majority of animals were tolerant, showing

little change in analgesia from day 4 to day 7, therefore although both 1 to 7 and 1 to 4 were

analyzed, the change in analgesia from treatment days 1 to 4 alone was deemed most relevant.

Compared to vehicle pretreatment, nicotine pretreatment had a faster rate of codeine tolerance

(p < 0.03; Figure 19C), presenting as a more rapid decrease (steeper slope, −10.0 vs. −3.8) in

Page 96: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

74

codeine analgesia across treatment days 1–4. Individual animal analgesia on treatment day 1

(%MPE at 30 min) correlated with the rate of codeine tolerance (p < 0.001; Figure 19D),

suggesting that initial codeine analgesic response predicted the rate of codeine tolerance (which

was higher and faster in the brain CYP2D induced group). Animal baseline TFLs prior to codeine

treatment did not change significantly across the treatment period (p > 0.7), suggesting no

increased sensitization (hyperalgesia) across repeated codeine administration, or between

pretreatment groups (p > 0.9), suggesting no effect of nicotine on baseline response.

Compared to vehicle pretreated rats (8.6 + 1.5 nmol/mg/h, mean + SEM), nicotine pretreated rats

had significantly higher ex vivo brain CYP2D activity (13.5 + 1.3 nmol/mg/h; p < 0.05 sacrificed

on treatment day 7 at 30 min after codeine administration) consistent with brain CYP2D induction

by nicotine. There was no change in ex vivo hepatic CYP2D activity between vehicle

(0.17 + 0.01 nmol/mg/min) and nicotine (0.19 + 0.03 nmol/mg/min; p > 0.6) pretreatment groups.

Consistent with increased brain, but not liver, CYP2D enzyme activity, animals pretreated with

nicotine trended towards a higher brain morphine/codeine ratio (0.18 + 0.05, mean + SEM,

compared to 0.09 + 0.02; p < 0.2) with no impact on the plasma morphine/codeine ratio

(0.49 + 0.09 compared to 0.41 + 0.18; p > 0.7), suggesting an effect of nicotine pretreatment on

central, but not peripheral, metabolism of codeine to morphine.

Page 97: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

75

Figure 19: Nicotine pretreatment increased acute codeine-induced analgesia; repeated codeine injections resulted in analgesic tolerance, the rate of which was increased by nicotine pretreatment

Figure 19: Nicotine pretreatment increased acute codeine-induced analgesia; repeated

codeine injections resulted in analgesic tolerance, the rate of which was increased by nicotine

pretreatment. Compared with vehicle, nicotine pretreatment increased %MPE and analgesic

AUC0-60 min (a) after acute codeine administration (40 mg/kg, p.o.; n = 12–13/group); repeated

daily codeine administration resulted in analgesic tolerance (b). Compared to vehicle, nicotine

pretreatment increased the rate of onset of codeine tolerance (c); individual animal peak analgesia

on day 1 (at 30 min) correlated to the decrease in analgesia between days 1–4 (d). *p < 0.05

and **p < 0.01 using repeated measures two-way ANOVA with Bonferroni post-hoc test and t-

tests. For correlations, Pearson coefficients were used.

Effect of brain CYP2D induction was maintained and there was no significant change in

analgesia after a one-week washout between codeine administrations

To determine whether the changes in analgesia across repeated codeine injections were due to

tolerance to codeine’s analgesic effect, control groups receiving all pretreatments, but daily vehicle

instead of codeine injections on treatment days 2–6, were assessed. This also tested the extent to

which vehicle injections on treatment days 2–6 and daily analgesia testing sensitized animals to

Page 98: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

76

the behavioural assay. As a replication of the findings in Figure 19A, after acute codeine

administration compared to vehicle pretreatment, nicotine pretreatment resulted in significantly

higher %MPE and AUC0-60 min (n = 12–13/group; Figure 20A) as seen in test codeine animals

(Figure 19A). On treatment day 7, after one-week without codeine, the effect of nicotine-mediated

induction of brain CYP2D on codeine analgesia remained; animals pretreated with nicotine had

higher %MPE and AUC0-60 min (Figure 20B) after codeine than vehicle pretreated animals. After

each vehicle administration on treatment days 2–6 (Figure 20C) there was no significant difference

in %MPE between pretreatment groups (p > 0.4 across all days) or TFLs compared to baseline

(p > 0.2). Comparing codeine response on treatment days 1 and 7, there was no correlation between

individual animal peak analgesia on day 1 (%MPE at 30 min) and change in response from day 1

to day 7 (flat slope; Figure 20D), suggesting no effect of repeated vehicle injections and testing on

codeine induced analgesia.

Inducing brain CYP2D did not affect acute morphine analgesia or the rate of morphine

analgesic tolerance; there was no change in morphine analgesia after a one-week washout

Morphine is the active analgesic metabolite of codeine and is not a substrate of CYP2D; thus,

inducing brain CYP2D had no effect on acute morphine analgesia (McMillan and Tyndale, 2015)

and should not alter the rate of morphine tolerance. On treatment day 1 after acute morphine

administration, compared to vehicle pretreatment, nicotine pretreatment did not alter %MPE or

AUC0-60 min (n = 6–7/group; Figure 21A). On treatment day 7, animals showed negligible

analgesia and there remained no effect of nicotine pretreatment on %MPE and AUC0-

60 min (Figure 21B). Similar to the codeine experiment (Figure 19C), the majority of test animals in

both pretreatment groups experienced tolerance by treatment day 4 with negligible remaining

analgesia and little change through day 7. Analgesia decreased at the same rate in both pretreatment

groups (similar slopes, p > 0.9; Figure 21C) from treatment days 1–4 of morphine administration.

Similar to codeine, individual animal analgesia on day 1 (%MPE at 30 min) correlated with the

rate of morphine tolerance (p < 0.001; Figure 21D), suggesting that initial response to codeine or

morphine predicted the rate of tolerance; the higher initial analgesia, the faster the decline in

analgesia.

Page 99: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

77

Figure 20: Control animals showed no change in analgesia after a one-week washout between codeine injections; analgesia was not altered by daily p.o. (i.e. codeine vehicle) injections or behavioural testing

Figure 20: Control animals showed no change in analgesia after a one-week washout

between codeine injections; analgesia was not altered by daily p.o. (i.e. codeine vehicle)

injections or behavioural testing. Compared with vehicle, nicotine pretreatment increased

%MPE and analgesic AUC0-60 min(a) after acute codeine administration (40 mg/kg, p.o.; n = 12–

13/group) as expected and observed previously (Fig. 19a); the effect of nicotine pretreatment on

codeine analgesia was sustained after a one-week washout (b). There was no effect of p.o. vehicle

injections on %MPE across days 2–6 (c); negative slope shows slight decrease in analgesia

between codeine injections one-week apart (d). *p < 0.05, **p < 0.01 and ***p < 0.001 using

repeated measures two-way ANOVA with Bonferroni post-hoc test and t-tests. For correlations,

Pearson coefficients were used.

Page 100: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

78

Figure 21: Nicotine pretreatment did not alter morphine-induced analgesia; repeated morphine injections resulted in analgesic tolerance, the rate of which was not altered by nicotine pretreatment

Figure 21: Nicotine pretreatment did not alter morphine-induced analgesia; repeated

morphine injections resulted in analgesic tolerance, the rate of which was not altered by

nicotine pretreatment. Compared with vehicle, nicotine pretreatment did not alter %MPE and

analgesic AUC0-60 min (a) after acute morphine administration (20 mg/kg, p.o.; n = 6/group);

repeated daily morphine administration resulted in analgesic tolerance (b). Compared to vehicle,

nicotine pretreatment did not alter the rate of onset of morphine tolerance (c); individual animal

peak analgesia on day 1 (at 30 min) correlated to the decrease in analgesia between days 1–4 (d).

Repeated measures two-way ANOVA with Bonferroni post-hoc test, t-tests and Pearson

coefficients were used.

In control animals, compared to vehicle pretreatment, nicotine pretreatment did not alter %MPE

or AUC0-60 min (n = 6–7/group; Figure 22A) after acute morphine administration on day 1, or

morphine on day 7 after a one-week washout (Figure 22B). Similar to codeine control animals

(Figure 20), there was no significant difference in %MPE between pretreatment groups (p > 0.3

across all days) or TFLs compared to baseline (p > 0.7) after vehicle administration on treatment

days 2–6 (Figure 22C), as well as no correlation between individual animal peak analgesia on day

Page 101: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

79

1 and change in response after washout (flat slope; Figure 22D), overall suggesting no effect of

repeated vehicle injections and testing on morphine induced analgesia.

Figure 22: Control animals showed no change in analgesia after a one-week washout between morphine injections; analgesia was not altered by daily p.o. (i.e. morphine vehicle) injections and behavioural testing

Figure 22: Control animals showed no change in analgesia after a one-week washout

between morphine injections; analgesia was not altered by daily p.o. (i.e. morphine vehicle)

injections and behavioural testing. Compared with vehicle, nicotine pretreatment did not alter

%MPE and analgesia AUC0-60 min(a) after acute morphine administration (20 mg/kg, p.o.;

n = 6/group); analgesia was sustained after a one-week washout (b). There was no effect of p.o.

vehicle injections on %MPE across days 2–6 (c); negative slope shows slight decrease in analgesia

between morphine injections one-week apart (d). Repeated measures two-way ANOVA with

Bonferroni post-hoc test, t-tests and Pearson coefficients were used.

Rate of tolerance correlated directly to initial analgesic response, independent of

pretreatment and codeine or morphine dose

To further assess the relationship between increasing initial analgesia and development of

tolerance to codeine and morphine, a group of animals without pretreatments (i.e. unaltered brain

Page 102: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

80

CYP2D activity) were given higher doses of daily codeine (50 or 60 mg/kg; p.o.) and morphine

(25 or 30 mg/kg, p.o.). Increased doses of codeine and morphine resulted in significantly higher

day 1%MPE and analgesic AUC (p < 0.001 and p = 0.05, respectively) using two-way ANOVA.

As seen previously (Figures 19 and 21), with daily dosing, these animals became tolerant by

treatment day 4 (Fig. 23C); individual animal peak-analgesia on day 1 (%MPE at 30 min)

correlated with the rate of tolerance (p < 0.001; Figure 23D). Combining the data from codeine

and morphine alone (Fig. 23), with data following brain CYP2D induction (Figures 19 and 21),

confirmed that the initial peak analgesia from acute exposure (%MPE at 30 min on treatment day

1) correlated with the mean rate of tolerance from treatment days 1–4 in all test groups

(p < 0.001; Figures 24A and B). This correlation remained significant when comparing initial

peak analgesia to slope from day 1 to day 7 (R = 0.95, p < 0.001). This again confirmed that acute

day 1 initial analgesic response predicted the rate of tolerance and that the pretreatments simply

altered the magnitude of initial analgesic response. Codeine, via CYP2D metabolism to morphine,

and morphine, via direct action, elicit analgesia through morphine, suggesting a relationship

between morphine action at the opioid receptor and the rate of tolerance.

Page 103: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

81

Figure 23: Initial analgesia correlated to rate of tolerance in animals given higher doses of codeine and morphine

Figure 23: Initial analgesia correlated to rate of tolerance in animals given higher doses of

codeine and morphine. Acute codeine (50 or 60 mg/kg, p.o.) and morphine (25 or 30 mg/kg; p.o.)

administration produced peak analgesia in a dose dependent manner (n = 5–6/group, p < 0.001 and

p = 0.05 compared to earlier doses of codeine and morphine, respectively; a); repeated daily

administration resulted in analgesic tolerance (b). Rate of tolerance (slope from day 1 to 4) was

dose dependent (c) and correlated to peak analgesia after acute administration on day 1

(d). *p < 0.05, **p < 0.01 between codeine doses and #p < 0.01 between morphine doses using two-

way ANOVA with Bonferroni post-hoc. Pearson coefficients were used.

Page 104: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

82

Figure 24: Initial analgesia predicts the rate of onset of tolerance across multiple codeine and morphine doses

Figure 24: Initial analgesia predicts the rate of onset of tolerance across multiple codeine

and morphine doses. Peak analgesia after the first codeine or morphine injection correlated to the

change in peak analgesia across the first four days of codeine (n = 12–13/group; n = 5–6/group for

higher doses) or morphine (n = 6/group; n = 5–6/group for higher doses) administration, regardless

of dose (Codeine: 40, 50, and 60 mg/kg p.o.; Morphine: 20, 25, and 30 mg/kg p.o.) or pre-treatment

(seven-day nicotine, seven-day vehicle, or no pretreatment). Data are displayed as group means

(a) and individual animals (b). Pearson correlations were used.

3.5 Discussion

This is the first study to show that nicotine-induced brain CYP2D metabolism of codeine

to morphine can alter the response to repeated codeine (i.e. tolerance), providing evidence that

brain CYP-mediated metabolism can impact chronically administered CNS-acting drug effects.

During chronic daily opioid induced analgesia, induced brain CYP2D activity increases initial

codeine analgesia, resulting in a faster rate of codeine induced analgesic tolerance. Further, it

demonstrated that increasing initial analgesia, from either codeine or morphine, increased the rate

of tolerance (decrease in peak analgesia across days) with no effect on time to full tolerance.

Inducing brain CYP2D with nicotine increased initial codeine analgesia, and resulted in a

significantly greater rate of decrease in analgesia across the first four days of treatment (steeper

slope), compared to vehicle pretreated animals that had less initial codeine analgesia, and a

comparatively decreased rate of tolerance (shallower slope). The rate was increased, rather than

Page 105: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

83

resulting in an equal rate of tolerance but longer sustained analgesia in animals with a higher initial

analgesic response. In animals given daily morphine (not a substrate of CYP2D), there was no

effect of nicotine induction of brain CYP2D on initial morphine analgesic response or on the rate

of tolerance. The relationship between initial analgesia and tolerance was further supported in

animals without brain CYP2D pretreatments, where increased codeine and morphine doses

administered increased initial analgesia, and thereby resulted in an increase in the absolute drop in

analgesia, or rate of tolerance, but not in the time to reach tolerance (i.e. days to negligible

analgesia). These findings are related to a 1969 paper which indicated that degree of tolerance to

levorphanol-induced running-fit in mice was related to its original (first injection)

intensity (Goldstein and Sheehan, 1969). This finding suggested a link between acute and chronic

opioid response, using a distinct animal model of excitatory opioid action; we are unaware of any

publications illustrating the relationship between initial analgesia and rate of analgesic tolerance

to opioids.

The initial analgesic response and rate of tolerance correlated in all animals assessed, independent

of brain CYP2D activity (vehicle or nicotine pretreatment) or dose of compound (codeine or

morphine), confirming that the rate of tolerance was related to initial analgesic response. In all

experimental conditions, the majority of test animals experienced analgesic tolerance by the fourth

opioid administration, which is interesting to consider as it relates to the rate of codeine tolerance

with and without nicotine-induced brain CYP2D. Nicotine pretreated animals experienced a

similar proportional daily decrease (28% per day) in codeine analgesia to vehicle pretreated

animals (33%), however the absolute drop was greater (11.2%MPE per day vs. 4.7%MPE). In

individuals who experience greater initial pain relief, a 50% decrease in efficacy of an opioid may

present as a greater, or more noticeable, drop in drug efficacy than those who experience less initial

analgesia. Thus, while the drop is proportional, for those with higher initial analgesia, the absolute

drop is greater. A greater drop in response could, in turn, lead to a greater motivation to seek better

pain control resulting in quicker or more pronounced dose-escalation. In individuals with cancer

pain challenged with a morphine dose before and after one week of regular morphine treatment, a

∼1.5-fold greater morphine dose was required to achieve the same degree of pain relief (Houde

and Beaver, 1966). Our findings suggest that a greater magnitude of response from the first

morphine challenge, potentially resulting in more rapid tolerance, can result in dose escalation in

Page 106: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

84

a shorter time frame than one week. In support of this, individuals treated by “high-dose”

emergency physicians showed significantly increased odds of long-term opioid use compared to

those treated by “low-dose” emergency physicians (and “high-intensity” compared to “low-

intensity” physicians) (Barnett et al, 2017). Chronic opioid dose escalation in pain patients and

illicit opioid users is well documented; our findings on the impact of altered brain CYP2D

metabolic activation of codeine shed some light on one potential mechanism which might motivate

differential dose escalation (Collett, 1998; Cowan et al, 2001).

These findings provide one possible mechanistic link for an impact of smoking on increasing

codeine dependency. The nicotine pretreatment paradigm used in this study resulted in average

daily nicotine plasma levels similar to those seen in human smokers and experienced electronic

cigarette (e-cigarette) users (20–50 ng/ml) (Farsalinos et al, 2015; McMillan and Tyndale, 2015;

Velez de Mendizabal et al, 2015); similar to nicotine-treated animals, smoking is associated with

higher human brain, but not hepatic, CYP2D (Miksys and Tyndale, 2004). Studies assessing the

impact of smoking on opioid tolerance are often conducted in chronic, and post-operative, pain

situations where findings are inconsistent; smoking has been found to increase, decrease and have

no effect on opioid escalation across time (Chiang et al, 2016; Skurtveit et al, 2010; Weingarten

et al, 2011). However, the acute and chronic effects of nicotine on nociception, pain sensitivity,

and interaction with the wide-range of opioids with analgesia affected (e.g. codeine) or unaffected

(e.g. morphine) by potential changes in CYP2D activity, is complex and multifactorial (Girdler et

al, 2005). The effect of nicotine on increasing brain CYP2D-mediated codeine metabolic

activation and analgesia, and the resulting acceleration in the rate of analgesic tolerance could

contribute to potential dose-escalation, which may increase the prevalence of codeine-dependency

among smokers.

One concern with continued nicotine pretreatment throughout the treatment phase, was the

potential interaction of nicotine with codeine and morphine-induced analgesia. Nicotine can play

a role in inhibitory-pain control, and can elicit analgesia in animal models through the involvement

of endogenous opioid transmission (Berrendero et al, 2002). Nicotine and opioids also

exhibit cross-tolerance or cross-sensitization (Vihavainen et al, 2008; Zarrindast et al, 2003). We

have previously shown, as well as confirmed here, that under the current nicotine dosing regime,

Page 107: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

85

just prior to codeine or morphine administration, nicotine levels are undetectable (McMillan and

Tyndale, 2015). We also showed that nicotine pretreatment did not alter pre- or post-injection

baseline latencies (suggesting no effect on locomotion), ex vivo hepatic CYP2D activity and

hepatic morphine/codeine ratios in codeine test animals, nor did it have an effect on acute morphine

analgesia, or the rate of morphine tolerance. This is consistent with a lack of direct nicotine effect

itself, on opioid-induced analgesia, via nAChR activation or otherwise. We saw an effect of

nicotine pretreatment on ex vivo brain CYP2D activity but failed to see a significant change on

brain morphine/codeine ratio; ratios were examined in whole brain homogenates (minus

cerebellum), which may have blunted the ability to see regional impacts expected due to the

heterogeneity in CYP2D expression and induction (Miksys and Tyndale, 2004; Yue et al, 2008).

An additional limitation of this study is in modelling human brain CYP2D6 with rat brain CYP2D;

while CYP2D6 is the only functional CYP2D isoform in humans, six homologous isoforms have

been identified in rats which show some variation in substrate specificity and tissue

expression (Haduch et al, 2011; Hiroi et al, 2002; Hiroi et al, 1998a). Rat CYP2D isoforms and

human CYP2D6 are expressed in the brain, catalyze the O-demethylation of codeine to morphine

and dextromethorphan to dextrorphan, and mediate the mechanism-based (suicide) inhibition

by propranolol (Grobe et al, 2012; Hiroi et al, 2002; Hiroi et al, 1998a; Rowland et al, 1994;

Zanger et al, 2004). In addition, both rat and non-human primate (i.e. African Green Monkey)

brain, but not liver, CYP2D are induced by nicotine in vivo consistent with the higher brain, but

not liver, CYP2D in human smokers (Mann et al, 2008; Miksys et al, 2002; Miller et al, 2014;

Yue et al, 2008; Zanger and Schwab, 2013). Together this suggests that while the precise isoform

involved is unknown, the rat is useful for modelling these CNS aspects of human brain CYP2D6.

The tail-flick reflex assay is a reliable and reproducible assay indicating spinal and supra-spinally

mediated analgesia; the tail-flick reflex also shows little inter-animal variability in baselines or

with repeated testing, rarely occurs spontaneously, and can predict the analgesic effects of opioids

in humans (Grossman et al, 1982; Le Bars et al, 2001). We have previously shown that nicotine

induction of brain CYP2D resulted in an increase in codeine, but not morphine, analgesia measured

by the hot-plate assay (McMillan and Tyndale, 2015). Lastly, the mechanism of induction of brain,

but not hepatic, CYP2D expression with chronic nicotine pretreatment is currently unknown, but

occurs in the absence of changes in mRNA suggesting it may be mediated by increased enzyme

stability or decreased degradation (Yue et al, 2008).

Page 108: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

86

The implications of our findings are that individuals with higher brain CYP2D activity (e.g.

genetically ultra-rapid CYP2D metabolizers or smokers), those taking larger initial doses, or those

more sensitive to opioids via an alternative mechanism (e.g. greater mu-opioid receptor

availability) may experience a greater initial response to, and therefore more rapid rate of tolerance

to, codeine. The influence of brain CYP2D metabolism on acute codeine response has been

observed with tramadol, another opioid analgesic metabolized by CYP2D. Selective rat brain

CYP2D inhibition and induction decreased and increased, respectively, central tramadol

metabolism and analgesia (Wang et al, 2015). This is further evidence that altered brain CYP2D

metabolism alters acute opioid metabolic activation and analgesia, suggesting the potential for

altering tolerance to tramadol response and potentially to other CYP2D6 substrate opioids

including oxycodone and hydrocodone (Hutchinson et al, 2004; Lalovic et al, 2006). Beyond

genetics and smoking, there are numerous environmental and endogenous factors that can further

contribute to variable brain CYP2D, including hormonal, sex and developmental factors (Wang et

al, 2014). Monkeys self-administering ethanol had a dose-dependent increase in brain, but not

liver, CYP2D, consistent with increased brain CYP2D levels in human alcoholics (Miksys et al,

2002; Miller et al, 2014). The neuroleptic clozapine and neurotoxin toluene have also been shown

to induce rat brain CYP2D (Hedlund et al, 1996). While sex-based differences in brain CYP2D

activity and expression remain unclear, sex hormones play a role in regulating expression of brain

CYP2D; testosterone and progesterone produced a marked increase and decrease, respectively, of

brain CYP2D mRNA levels female rats (Aichhorn et al, 2005; Baum and Strobel, 1997; Bergh

and Strobel, 1996; Hildebrandt et al, 2003; Li et al, 2015a; Waxman and Holloway, 2009).

Furthermore, the expression of human brain CYP2D6 increases with age (Mann et al, 2012). Thus,

being a smoker, having rapid CYP2D genetics, taking higher initial doses, or any prior-mentioned

cause for induced brain CYP2D metabolism may increase the rate of tolerance and risk of dose

escalation and/or dependence, providing an argument for lower initial doses in pain treatment.

Likewise, even with repeated morphine administration, the higher initial response predicted a

faster rate of tolerance suggesting that even among non-enzymatically activated opioids initiating

pain relief with the lowest possible doses may be prudent.

Page 109: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

87

This study provides a novel mechanistic approach to understanding the potential role of central

metabolism on individual differences in pain relief and tolerance to codeine and potentially other

CYP2D substrate opioids (e.g. hydrocodone and tramadol). To our knowledge there has been little

investigation of whether initial analgesia predicts tolerance, and if it does, whether initial analgesia

is related to a proportional drop in analgesia (as seen by a change in slope) or an absolute drop in

analgesia (similar slopes, and thus a longer time to tolerance in those with higher initial analgesia).

Codeine remains a commonly abused opioid analgesic, largely due to its use in prescription and

over-the-counter analgesic and cough syrup combinations (Cooper, 2013; Roussin et al, 2013).

Codeine use also extends beyond prescription; in the US in 2014, 64.3 million people reported

some lifetime non-medicinal opioid use (19.4 million attributed to codeine products), with

12.5 million people reporting current opioid misuse in 2015 alone (NSDUH, 2016, 2017).

However, most opioid drugs are metabolized by CYP2D to compounds that are more potent µ-

opioid receptor agonists than the parent opioid, suggesting that the findings from this study provide

a proof-of-concept that could extend beyond codeine to other commonly abused opioid analgesics

(e.g. hydrocodone and tramadol). With the ongoing rise in risk of opioid use, abuse, dependency

and overdose, the role of opioid metabolism within the brain, and variability in this metabolism,

may be a novel source of inter-individual differences in opioid response and tolerance.

3.6 Acknowledgements

We would like to thank Dr. Sharon Miksys and Dr. Bin Zhao for their invaluable technical

assistance and scientific guidance, and Fariba Baghai Wadji for her expert assistance with animal

procedures. This work was supported by a Canada Research Chair in Pharmacogenetics, Canadian

Institutes of Health Research [grant numbers TMD 132557 and MOP 136937]; the Centre for

Addiction and Mental Health and the CAMH Foundation; the Canada Foundation for Innovation

[grant numbers 20289 and 16014]; and the Ontario Ministry of Research and Innovation. RFT has

consulted for Apotex on topics unrelated to this work. DMM has no conflicts of interest to declare.

Page 110: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

88

3.7 Significance to Thesis

This original article expands on our findings of an impact of brain CYP2D metabolism on acute

codeine administration, by showing a novel role for brain CYP2D in the development of codeine

analgesic tolerance after repeated codeine administration. This study used oral gavage, a more

clinically relevant route of codeine administration, and replicated findings from our first study that

inducing rat brain CYP2D increased initial codeine analgesia. This increase in initial codeine

analgesia resulted in an increased the rate of codeine analgesic tolerance following repeated

codeine administration. This increased rate of tolerance presented as a greater absolute drop in

codeine analgesia (similar proportional drop) per oral injection. Furthermore, this study replicated

findings that inducing brain CYP2D did not alter morphine-induced initial analgesia (not a CYP2D

substrate), again using oral gavage administration, and as expected found no impact of inducing

brain CYP2D on the rate of morphine tolerance across repeated morphine administration. This

study showed that the rate of codeine or morphine tolerance correlated to initial peak analgesia

after the first codeine or morphine administration. This relationship was observed when initial

codeine, but not morphine, analgesia was increased by inducing brain CYP2D and also when initial

codeine and morphine analgesia was increased by giving greater doses.

We demonstrated here for the first time that initial analgesia 1) is related to the rate of tolerance,

and 2) does not change the time to reach tolerance. Increasing initial analgesia produced a greater

absolute decrease in analgesia or rate of tolerance (slope) but similar time to tolerance (4 days in

all groups) rather than a similar absolute decrease in analgesia and therefore longer time to

tolerance. The clinical implications of these findings are that individuals who experience greater

initial opioid effect would experience a more dramatic decrease in drug effect with repeated opioid

taking. The consequence of a greater perceived decrease in opioid-induced analgesia over time

may be that these individuals are more likely to increase opioid dose to achieve the initial level of

analgesia, and therefore they may be more vulnerable to the associated risks of higher opioid doses.

Consequently, this study provides a further rationale for initiating opioid therapy at the lowest

analgesic doses possible.

Page 111: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

89

4. Chapter 3: Rat brain CYP2D activity alters in vivo central oxycodone metabolism, levels,

and resulting analgesia

Douglas M. McMillan, Sharon Miksys, and Rachel F. Tyndale

Reprinted with permission from Wiley: Addiction Biology

Epub ahead of print © 2017 Wiley. All rights reserved.

Dr. Rachel F. Tyndale, Dr. Sharon Miksys and Douglas M. McMillan designed the overall study.

DMM 1) conducted all analgesia, microdialysis, and ex vivo metabolism experiments, 2)

performed the data analysis, and 3) wrote the manuscript. SM performed some pilot studies. Fariba

Baghai Wadji assisted in animal cannulation surgeries and optimizing the microdialysis assay for

oxycodone and metabolite detection. Dr. Bin Zhao developed and performed the LCMS assay for

quantification of oxycodone and metabolites. RFT guided the manuscript writing and RFT and

SM edited the manuscript.

Page 112: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

90

4.1 Abstract

Oxycodone is metabolized by CYP2D to oxymorphone. Despite oxymorphone being a more potent

opioid-receptor agonist, its contribution to oxycodone analgesia may be minor because of low

peripheral production, low blood-brain barrier permeability and central nervous system efflux.

CYP2D metabolism within the brain may contribute to variation in central oxycodone and

oxymorphone levels, thereby affecting analgesia. Brain CYP2D expression and activity are subject

to exogenous regulation; nicotine induces rat brain, but not liver, CYP2D consistent with higher

brain CYP2D in smokers. We assessed the role of rat brain CYP2D in orally administered

oxycodone metabolism (in vivo brain microdialysis) and analgesia (tail-flick test) by inhibiting

brain CYP2D selectively with intracerebroventricular propranolol (mechanism-based inhibitor)

and inducing brain CYP2D with nicotine. Inhibiting brain CYP2D increased brain oxycodone

levels (1.8-fold; P < 0.03) and analgesia (1.5-fold AUC0-60; P < 0.001) after oxycodone, while

inducing brain CYP2D increased brain oxymorphone levels (4.6-fold; P < 0.001) and decreased

analgesia (0.8-fold; P < 0.02). Inhibiting the induced brain CYP2D reversed the change in

oxycodone levels (1.2-fold; P > 0.1) and analgesia (1.1-fold; P > 0.3). Brain, but not plasma,

metabolic ratios were affected by pre-treatments. Peak analgesia was inversely correlated with ex

vivo brain (P < 0.003), but not hepatic (P > 0.9), CYP2D activity. Altering brain CYP2D did not

affect analgesia from oral oxymorphone (P > 0.9 for AUC0-60 across all groups), which is not a

CYP2D substrate. Thus, brain CYP2D metabolism alters local oxycodone levels and response,

suggesting that people with increased brain CYP2D activity may have reduced oxycodone

response. Factors that alter individual oxycodone response may be useful for optimizing treatment

and minimizing abuse liability.

Page 113: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

91

4.2 Introduction

Oxycodone is a semisynthetic opioid analgesic widely used to treat moderate to severe pain (Riley

et al, 2008), with analgesic efficacy for neuropathic (e.g. diabetic neuropathy), somatic (e.g.

osteoarthritis), visceral (e.g. myocardial ischemia) and cancer pain (Riley et al, 2008). Despite

recent efforts to reduce both oxycodone prescribing and misuse with the development of abuse-

deterrent delivery formulations, oxycodone abuse remains a major public health concern (Webster,

2009). In 2015, in all Americans over the age of 12 years, approximately 28 million reported

oxycodone use, and 4.3 million (15.3 percent of users) reported oxycodone misuse (NSDUH,

2016). In these individuals, oxycodone-containing products accounted for 29 percent of total

opioid use and 34 percent of opioid misuse (NSDUH, 2016). Elucidating potential sources of

variation in opioid response remains important for understanding individual differences in opioid

analgesic response and risk of abuse.

Oxycodone is metabolized by cytochrome P450 (CYP) enzymes to three primary metabolites:

oxymorphone (by CYP2D6 in humans; 11 percent of urinary recovery), noroxycodone (by

CYP3A4 in humans; 45–47 percent of urinary recovery) and noroxymorphone (by CYP3A4 and

CYP2D6 in humans; 14 percent of urinary recovery) (Klimas et al, 2013; Lalovic et al, 2006;

Lalovic et al, 2004). Parent opioid and/or metabolite contribution to analgesia depends on

numerous factors including target receptor affinity (primarily the μ-opioid receptor), proportional

peripheral metabolism, influx across the blood–brain barrier (BBB) relative to efflux from the

central nervous system (CNS) and concentrations in the brain (Bostrom et al, 2005; Mercer and

Coop, 2011); the relative contribution of oxycodone and its metabolites to oxycodone analgesia is

unclear (Lalovic et al, 2006).

Cytochrome P450 enzymes are present and active in the brain and can be induced independently

from hepatic CYPs (Miksys and Tyndale, 2013). While hepatic CYP2D levels are regulated

primarily by genetics, brain CYP2D expression may be altered by genetics, age and environmental

exposures such as to smoking and nicotine (Miksys and Tyndale, 2004). Modifying rat brain-

specific CYP2D enzyme activity in vivo can alter the central metabolism and effect of CNS-acting

CYP2D substrates, including opioids (e.g. codeine and tramadol) (McMillan and Tyndale, 2015;

Wang et al, 2015). Codeine requires activation by CYP2D to morphine to produce analgesia;

Page 114: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

92

individuals who are genetically CYP2D poor metabolizers (lacking CYP2D6 activity), or who are

given CYP2D inhibitors, produce little-to-no morphine and exhibit little-to-no codeine analgesia

(Chen et al, 1991b; Sindrup et al, 1992; Sindrup et al, 1990). Selectively inhibiting and inducing

rat brain CYP2D decreased and increased, respectively, brain, but not plasma, morphine levels and

analgesia after codeine administration (McMillan and Tyndale, 2015). Unlike codeine, which is

essentially inactive, oxycodone is a potent μ-opioid receptor agonist, and compared with its

CYP2D metabolite oxymorphone, oxycodone is present at higher concentrations and has a higher

partition across the BBB (rat brain/plasma ratio of 2.07 versus 0.23 at 60 minutes after intragastric

oxycodone) (Bostrom et al, 2008; Lalovic et al, 2006). Therefore, while the influence of brain

CYP2D metabolism on codeine analgesia is via metabolic activation, the influence of brain

CYP2D metabolism on oxycodone analgesia is unclear and likely multifactorial. Brain CYP2D

metabolism may contribute to oxycodone analgesia through formation of oxymorphone, which has

a greater μ-opioid receptor affinity than oxycodone, may reduce oxycodone analgesia through

local oxycodone metabolism or may have no impact on oxycodone analgesia (Klimas et al, 2013).

Our objective was to investigate the central CYP2D-mediated metabolism of oxycodone to

oxymorphone and resulting analgesia, by selectively inhibiting and/or inducing rat brain CYP2D

activity. The pre-treatments used to manipulate brain CYP2D metabolism have been shown to

modify brain-specific CYP2D activity without altering (1) hepatic CYP2D, (2) baseline

nociception or (3) antinociception from non-CYP2D substrates (e.g. morphine-induced analgesia)

(McMillan and Tyndale, 2015; Miksys et al, 2017). Because of the possible opposing impacts of

brain CYP2D on oxycodone analgesia as described previously, we used in vivo brain microdialysis

to assess brain oxycodone and major metabolite levels. This enabled assessment of the relationship

over time between brain CYP2D-mediated oxycodone metabolism, resulting oxycodone and

oxymorphone levels, and oxycodone analgesia.

4.3 Materials and Methods

Animals

Male Wistar rats (Charles River Laboratories, Saint-Constant, Canada) were house in triplets

with ad libitum food and water, under a 12-hour artificial light/dark cycle with experimentation

occurring during the light cycle. Adult animals were used; over the duration of the experiments,

Page 115: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

93

ages ranged from 8 to 14 weeks old and weights from 250 to 500 g. All procedures were conducted

in accordance with the Canadian Council on Animal Care guidelines for the care and use of

laboratory animals and approved by the Animal Care Committee at the University of Toronto.

Drug treatment

Oxycodone hydrochloride (Professional Compounding Centers of America, London, Canada) and

oxymorphone hydrochloride (Toronto Research Chemicals Inc., Toronto, Canada) were dissolved

in dH2O and administered by oral gavage (p.o.). Selection of doses is outlined in Figure 25.

Propranolol hydrochloride (Sigma-Aldrich, Oakville, Canada) was dissolved in 20 percent (w/v)

2-hydroxypropyl-β-cyclodextrin (cyclodextrin, Sigma-Aldrich) in dH2O to deliver 20 μg base in

4 μl intracerebroventricularly (i.c.v.) 24 hours prior to oxycodone (or oxymorphone)

administration (McMillan and Tyndale, 2015; Zhou et al, 2013). Propranolol is a CYP2D

mechanism-based inhibitor (Narimatsu et al, 2001); in rats, i.c.v. propranolol pre-treatment

inhibits brain, but not liver, ex vivo dextromethorphan and codeine metabolism, indicative of in

vivo brain, but not liver, irreversible CYP2D inhibition (Miksys et al, 2017; Zhou et al, 2013). To

deliver propranolol or vehicle i.c.v., guide cannulae were surgically implanted into the right lateral

cerebral ventricle, and fixed with adhesive for repeated use, as previously described (McMillan

and Tyndale, 2015; Miksys et al, 2017; Zhou et al, 2013). Compounds (e.g. mechanism-based

inhibitors and dyes) injected i.c.v. into one lateral ventricle in rats have been shown to spread

bilaterally throughout all regions of the brain (DeVos and Miller, 2013; Miksys and Tyndale,

2009). Propafenone hydrochloride (Sigma-Aldrich), a CYP2D competitive inhibitor (Xu et al,

1995), was dissolved in 20 percent cyclodextrin (Sigma-Aldrich) in dH2O to deliver 40 μg base in

4 μl i.c.v. 30 minutes prior to oxycodone administration. Seven days of nicotine pre-treatment

induces rat brain, but not liver CYP2D, consistent with higher brain CYP2D levels found in human

smokers (Mann et al, 2008; Miksys et al, 2002; Yue et al, 2008); nicotine pre-treatment

increases in vivo activation of codeine to morphine and ex vivo dextromethorphan metabolism in

brain, but not liver, indicative of increased CYP2D activity (McMillan and Tyndale, 2015; Miksys

et al, 2017). Nicotine bitartrate (MP Biomedicals, Solon, OH, USA) was given subcutaneously

(s.c.) at 1 mg base/kg (in saline, 0.9 percent NaOH; pH 7) daily for 7 days, and oxycodone (or

oxymorphone) was given at 8 hours after the last nicotine injection, when induction of brain

Page 116: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

94

CYP2D is maximal and nicotine has been cleared from both the central and peripheral systems

(Yue et al, 2008).

Figure 25: Oxycodone and oxymorphone doses were chosen from dose–response curves

Figure 25: Oxycodone and oxymorphone doses were chosen from dose–response curves.

Oxycodone (OC) at 12.5 mg/kg p.o. (n = 13) and oxymorphone (OM) at 10 mg/kg p.o. (n = 10)

produced analgesic peaks of 62 and 70 percent, respectively, similar to the doses of codeine (COD,

20 mg/kg i.p.; 64 percent) and morphine (MOR, 3.5 mg/kg i.p.; 69 percent) used in a previous

study assessing the impact of brain CYP2D metabolism on COD metabolism and analgesia. The

COD and MOR i.p. dose–response data were previously published by (McMillan and Tyndale,

2015). %MPE, percentage of maximal possible effect; SEM, standard error of the mean.

Nociceptive testing of analgesia

Antinociception (denoted throughout as analgesia) was measured as the percentage of maximal

possible effect (%MPE = [postinjection latency − baseline latency]/[maximum cut-off − baseline

latency] × 100 percent) of tail-flick latency (TFL) using a tail-flick meter (Columbus Instruments,

Columbus, OH, USA), as previously described (McMillan and Tyndale, 2015). The thermal

strength was adjusted to obtain baseline TFLs between 3 and 4 seconds; a cut-off of 10 seconds

was used to avoid tissue damage. Individual baseline TFLs were the average of two measurements

prior to experimentation.

Page 117: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

95

Analgesia experimental design

Full analgesia paradigms, including pre-treatment timing, are outlined in Supplementary

(Figure 30). Food was restricted 1 hour prior to, and during, experimentation to decrease

variability in oral drug absorption. After oxycodone or oxymorphone administration, analgesia

was assessed over time, focussing on 0–60 minutes where oxycodone and oxymorphone analgesia

peak (TFL timepoints: 5, 10, 15, 20, 25, 30, 40, 50, 60, 75 and 90 minutes).

Microdialysis cannulation and procedure

Rats were surgically implanted with guide cannulae (MD-2250, Bioanalytical Systems, Inc., West

Lafayette, IN, USA) into the right nucleus accumbens shell (anteroposterior: +2.2 mm;

mediolateral: −1.0 mm; and dorsoventral −5.8 mm from Bregma (Nelson et al, 2011)), as

previously described (Garcia et al, 2016). Animals recovered for 7 days before beginning pre-

treatment, and were housed singly with access to food, restricted to 20–25 g/day to minimize

weight gain. A within-animal study design was used, where rats received each of four pre-

treatments (vehicle, propranolol, nicotine, or nicotine and propranolol, as previously described) in

randomized order (n = 9). Animals were connected to the microdialysis probes (concentric silica-

coated, 2-mm membrane; MD-2200, Bioanalytical Systems, Inc.) and allowed to acclimate for at

least 15 minutes while perfusion medium (Ringer's solution; 147 mM Na+, 2 mM Ca2+, 4 mM K+,

155 mM Cl−, pH 6.0) was pumped at a flow rate of 2 μl/min. Baseline dialysate was collected on

ice in three intervals of 15 minutes (45 minutes total) prior to oxycodone administration

(12.5 mg/kg p.o.), after which dialysate was collected in 15-minute intervals for 2 hours (eight

samples total). Microdialysis was performed in extracellular fluid as opioid receptors face the

extracellular compartment; therefore, extracellular opioid concentrations are relevant to functional

opioid response (Hammarlund-Udenaes et al, 2008).

Tissue collection, membrane preparation and ex vivo oxidation

Immediately following the microdialysis experiments, to confirm the impact of pre-treatments on

brain CYP2D activity, with no impact of liver CYP2D activity and plasma oxycodone levels,

animals were administered oxycodone and sacrificed at 30 minutes postinjection (approximately

peak analgesia). Trunk blood was centrifuged at 5000 g for plasma isolation. Liver microsomal

and brain cerebellum membranes were prepared as previously described (Miksys et al, 2017).

Page 118: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

96

Formation of dextrorphan from dextromethorphan was assessed as a measure of ex vivo CYP2D

activity, as previously described (Miksys et al, 2017; Tyndale et al, 1999).

Measurement of compounds

Oxycodone and its metabolites (eight total compounds) were measured by modified published

liquid chromatography–mass spectrometry methods (Andreassen et al, 2011; Lalovic et al, 2006);

full methodological details can be found in the Supplementary Information.

Statistical analyses

Data were analyzed with GraphPad Prism v.6.01 (GraphPad Software Inc., La Jolla, CA, USA),

by two-way ANOVA followed by either planned comparisons using one-way ANOVA (AUCs

and CYP2D ratios), independent samples t-tests (AUCs) or Tukey's post hoc testing adjusted for

multiple comparisons (%MPE at individual timepoints). Repeated measures were used in within-

animal experiments. Relationships between data sets were assessed by Pearson correlation

coefficients. Standard error of the mean (SEM) was displayed as unidirectional error bars for visual

simplification.

4.4 Results

Opioid analgesic dose–response was used to optimize oral oxycodone and oxymorphone

doses

Peak analgesia from a range of doses of oxycodone (5–15 mg/kg p.o.) and oxymorphone (2.5–

10 mg/kg p.o.) was compared with that after various codeine and morphine doses and

administration routes (Figure 25). The selected doses of oxycodone (12.5 mg/kg p.o.) and

oxymorphone (10 mg/kg p.o.) (Figure 25, white circled) provided submaximal peak analgesic

responses, allowing detection of both increases and decreases in analgesia, and compared with

peak analgesia from doses of codeine (20 mg/kg i.p.) and morphine (3.5 mg/kg i.p.) used in a prior

study assessing the impact of brain CYP2D on codeine response (Figure 25, black circled)

(McMillan and Tyndale, 2015). Of note, a 4.3-fold lower oral oxycodone dose (used in this study)

was required to produce similar peak analgesia relative to oral codeine (from Figure 25).

Page 119: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

97

Modifying brain CYP2D activity resulted in altered oxycodone response

Compared with vehicle, propranolol (CYP2D inhibitor) i.c.v. pre-treatment increased %MPE at

multiple timepoints (pre-treatment P = 0.044; Figure 26A) after oxycodone administration

(12.5 mg/kg p.o.), resulting in a 1.9-fold increase of the analgesic area under the analgesic-time

curve from 0 to 60 minutes (AUC0–60, P = 0.055; Figure 26B). A within-animal study design was

used; animals received i.c.v. vehicle and i.c.v. propranolol in randomized order, with a 2-week

washout between oxycodone (n = 15). In addition, brain CYP2D was inhibited using a

mechanistically (competitive versus mechanism-based inhibitor) and structurally distinct

compound, propafenone. Compared with vehicle pre-treatment, propafenone pre-treatment

increased %MPE at multiple timepoints (pre-treatment P = 0.003) after oxycodone administration

(12.5 mg/kg p.o.) resulting in a 3.7-fold increase of the analgesic AUC0–60 (P = 0.004; see

Supplementary, Figure 31), providing further evidence that inhibiting brain CYP2D increases

oxycodone analgesia (n = 7–8).

Page 120: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

98

Figure 26: Inhibiting brain CYP2D increased, and inducing brain CYP2D decreased, oxycodone analgesia

Figure 26: Inhibiting brain CYP2D increased, and inducing brain CYP2D decreased,

oxycodone analgesia. Compared with vehicle pre-treatment (V), propranolol pre-treatment (PL)

resulted in significantly increased (a) analgesia (%MPE) and (b) area under the analgesia-time

curve (AUC0–60) after oxycodone administration (12.5 mg/kg p.o., n = 15). Pre-treatment F(1,

14) = 4.89, P = 0.04. For AUC, t = 2.10, d.f. = 14, P = 0.05. Compared with vehicle pre-

treatment, nicotine pre-treatment (NIC) resulted in significantly decreased (a) %MPE and (b)

analgesic AUC0–60 after oxycodone administration (n = 17). Pre-treatment F(1,

16) = 12.76, P = 0.003. For AUC, t = 3.76, d.f. = 16, P = 0.002. Data are from two separate

studies, and the averaged vehicle arm is shown; statistics are based on the individual

experiments. *P < 0.05, **P < 0.01, ***P < 0.001 compared with vehicle. %MPE, percentage of

maximal possible effect; SEM, standard error of the mean.

Compared with vehicle, nicotine (CYP2D inducer) pre-treatment decreased %MPE at multiple

timepoints (pre-treatment P = 0.003; Figure 26A) after oxycodone administration (12.5 mg/kg

p.o.), resulting in a 1.6-fold decrease of the analgesic AUC0–60 (P = 0.002; Figure 26B). A within-

animal study design was used; animals received 7-day s.c. vehicle and 7-day s.c. nicotine in

randomized order, with a 2-week washout between oxycodone (n = 17). These data together

suggest that the parent compound oxycodone, rather than the more potent metabolite

oxymorphone, is responsible for oxycodone analgesia.

Page 121: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

99

Modifying brain CYP2D alters oxycodone, but not oxymorphone, analgesia

The impact of modifying brain CYP2D activity on oxycodone analgesia was replicated (Figures

26 and 27A); compared with vehicle pre-treatment, inhibiting brain CYP2D with propranolol

resulted in higher %MPE and analgesic AUC0–60 (P < 0.01), while inducing brain CYP2D with

nicotine resulted in a lower %MPE at 30 minutes, and lower analgesic AUC0–60 (P < 0.001)

compared with propranolol pre-treatment, after oxycodone administration (12.5 mg/kg p.o.).

Inhibiting the nicotine pre-treatment-induced brain CYP2D with i.c.v. propranolol yielded no

difference in %MPE or analgesic AUC0–60 (P = 0.47; Figure 27A) after oxycodone administration,

compared with vehicle (n = 10–13). Inhibition of the nicotine pre-treatment effect on oxycodone

analgesia suggests that propranolol and nicotine pre-treatment impact analgesia through a common

mechanism, i.e. modification of brain CYP2D activity.

Oxymorphone is the CYP2D metabolite of oxycodone and is not further metabolized by CYP2D;

pre-treatments that selectively inhibit or induce brain CYP2D should therefore not affect

oxymorphone analgesia. Compared with vehicle pre-treatment, propranolol pre-treatment,

nicotine pre-treatment and combined pre-treatment of nicotine and propranolol did not alter

%MPE (at any timepoints) or AUC0–60 (P > 0.6 for all pre-treatment groups; Figure 27B) after

oxymorphone administration (10 mg/kg p.o., n = 10–11). This suggests that the effect of pre-

treatments (propranolol and/or nicotine) on oxycodone analgesia was likely due to altered central

CYP2D-mediated metabolism of oxycodone rather than alternative analgesic or kinetic effects

(e.g. on BBB transport or opioid-receptor activation).

Baseline TFLs (in seconds) were assessed prior to experimentation, and there were no significant

differences across pre-treatment groups just prior to oxycodone (P = 0.21; vehicle = 3.6 + 0.1,

mean + SEM, propranolol = 3.2 + 0.2, nicotine = 3.2 + 0.1 and nicotine and

propranolol = 3.4 + 0.2) or oxymorphone (P = 0.88; vehicle = 4.5 + 0.4, propranolol = 4.3 + 0.3,

nicotine = 4.7 + 0.4 and nicotine and propranolol = 4.7 + 0.2) administration, as previously

observed (McMillan and Tyndale, 2015).

Page 122: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

100

Figure 27: Inhibiting induced brain CYP2D reversed the changes in oxycodone analgesia; variation in brain CYP2D activity did not affect oxymorphone analgesia

Figure 27: Inhibiting induced brain CYP2D reversed the changes in oxycodone analgesia;

variation in brain CYP2D activity did not affect oxymorphone analgesia. Compared with

vehicle pre-treatment (V), propranolol pre-treatment (PL) or nicotine pre-treatment (NIC) alone

resulted in significantly higher and lower, respectively, (a) %MPE and analgesic AUC0–60 after

oxycodone administration (12.5 mg/kg p.o., n = 10–13/group). Compared with vehicle, combined

nicotine and propranolol pre-treatment (NICPL) resulted in a significantly different %MPE and

AUC0–60 than nicotine or propranolol pre-treatment alone and no change in %MPE or AUC0–

60 from vehicle pre-treatment. Pre-treatment F(3, 42) = 6.90, P = 0.001. For AUCs, F(3,

42) = 7.53, P < 0.001. Compared with vehicle pre-treatment, a higher portion of animals pre-

treated with propranolol reached maximal analgesia, while no animals pre-treated with nicotine

reached maximal analgesia (a, table). Compared with vehicle pre-treatment, propranolol pre-

treatment, nicotine pre-treatment and co-pre-treatment of nicotine and propranolol did not result

in significantly different (b) %MPE or AUC0–60 after oxymorphone administration (10 mg/kg

p.o., n = 10–11/group). For analgesia curves, pre-treatment F(3, 34) = 0.21, P = 0.89. For

AUCs, F(3, 34) = 0.14, P = 0.94. A similar percentage of animals reached maximal oxymorphone

analgesia across all pre-treatment groups (b, table). *P <0.05, **P < 0.01, ***P < 0.001 compared

with vehicle and #P < 0.05, ##P < 0.01, ###P < 0.001 compared with nicotine and propranolol

combined pre-treatment. %MPE, percentage of maximal possible effect; SEM, standard error of

the mean.

Page 123: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

101

Brain oxycodone, not oxymorphone, levels are responsible for oxycodone analgesia

To investigate how the differences in oxycodone analgesia were related to changes in brain

oxycodone or oxymorphone levels in vivo, microdialysis was used to assess brain analyte levels

over time after oxycodone. After oxycodone administration (12.5 mg/kg p.o.) in vehicle pre-

treated animals, oxycodone and noroxycodone were the most abundant analytes, followed by

noroxymorphone and oxymorphone-glucuronide, and oxymorphone was the least abundant

(Figure 28A; curves for all analytes in the pre-treatment arms are in Supplementary Information,

Figure 32).

Compared with vehicle, propranolol pre-treatment significantly increased brain oxycodone levels

(P < 0.05; Figure 28C) after oxycodone administration, consistent with an increase in oxycodone

analgesia (Figure 28B). Nicotine pre-treatment increased brain oxymorphone levels (P < 0.001;

Figure 28G); this was not consistent with the decrease in oxycodone analgesia. Together these

brain analyte and time course data provide evidence to suggest that oxycodone, rather than the

more potent CYP2D metabolite oxymorphone, is responsible for oxycodone analgesia. Combined

nicotine and propranolol pre-treatment blocked the effect of propranolol on brain oxycodone levels

(P = 0.12; Figure 28I) and nicotine on brain oxymorphone levels (P = 0.56; Figure 28J), consistent

with the oxycodone analgesia data (Figure 28H).

Page 124: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

102

Figure 28: Manipulating brain CYP2D further suggests that brain oxycodone levels, not oxymorphone levels, are responsible for oxycodone analgesia

Figure 28: Manipulating brain CYP2D further suggests that brain oxycodone levels, not

oxymorphone levels, are responsible for oxycodone analgesia. Using in vivo brain

microdialysis, oxycodone (OC) and four other metabolites [noroxycodone (NOC), oxymorphone

(OM, on secondary axis), noroxymorphone (NOM) and oxymorphone-glucuronide (OMG)] could

be detected after oxycodone administration (12.5 mg/kg p.o., vehicle arm in a). Compared with

vehicle pre-treatment (V), propranolol pre-treatment (PL) increased (b) analgesia (from

Figure 27A) and (c) brain oxycodone levels (F(1, 15) = 5.20, P = 0.03), but had no effect on (d)

brain oxymorphone levels (F(1, 15) = 0.28, P = 0.60), after oxycodone administration. Nicotine

pre-treatment (NIC) decreased (e) analgesia (from Figure 27A) and increased (g) brain

oxymorphone levels (F(1, 13) = 15.85, P < 0.001) but had no effect on (f) brain oxycodone levels

(F(1, 13) = 1.52, P = 0.22), after oxycodone administration. Combined nicotine and propranolol

pre-treatment (NICPL) did not affect (h) analgesia (from Figure 27A), (i) brain oxycodone levels

(F(1, 15) = 2.54, P = 0.12) or (j) brain oxymorphone levels (F(1, 15) = 0.34, P = 0.56) after

oxycodone administration (n = 5–9/group). *P < 0.05, ***P < 0.001. %MPE, percentage of

maximal possible effect; SEM, standard error of the mean.

Page 125: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

103

Manipulating brain CYP2D altered brain, but not liver, oxycodone metabolism: oxycodone

analgesia correlated with brain CYP2D activity

Brain CYP2D metabolic ratios (metabolite oxymorphone level/ substrate, oxycodone

level; n = 5/9) from microdialysis data and plasma CYP2D metabolic ratios at 30 minutes post-

oxycodone (12.5 mg/kg p.o., n = 12) were calculated for each pre-treatment group. The overall

brain CYP2D metabolic ratio AUC0–60 (encompassing peak analgesia) was greater in nicotine pre-

treated animals (P < 0.05 against all groups) and different across pre-treatment groups (F(3,

24) = 4.34, P = 0.014). The brain CYP2D metabolic ratio from the microdialysis collection time

of 15–30 minutes was illustrated (Figure 29A) to contrast most closely with the plasma CYP2D

metabolic ratio from the plasma sampling time of 30 minutes. Compared with vehicle, propranolol

pre-treatment decreased the brain CYP2D metabolic ratio (n = 5–9; Figure 29A), but did not alter

the plasma CYP2D metabolic ratio (Figure 29C), and nicotine pre-treatment increased the brain

CYP2D metabolic ratio (P < 0.05 against all groups; Figure 29A), but did not alter the plasma

CYP2D metabolic ratio (Figure 29C). Combined nicotine and propranolol pre-treatment blocked

the effects of nicotine or propranolol pre-treatment alone, evidenced through no change in brain

or plasma CYP2D metabolic ratios (Figures 29A and C). Compared with vehicle

(7.82 + 0.38 nmol/g/h, mean + SEM), propranolol pre-treatment modestly decreased

(7.01 + 0.35 nmol/g/h), nicotine pre-treatment modestly increased (8.74 + 0.55 nmol/g/h) and

combined nicotine and propranolol pre-treatment resulted in no change in (7.91 + 0.55 nmol/g/h)

brain CYP2D activity (F(3, 82) = 1.37, P = 0.26). Hepatic CYP2D activity was unchanged across

all four pre-treatment groups (F(3, 44) = 0.02, P = 0.99). Analgesia (at 30 minutes, approximately

peak) negatively correlated with ex vivo brain CYP2D activity (n = 20–23; P = 0.002; Figure

29B), but not with hepatic CYP2D activity (n = 12; P = 0.42; Figure 29D). These findings further

indicate that manipulations of brain CYP2D metabolism alter brain, but not hepatic, oxycodone

metabolism and resulting analgesia.

Page 126: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

104

Figure 29: Manipulating brain CYP2D altered the CYP2D metabolic ratio in the brain; oxycodone analgesia was inversely correlated with brain CYP2D activity

Figure 29: Manipulating brain CYP2D altered the CYP2D metabolic ratio in the brain;

oxycodone analgesia was inversely correlated with brain CYP2D activity. (a) Compared with

vehicle pre-treatment (V), the brain CYP2D metabolic ratio (oxymorphone/oxycodone levels) was

slightly lower in animals given propranolol pre-treatment (PL), higher in animals given nicotine

pre-treatment (NIC) and unaltered in the combined propranolol and nicotine (NICPL) pre-

treatment group after oxycodone administration (12.5 mg/kg p.o., n = 5–9/group; F(3,

24) = 0.96, P = 0.43). (b) Oxycodone analgesia was significantly and inversely correlated with

brain CYP2D activity (P < 0.002) at 30 minutes (approximately peak) after oxycodone

administration (n = 20–23/group). (c) The plasma CYP2D metabolic ratio was similar across all

pre-treatment groups (n = 12/group; F(3, 67) = 0. 037, P = 0.99). (d) Oxycodone analgesia was

not correlated with hepatic CYP2D activity (P = 0.42) at 30 minutes after oxycodone

administration (n = 12/group). %MPE, percentage of maximal possible effect; SEM, standard

error of the mean.

4.5 Discussion

This study demonstrated an impact of brain CYP2D on central oxycodone metabolism to

oxymorphone and resulting analgesia. Inhibiting rat brain CYP2D increased oxycodone levels and

analgesia, while inducing rat brain CYP2D increased oxymorphone levels and decreased

analgesia; inhibiting the induced brain CYP2D blocked the decrease in oxycodone analgesia.

Together, this demonstrates that variation in brain CYP2D alters oxycodone brain levels and

Page 127: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

105

response. Pre-treatments modestly altered CYP2D metabolic ratios in brain, but not in plasma;

oxycodone analgesia was inversely correlated with ex vivo brain, but not liver, CYP2D activity.

Additionally, pre-treatments altering brain CYP2D activity did not affect baseline TFLs or

oxymorphone analgesia, indicating that the pre-treatments had no effect on (1) nociception, (2)

pathways mediating antinociception, (3) transport across the BBB and (4) peripheral CYP2D

activity. These findings provide strong evidence that brain oxycodone, rather than the more potent

μ-opioid receptor activating metabolite oxymorphone, is the primary contributor to oxycodone

analgesia and that levels of brain CYP2D influence oxycodone levels and response.

The role of brain CYP2D metabolism in oxycodone analgesia directly contrasts with its role in

codeine and tramadol analgesia. Like oxycodone, codeine and tramadol are metabolized by

CYP2D to more potent opioid-receptor agonists morphine and O-desmethyltramadol, respectively

(Chen et al, 1991b; Grond and Sablotzki, 2004). However, codeine and tramadol analgesia were

increased as a result of brain CYP2D metabolism increasing brain levels of the CYP2D

metabolites; codeine analgesia correlated with brain morphine levels, while tramadol analgesia

correlated with CSF O-desmethyltramadol levels (McMillan and Tyndale, 2015; Wang et al,

2015). Thus, brain CYP2D has opposing impacts on analgesia from different opioids; higher brain

CYP2D lowers oxycodone analgesia, likely through reduced brain oxycodone levels, but increases

codeine and tramadol analgesia, likely through higher levels of the active metabolites.

Despite an established role for hepatic CYP2D in metabolism of oxycodone, both pre-clinical and

human studies show little evidence for a role of hepatic CYP2D metabolism in oxycodone

analgesia. Inhibiting rat CYP2D (quinine) in vivo did not alter oxycodone analgesia (Cleary et al,

1994). Likewise, while inhibiting human CYP2D6 in vivo (quinidine or paroxetine) reduced

plasma oxymorphone AUC0–48 h (40 percent) and increased plasma noroxycodone AUC0–48 h (30

percent), it did not alter oxycodone plasma concentrations or analgesia (Gronlund et al, 2010).

Furthermore, inhibiting human CYP2D6 in vivo does not impact the analgesic or reinforcing

effects of other structurally related 6-keto-opioids hydrocodone and dihydrocodeine (Kaplan et al,

1997; Lelas et al, 1999; Schmidt et al, 2003; Webb et al, 2001). The finding here that brain

oxycodone, despite being a less potent μ-opioid receptor activator than oxymorphone, is primarily

responsible for analgesia is counterintuitive; the expectation being greater formation of the more

Page 128: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

106

potent oxymorphone should have increased analgesia. Oxycodone is a relatively potent μ-opioid

receptor agonist and readily crosses the BBB into the CNS, whereas oxymorphone has low

systemic levels and BBB permeability (1.6-fold lower brain-to-plasma ratio in rats) (Bostrom et

al, 2006; Klimas et al, 2013; Lalovic et al, 2006; Riley et al, 2008; Sadiq et al, 2013). The

differences in brain-to-plasma partitioning, sequestration, relative amounts and clearance between

oxycodone and oxymorphone may be sufficient to prevent changes in brain oxymorphone levels

from influencing oxycodone analgesia under these conditions (i.e. increased oxymorphone levels

when brain CYP2D is induced).

Our nicotine pre-treatment paradigm in rats, which induces brain CYP2D, results in average daily

nicotine plasma levels (25 ng/ml) comparable with smokers (20–50 ng/ml); smoking is associated

with elevated human brain CYP2D (Benowitz, 1999; Mann et al, 2008; Miksys et al, 2002; Yue

et al, 2008). Non-medicinal prescription opioid use is higher in smokers than non-smokers;

however, the isolated effects of smoking on acute oxycodone response are unclear (Skurtveit et al,

2010). In a study assessing the influence of smoking status on oxycodone response, smokers

experienced less oxycodone-induced nausea (Zacny et al, 2013), which is a common opioid-

receptor-mediated side effect of oxycodone treatment (He et al, 2016). Increased brain CYP2D

activity in smokers, increasing central metabolism of oxycodone and potentially reducing μ-opioid

receptor occupancy, could contribute to the decreased oxycodone-induced nausea experienced in

smokers. It is possible that decreases in opioid response and/or nausea alter the opioid-taking

experience and abuse potential of oxycodone among smokers.

The effect of smoking on oxycodone abuse liability is currently unknown. Theoretically, it is

difficult to discern whether inducing brain CYP2D might decrease the abuse potential of

oxycodone due to decreased acute effect or potentially increase the abuse potential due to

decreased adverse side effects. In a study assessing the reinforcing effects of oxycodone, 67

percent of the oxycodone drug abusers were smokers compared with 22 percent of the non-abusers

(Comer et al, 2010). Cigarette smoking is strongly associated with illicit drug use (Eckhardt et al,

1994); separating the effect of smoking on brain reward systems and on inducing brain CYP2D

requires alternative methods. Numerous pharmacological characteristics have been linked to

opioid abuse potential, such as onset and duration of action (Wightman et al, 2012). In this study,

Page 129: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

107

nicotine pre-treatment modestly delayed time to peak analgesia (30.5 + 1.30 minutes,

mean + SEM) compared with all other pre-treatments (vehicle: 27.5 + 1.7 minutes; propranolol:

24.2 + 1.8 minutes; and nicotine and propranolol: 26.8 + 1.4 minutes; F(3, 42) = 1.66, P = 0.19).

Thus, the subjective effects of a decreased and delayed peak oxycodone effect on altering the

rewarding properties and the transition to abuse remain to be directly investigated.

Oxycodone and metabolite levels were assessed by microdialysis in the cerebral ventricles of a

subset of animals and compared with levels in dialysate from the nucleus accumbens. There was

no difference in rank order of oxycodone and metabolites at any timepoint (P > 0.05) and no

difference in metabolite levels across (ANOVA of brain CYP2D ratio: F(1,9) = 0.016, P = 0.90),

suggesting drug levels in the nucleus accumbens shell and the cerebral ventricles were similar.

Therefore, the nucleus accumbens was used to enhance our ability in the future to pair brain opioid

levels with opioid-induced dopamine release. Only male rats were used in this study as previous

studies optimizing the pre-treatment paradigms were performed in male rats; exploration of male–

female sex differences in brain CYP2D activity and functional influences is currently being

planned. We used one behavioral assay for antinociception (tail flick) because of its reliability and

reproducibility (Grossman et al, 1982); future studies expanding these findings with additional

antinociception assays (e.g. hot plate), as well as additional outcomes of human clinical oxycodone

use (e.g. opioid-induced side effects, tolerance and dependence), would assist in understanding the

extent of the impact of central oxycodone metabolism.

This study provides further evidence of an impact of rat brain CYP2D on opioid metabolism and

response. Increasing (or decreasing) brain CYP2D activity and the consequent increase (or

decrease) in oxycodone metabolism decreased (or increased) oxycodone analgesia. Together with

microdialysis assessments, the results suggest that oxycodone is the active analgesic moiety in this

paradigm. Individuals with increased brain CYP2D activity (e.g. smokers and genetically rapid

CYP2D metabolizers) may experience lower oxycodone analgesia, while those with lower brain

CYP2D activity (e.g. non-smokers or those taking centrally acting CYP2D inhibitors) may

experience greater analgesia. This work also illustrates a contrasting role for brain CYP2D in

oxycodone versus codeine and tramadol analgesia. Hormones (e.g. testosterone and estrogen) and

developmental factors (e.g. age) can also influence brain CYP2D activity (Baum and Strobel,

Page 130: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

108

1997; Mann et al, 2012); the combined impact of genetic, environmental and hormonal factors on

brain CYP activity and on clinically relevant outcomes of altered CNS drug metabolism is still a

relatively unknown source of inter-individual variation in drug response. Investigation into

potential sources of individual variation in opioid response remains essential for identifying risk

factors for altered analgesic response and opioid abuse.

4.6 Acknowledgements

We would like to thank Dr. Bin Zhao for his invaluable technical assistance with the liquid

chromatography–mass spectrometry assay and Fariba Baghai Wadji for her expert support with

microdialysis animal procedures. This work was supported by a Canada Research Chair in

Pharmacogenomics; the Canadian Institutes of Health Research (grant numbers FDN 154294,

TMD 132557 and MOP 136937); the Campbell Family Mental Health Research Institute of the

Centre for Addiction and Mental Health (CAMH) and the CAMH Foundation; the Canada

Foundation for Innovation (grant numbers 20289 and 16014); and the Ontario Ministry of

Research and Innovation. Dr. Rachel F. Tyndale has consulted for Apotex on topics unrelated to

this work. Dr. Sharon Miksys and Douglas M. McMillan have no conflicts of interest to declare.

4.7 Supplementary Information

LC-MS quantification of oxycodone and analyte levels in biological samples (adapted from

(Andreassen et al, 2011; Lalovic et al, 2006))

Abbreviations

Oxycodone (OC), noroxycodone (NOC), oxymorphone (OM), noroxymorphone (NOM), 6-α-

oxycodol (OCOL), 6-α-oxymorphol (α-OMOL), 6-β-oxymorphol (β-OMOL), oxycodone N-

oxide (OCO) and oxymorphone-3-β-D-glucuronide (OMG) and their internal standards 6-α-

oxycodol-d3 (OCOL-d3), and noroxycodone-d3 (NOC-d3) were purchased from Toronto

Research Chemicals.

Page 131: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

109

Instrumentation

HPLC System:

HPLC system (Agilent 1260 LC system): Agilent 1260 Quaternary pump, Agilent 1260 Infinity

Standard Autosampler and temperature-controlled column compartment. OC, NOC, OM, NOM,

OCOL, α-OMOL, β-OMOL, OCO, OMG, OCOL-d3 and NOC-d3 were separated on an Agilent

ZORBAX SB-C18 column (15 cm x 2.1 mm I.D.; 5 µm particle size; Agilent Technologies, Palo

Alto, CA) at ambient temperature with a flow rate of 0.3 ml/min.

HPLC gradient:

Time (min) Flow (ml/min) 10mM Ammonium Acetate /

0.1 % Acetic Acid in dH2O (%)

Acetonitrile (%)

0 0.3 95 5

6.0 0.3 95 5

15.0 0.3 80 20

16.0 0.3 95 5

25.0 0.3 95 5

Mass spectrometry:

Instrument: Agilent 6430 Triple Quadrupole LC/MS system

Software: MassHunter software.

Acquisition Mode: multiple reaction monitoring (MRM)

Ionization: electrospray ionization (ESI)

Gas temperature: 350° C

Gas flow: 10 l/min

Nebulizer pressure: 35 psi

Capillary voltage: 3000 V

Page 132: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

110

Internal standards:

Working solution 20 ng/ml of OCOL-d3 and NOC-d3 prepared in water.

Analyte Transition Collision

Energy (eV) RT (min) IS used

OC 316.2 298.1 17 15.0 NOC-d3

NOC 302.1 284.1 13 14.6 NOC-d3

OM 302.1 284.1 17 4.8 NOC-d3

NOM 288.1 270.0 17 4.2 NOC-d3

OCOL 318.1 300.1 17 10.6 OCOL-d3

α-OMOL 304.1 286.1 21 3.2 OCOL-d3

β-OMOL 304.1 286.1 17 4.5 OCOL-d3

OCO 332.1 315.1 25 16.9 NOC-d3

OMG 478.1 284.1 29 1.9 NOC-d3

OCOL-d3 321.2 303.2 17 10.3 -

NOC-d3 305.2 287.1 13 14.3 -

Sample Preparation

Microdialysis

Dialysis samples (30 µl) were mixed with internal standards (10 µl of 20 ng/ml deuterated

metabolites in dH2O). Samples were centrifuged at 12,000 rpm for 10 min, then 40 µl was injected

into the LCMS system. LOQ = 0.1ng/ml for OC, NOC, OM, NOM, OCOL, α-OMOL, β-OMOL,

OCO and OMG (30ul dialysis sample used). The standard curve was calibrated with linear function

and 1/x weighing. Coefficient of correlation should be > 0.99. The calibration curve was prepared

in the same biological matrix as the samples (Ringer’s solution) with known concentrations from

a working solution of all analytes assessed; calibration curve range: 0.1, 0.2, 0.5, 1, and 10 ng/ml.

Plasma and Brain Homogenate

Internal standards (20 µl of 20 ng/ml deuterated metabolites in dH2O) were added to 100 µl of

sample, and protein was precipitated with acetonitrile (0.9 ml). Samples were vortexed for 30

seconds, centrifuged at 12,000 rpm for 10 min, the supernatant evaporated to dryness under

nitrogen at 38°C, reconstituted with 100 µl of mobile phase (ammonium acetate buffer /

Page 133: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

111

acetonitrile, 95 / 5 %), and injected (60 µl) into the LCMS system. LOQ: 0.2ng/ml (100ul used)

for OC, NOC, OM, OCO, OMG, OCOL, 6-alpha-OMOL, 6-beta-OMOL; 0.5ng/ml for NOM

(100ul used). The standard curve was calibrated with linear function and 1/x weighing. Coefficient

of correlation should be > 0.99. The calibration curve was prepared in the same biological matrix

as the samples (control plasma) with known concentrations from a working solution of all analytes

assessed; calibration curve range: 0.2, 0.5, 1, 10, 100, and 500 ng/ml.

Figure 30: Study paradigm of pretreatment timing prior to oxycodone or oxymorphone administration

Figure 30: Study paradigm of pretreatment timing prior to oxycodone or oxymorphone

administration. Nicotine pretreatment is given subcutaneously (1 mg/kg, s.c.) once daily for

seven days; oxycodone or oxymorphone are given at 8 hr after the last nicotine pretreatment.

Propranolol pretreatment is by a single intracerebroventricular injection (20 μg in 4ul cyclodextrin,

i.c.v.) 24 hr prior to oxycodone or oxymorphone administration.

Page 134: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

112

Figure 31: Inhibiting brain CYP2D with the competitive inhibitor propafenone increased oxycodone analgesia

Figure 31: Inhibiting brain CYP2D with the competitive inhibitor propafenone increased

oxycodone analgesia. Compared with vehicle pretreatment, propafenone pretreatment (brain

CYP2D inhibitor, 30 min prior) resulted in significantly increased %MPE (A; F (1, 13) = 13.32, p

= 0.003) and AUC0-60 min (B; un-paired t = 3.51, df = 13, p = 0.004) after oxycodone administration

(12.5 mg/kg p.o., n=7-8/group). The weighted average of the vehicle groups (including vehicle

groups from Figure 26) was calculated and shown for illustration of the comparisons with those in

Figure 26; statistics shown here are based on this individual experiment. **p<0.01, ***p<0.001.

Page 135: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

113

Figure 32: Full microdialysis analyte curves for each pretreatment group after oxycodone administration

Figure 32: Full microdialysis analyte curves for each pretreatment group after oxycodone

administration. Drug levels by time are displayed for vehicle pretreatment (A), propranolol

pretreatment (B), nicotine pretreatment (C) and combined nicotine and propranolol pretreatment

(D). Oxycodone (OC) and its three primary metabolites noroxycodone (NOC), oxymorphone

(OM, on secondary axis), and noroxymorphone (NOM), along with the additional metabolite

oxymorphone-glucuronide (OMG) could be detected in all four pretreatment groups after

oxycodone administration (12.5 mg/kg, p.o., n=5-9/group).

Page 136: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

114

4.8 Significance to Thesis

This study provides further evidence of an impact of brain CYP2D on opioid metabolism and

response, building from the prior two studies by using a distinct opioid compound oxycodone.

Inhibiting rat brain CYP2D increased analgesia, while inducing rat brain CYP2D decreased

analgesia after oral oxycodone administration. Furthermore, using in vivo brain microdialysis

optimized to detect brain levels of oxycodone and its primary metabolites over time, we found that

inhibiting brain CYP2D increased brain oxycodone levels while inducing brain CYP2D increased

brain oxymorphone (CYP2D-metabolite of oxycodone) levels in vivo. While oxycodone, like

codeine, is metabolized by CYP2D to a more potent opioid receptor-activating metabolite

oxymorphone, our in vivo brain microdialysis data indicated that the parent compound oxycodone

is predominantly responsible for oxycodone-induced analgesia. This is in contrast to our findings

on the impact of brain CYP2D metabolism on codeine response, where the CYP2D-metabolite

morphine was primarily responsible for codeine-induced analgesia, suggesting different roles for

brain CYP2D in response to these two opioids.

Findings from this chapter suggest that individuals with high brain CYP2D activity (e.g. smokers,

individuals taking nicotine) may exhibit a lower response to oxycodone due to increased central

metabolism of oxycodone to oxymorphone. Human studies have found no contribution of CYP2D6

genotype to overall oxycodone effect, therefore this study provides a potential novel source of

interindividual variation in oxycodone response. The findings outlined in this chapter support

future work on elucidating the impact of brain CYP2D metabolism on additional substrate opioid

compounds such as hydrocodone.

Page 137: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

115

5. General Discussion

5.1 Summary of findings

5.1.1 Brain CYP2D and codeine

The studies in this thesis demonstrated a novel impact of brain CYP2D on the central metabolism

of opioids and resulting analgesia in vivo, after acute and repeated opioid administration. In

chapter 1, rats were administered intraperitoneal doses of codeine and morphine adjusted to

produce similar plasma morphine levels at 15 min post-injection, the time of peak codeine

analgesia; animals administered codeine had higher brain morphine levels and analgesia than those

administered morphine, consistent with brain CYP2D metabolism of codeine. We showed that

inhibiting CYP2D specifically in the brain with direct intracerebroventricular injection of

mechanism-based, and competitive, CYP2D inhibitors decreased brain morphine levels and

analgesia after intraperitoneal codeine administration. We further showed that inducing brain

CYP2D with short-term nicotine pretreatment increased brain morphine levels and analgesia after

codeine, which was reversed by inhibiting brain CYP2D. Inhibiting and inducing brain CYP2D

did not alter morphine-induced analgesia, indicating the selectivity of pretreatment effect on

codeine analgesia was through brain CYP2D alone. Codeine analgesia correlated with brain, but

not plasma, morphine levels, consistent with brain CYP2D-mediated activation of codeine to

morphine primarily mediating codeine analgesic response (Figure 33).

In chapter 2, we investigated the role of brain CYP2D metabolism in response to chronic drug

administration. Specifically, we assessed whether inducing brain CYP2D, and thereby increasing

the brain CYP2D activation of codeine to morphine, would impact the rate of onset of codeine

analgesic tolerance during daily oral codeine administration. We found that inducing brain CYP2D

with short-term nicotine pretreatment increased acute codeine analgesia (replicating our findings

in chapter 1) and the rate of codeine tolerance, quantified as a greater decrease (greater slope) in

peak analgesia (30 minutes post-injection) across 7 days of oral codeine administration. Consistent

with morphine not being a CYP2D substrate, inducing brain CYP2D did not alter the rate of

tolerance to analgesia from daily oral morphine administration. The rate of tolerance to codeine

correlated with acute analgesia, which was increased by inducing brain CYP2D, while the rate of

tolerance to morphine also correlated with acute analgesia, which was unaffected by brain CYP2D.

Page 138: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

116

The same relationship was seen when acute analgesia was increased by administering higher doses

of codeine and morphine, further indicating acute analgesia was predominantly responsible for the

rate of tolerance. Individuals who experience a greater acute codeine analgesic effect, either by

taking a high codeine dose or by having higher brain CYP2D activity (e.g. due to genetics or

exposure to xenobiotic inducers), may experience a greater absolute decrease, but similar

proportional decrease, in codeine analgesia across repeated doses, than those with a lower acute

analgesic effect. The potential clinical outcome of this phenomenon is unknown, but is explored

further in discussion section 5.3.

Figure 33: Proposed model for the functional role of rat brain CYP2D in the activation of codeine to morphine in vivo

Figure 33: Proposed model for the functional role of rat brain CYP2D in the activation of

codeine to morphine in vivo. A) While codeine readily enters the CNS, morphine has difficulty

crossing the blood-brain barrier (BBB) and is actively effluxed. B) Inhibiting CYP2D specifically

in the brain with the mechanism-based inhibitor propranolol (PL, metabolized by CYP2D to

hydroxy-propranolol [OH-PL] which binds irreversibly to the active site of CYP2D rendering it

inactive) decreased brain morphine formation from codeine, and resulting codeine analgesia. C)

In contrast, inducing brain CYP2D with short-term nicotine (NIC) pretreatment increased brain

morphine formation from codeine and resulting codeine analgesia.

5.1.2 Brain CYP2D and oxycodone

In chapter 3, we extended our findings on brain CYP2D metabolism to oxycodone, an opioid

compound with a pharmacologic profile distinct from codeine. We were initially unsure if greater

brain CYP2D metabolism of oxycodone would increase oxycodone analgesic response as, like

codeine, oxycodone is metabolized by CYP2D to a more potent MOR-activating compound,

oxymorphone. However, inhibiting brain CYP2D with propranolol increased analgesia and brain

Page 139: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

117

oxycodone levels, measured by microdialysis after oral oxycodone administration, suggesting that

the parent compound was responsible for oxycodone analgesia. Inducing brain CYP2D with short-

term nicotine pretreatment increased brain oxymorphone levels from oral oxycodone, but

decreased analgesia, consistent with brain oxymorphone levels not contributing to oxycodone

analgesia under this paradigm. The effect of inducing brain CYP2D on oxycodone analgesia and

brain oxymorphone levels was blocked by inhibiting brain CYP2D. Similar to the morphine

control used in chapters 1 and 2, we found that oxymorphone analgesia (not a CYP2D substrate)

was not altered by the pretreatments used to increase and decrease brain CYP2D activity,

suggesting the effects of pretreatment on oxycodone analgesia and drug levels were due to changes

in brain CYP2D activity alone. Oxycodone analgesia negatively correlated with ex vivo brain, but

not liver, CYP2D activity consistent with oxycodone being primarily responsible for oxycodone

analgesia. Due to BBB permeability differences between oxycodone and oxymorphone, the brain

CYP2D metabolism of oxycodone may function to increase clearance of oxycodone from the CNS

(Figure 34); this concept is further explored in discussion section 5.2.

Figure 34: Proposed model for the functional role of rat brain CYP2D in the metabolism of oxycodone to oxymorphone in vivo

Figure 34: Proposed model for the functional role of rat brain CYP2D in the metabolism of

oxycodone to oxymorphone in vivo. A) Oxycodone partitions from plasma across the blood-

brain barrier (BBB) into the CNS, while oxymorphone is produced in low quantities peripherally

and centrally. B) Inhibiting CYP2D specifically in the brain with the mechanism-based inhibitor

propranolol (PL, metabolized by CYP2D to hydroxy-propranolol [OH-PL] which binds

irreversibly to the active site of CYP2D rendering it inactive) increased brain oxycodone levels,

and resulting oxycodone analgesia. C) In contrast, inducing brain CYP2D with nicotine (NIC)

increased brain oxymorphone from oxycodone, and decreased oxycodone analgesia.

Page 140: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

118

5.2 Predicting the contribution of opioid parent and CYP2D metabolite to drug effect

5.2.1 Pharmacokinetics, pharmacodynamics and blood-brain barrier transport

The studies in this thesis have focused on elucidating the relative contribution of parent (codeine

and oxycodone) and brain CYP2D-mediated metabolite (morphine and oxymorphone) to opioid

drug effect (analgesia and tolerance). This is predominantly dependent on the pharmacokinetic and

pharmacodynamic properties of each compound, including the proportion of parent opioid

compound metabolized by CYP2D, the metabolite production in the periphery (primarily liver)

relative to the brain, the influx across the BBB relative to efflux from the CNS, and target receptor

affinity (Smith, 2009, 2011). These variables differ between opioid compounds, and account for

differences in the impact of brain CYP2D metabolism on opioid effect, such as that seen in

chapters 1 and 3.

5.2.1.1 Codeine

The relative contribution of codeine and the CYP2D-mediated metabolite morphine to codeine-

induced analgesia is fairly straightforward. Morphine is a relatively minor metabolite of codeine,

representing 0–15% of a codeine dose, while 80% of codeine is metabolized by UGT2B7 to

codeine-6-glucuronide, and the remainder by CYP3A4 to norcodeine (Chen et al, 1991b; Smith,

2011). The affinity of morphine for the MOR is approximately 3000-fold greater than codeine and

morphine is therefore primarily responsible for codeine analgesia (Smith, 2011). There are

established differences in the amount of morphine produced from codeine and resulting codeine

analgesia between individuals given CYP2D6 inhibitors (e.g. quinidine) and vehicle, and between

individuals with different CYP2D6 genotypes (Sindrup et al, 1992; Sindrup et al, 1990). Our

findings in chapter 1 suggest that while morphine formed from codeine is responsible for codeine

analgesia, distinct properties of the two compounds, like BBB permeability, may reduce the

contribution of peripherally formed morphine (via hepatic CYP2D) to analgesia, which is a

centrally mediated effect.

There are marked differences in the ability of codeine and morphine to cross the BBB. This was

shown using in vivo brain microdialysis in rats infused intravenously with codeine or morphine,

where codeine rapidly entered into brain extracellular fluid, and exhibited identical influx and

Page 141: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

119

efflux clearances, producing an unbound brain-to-blood AUC ratio of 1.00 ± 0.18 (Xie and

Hammarlund-Udenaes, 1998). In comparison, morphine produced a 3.6-fold lower brain-to-blood

AUC ratio (0.28 ± 0.09) than codeine, and a net efflux clearance (26 µl/min*g-brain), indicating

both poor BBB permeability and active efflux (Bouw et al, 2000; Xie and Hammarlund-Udenaes,

1998). Consistent with this, in chapter 1, there was a correlation between plasma and brain

codeine, but not morphine, levels at 15 min post-codeine injection, indicating that codeine, but not

morphine, readily equilibrates across the BBB.

Comparing compartment-specific half-lives (t1/2) in rats infused with codeine or morphine, codeine

displayed a similar t1/2 in brain (26 min) and blood (24 min), while morphine had a significantly

longer t1/2 in brain (44 min) than in blood (30 min) suggesting potential redistribution within brain

tissue (Bouw et al, 2000). In chapter 1, when rats were given intraperitoneal doses of codeine (20

mg/kg) or morphine (3.5 mg/kg), producing similar plasma morphine levels at 15 min post-

injection, codeine analgesia peaked at approximately 15 min and dropped below 50% (MPE) by

30 min, while morphine analgesia peaked at 30 min post-injection and dropped below 50% at 60

min. The differences in compartment-specific half-lives, along with the slow BBB influx into the

CNS, could account for the delay in onset and greater duration of morphine analgesia compared

to codeine. Rats given codeine had approximately 6-fold greater brain morphine levels at the time

of peak codeine analgesia than rats given morphine. This suggests that central morphine levels and

analgesia at this time were likely derived from codeine (rapidly influxed) via central CYP2D-

mediated activation to morphine, rather than from peripheral morphine (slowly influxed) produced

via hepatic CYP2D.

Furthermore, efflux transporters at the BBB (e.g. P-glycoprotein [P-gp]) can function as

determinants of the bioavailability of opioids like codeine and morphine (Thompson et al, 2000;

Wandel et al, 2002). Codeine and morphine are both substrates of P-gp, evidenced by their ability

to increase P-gp mediated ATP hydrolysis in vitro (Cunningham et al, 2008). However, due to the

ability of codeine to passively permeate across the BBB, P-gp mediated active transport has no

effect on its distribution across the BBB (Cunningham et al, 2008). P-gp mediated efflux of

morphine is important in reducing morphine analgesic effect; P-gp knockout mice had greater

analgesia from morphine, and individuals homozygous for a variant SNP in the gene encoding P-

Page 142: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

120

gp (ATP-binding cassette B1 gene) had greater pain relief from morphine (Campa et al, 2008;

Thompson et al, 2000). Differences in P-gp efflux at the BBB may contribute to differences in

efflux of codeine and morphine from the CNS, and provide additional understanding for the

findings in chapters 1 and 2. The low BBB permeability of peripherally-formed morphine, as

well as active efflux via P-gp, suggest that morphine present in the brain shortly after codeine

administration is due to central metabolism of codeine. This explains, in part, why inhibiting and/or

inducing the brain CYP2D activation of codeine to morphine altered brain morphine levels shortly

after codeine administration and corresponding acute codeine analgesia.

Altering BBB transport was a potential confound in the studies included in this thesis, specifically

with regards to the pretreatments used to modify brain CYP2D activity. Studies assessing the

impact of nicotine and propranolol on P-gp function are conflicting. Nicotine, and its metabolite

cotinine, did not alter P-gp mediated ATPase activity in vitro, suggesting no interaction with P-gp

transport; however, nicotine treatment of rats for 14 days by minipump increased saquinavir (a

protease inhibitor and P-gp substrate) penetration into the brain (Manda et al, 2010; Wang et al,

2005b). Propranolol dose-dependently decreased digoxin transport in monolayers of Caco-2

(human colon carcinoma) cells, indicative of P-gp inhibition (Bachmakov et al, 2006). However,

our findings suggest that nicotine and propranolol, given in the pretreatment paradigms employed

in chapters 1 and 2, did not alter BBB transport due to 1) the timing of pretreatment (nicotine and

propranolol were administered 8 and 24 hours, respectively, prior to codeine and morphine), and

2) a lack of effect of nicotine and propranolol pretreatment on morphine analgesia.

5.2.1.2 Oxycodone

Compared to codeine and morphine, the contribution of brain CYP2D metabolism to oxycodone-

induced analgesia is more complex. Oxycodone is metabolized extensively by CYPs, primarily by

CYP3A4 into noroxycodone (45–47% of an oxycodone dose) and CYP2D6 into oxymorphone

(11%), which are further metabolized by CYP2D6 and CYP3A4, respectively, into

noroxymorphone (14%) (Lalovic et al, 2006; Lalovic et al, 2004). Similar to other opioids, the

CYP2D6-mediated metabolism of oxycodone produces a compound with greater affinity for the

MOR (Lalovic et al, 2006). Using radioligand displacement studies with membrane-expressed

ORs (from Chinese hamster ovary (CHO-K1) cells), oxymorphone displayed a greater

Page 143: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

121

displacement Ki than oxycodone for the MOR (0.36 nmol/L, compared to 16.0), KOR (148 nmol/L,

compared to >1000), and DOR (118 nmol/L, compared to >1000) (Klimas et al, 2013; Lalovic et

al, 2006). The affinity of oxymorphone for MOR specifically, compared to oxycodone,

noroxycodone, and noroxymorphone, respectively, was 44-, 158-, and 16-fold greater, while the

EC50 of oxymorphone-stimulated GTPγS binding to MOR Gα proteins was 8-, 45-, and 4-fold

greater (Klimas et al, 2013; Lalovic et al, 2006).

Based on receptor activity alone, our initial hypothesis in chapter 3, as with chapter 1, was that

increasing the brain CYP2D-metabolism of oxycodone to oxymorphone would increase

oxymorphone levels from oxycodone and increase analgesia. However, preclinical and human

studies show little evidence for a role of hepatic CYP2D metabolism of oxycodone to

oxymorphone in analgesia. Inhibiting rat CYP2D with i.p. quinine did not alter oxycodone

analgesia, while inhibiting human CYP2D6 with oral quinidine or paroxetine reduced plasma

oxymorphone levels from oxycodone but did not alter oxycodone analgesia (Cleary et al, 1994;

Gronlund et al, 2010). In an analysis of studies assessing oxycodone metabolism in vivo, mean

oxycodone levels were repeatedly calculated as the primary contributor to analgesia after oral

(83.0% of analgesia due to oxycodone) or intravenous (95.3%) oxycodone administration, as well

as with inhibition of the CYP3A4 (71.43%) or CYP2D6 (93.7%) metabolic pathways (Klimas et

al, 2013). This may be due, in part, to oxycodone being a relatively potent MOR agonist (5-fold

less than morphine) therefore, unlike codeine, oxycodone does not require CYP2D activation to

elicit analgesia (Klimas et al, 2013; Lalovic et al, 2006). In chapter 3, in contrast to codeine-

induced analgesia in chapter 1, inhibiting brain CYP2D increased brain oxycodone levels and

corresponding oxycodone-induced analgesia, while inducing brain CYP2D levels increased brain

oxymorphone levels from oxycodone and decreased analgesia.

Oxycodone readily crosses the BBB into the CNS while oxymorphone has low BBB permeability

(Bostrom et al, 2006; Klimas et al, 2013; Lalovic et al, 2006; Riley et al, 2008). Using in vivo

brain microdialysis and pharmacokinetic-pharmacodynamic modelling in rats, there was a 1.6-fold

smaller unbound brain-to-blood ratio of oxymorphone after intravenous oxymorphone (1.90), than

of oxycodone after intravenous oxycodone (3.01) (Bostrom et al, 2006; Sadiq et al, 2013). Also,

the influx clearance (CLin) of oxymorphone was 26.5-fold smaller than oxycodone (72 µl/min*g

Page 144: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

122

compared to 1910 µl/min*g) (Bostrom et al, 2006; Sadiq et al, 2013; Tunblad et al, 2003). While

oxymorphone may be actively influxed into the CNS (due to a high influx clearance and unbound

brain-to-blood ratio), this appears to occur at a significantly slower rate than for oxycodone.

Oxymorphone concentrations produced by peripheral metabolism of oxycodone are very low in

humans; healthy volunteers given 20 mg of oxycodone produced peak oxymorphone plasma

concentrations of 0.82 ng/ml, compared to 23.2 ng/ml of oxycodone (Kaiko et al, 1996).

Extrapolating from this dose in plasma using values for protein binding and the unbound brain-to-

blood ratios from literature yields an estimated brain concentration of 1.4 ng/ml oxymorphone,

compared to 38 ng/ml oxycodone, which is much lower than an estimated oxymorphone EC50 of

63 ng/ml in brain (Bostrom et al, 2006; Kaiko et al, 1996; Sadiq et al, 2013). While this explains

why oxymorphone produced peripherally does not contribute to oxycodone analgesia, it fails to

explain why increasing oxymorphone produced centrally (i.e. inducing brain CYP2D) results in a

decrease in oxycodone analgesia; therefore, additional considerations are required.

Differences in BBB efflux may contribute to the mechanism by which increased brain

oxymorphone production, via inducing brain CYP2D, resulted in decreased oxycodone analgesia.

If oxymorphone were effluxed more rapidly than oxycodone, then increasing the amount of

oxycodone metabolized to oxymorphone through the CYP2D pathway may expedite the removal

of oxycodone from the CNS by efflux. Through pharmacokinetic-pharmacodynamic modelling in

rats, oxycodone exhibited an efflux clearance (CLout) 16-fold greater than oxymorphone (630

µl/min*g compared to 39 µl/min*g) (Bostrom et al, 2006; Sadiq et al, 2013). However, this is

likely due to much higher brain levels of oxycodone than oxymorphone (influxed and/or produced

centrally), as oxycodone exhibits a net influx clearance 17-fold greater than oxymorphone (560

µl/min*g compared to 33 µl/min*g) (Bostrom et al, 2006; Sadiq et al, 2013). In rats infused with

the P-gp inhibitor PSC833, there was no difference in brain oxycodone levels, plasma oxycodone

pharmacokinetics, or oxycodone analgesia, compared to vehicle controls, indicating oxycodone is

not a P-gp substrate (Bostrom et al, 2005). Similarly, oxymorphone did not alter in vitro P-gp

mediated ATPase activity, indicating that it may be neither a P-gp substrate nor an inhibitor

(Metcalf et al, 2014). Therefore, while there do not appear to be major differences in the CNS

influx or efflux between oxycodone and oxymorphone, the net differences in brain/plasma

partitioning and clearance may be sufficient to negate the effect of changes in brain oxymorphone

Page 145: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

123

levels on oxycodone analgesia under these conditions. Any impact of P-gp on oxymorphone efflux

in vivo could be tested by inducing brain CYP2D concurrently with inhibition of P-gp (e.g. with

infusion of the P-gp inhibitor PSC833), and assessing the impact on oxycodone analgesia.

Although in vitro studies suggest oxymorphone is not a substrate of P-gp, if inducing brain CYP2D

metabolism of oxycodone to oxymorphone in vivo decreases oxycodone analgesia due to more

rapid efflux of oxymorphone, this should be blocked by P-gp inhibition.

There are numerous other factors that may account for changes in central oxymorphone levels not

contributing to oxycodone analgesia. While oxycodone acts primarily on MORs, oxymorphone

shows high affinity for MORs, KORs, and DORS (Lalovic et al, 2006). Agonists of DOR and

KOR can increase MOR-agonist response in vivo; intracerebroventricular injection of the DOR-

agonist DPDPE increased morphine-induced EEG bursts in rats, while repeated subcutaneous

administration of the KOR-agonist (-)U-50,488H increased MOR activity in mice (Narita et al,

2003; Stamidis and Young, 1992). Therefore, oxymorphone activation of KORs and DORs could

increase oxycodone activation of MORs, contributing to oxycodone’s predominant role in

analgesia. However, after brain CYP2D induction, increased brain oxymorphone levels from

oxycodone did not increase oxycodone-induced analgesia, suggesting that under these conditions,

KOR and DOR activation by oxymorphone is unlikely to play a role. Differences in short-term

MOR regulatory events, like desensitization of receptors, may also contribute to agonist-specific

differences in response (Whistler et al, 1999). In rat locus coeruleus neurons in vitro, oxymorphone

evoked a large change in potassium channel current (348 pA) but rapid desensitization (to 69% of

the peak), whereas oxycodone evoked a current (278 pA) similar to morphine but without

desensitization (<1% of peak) (Virk and Williams, 2008). In chapter 3, increased brain

oxymorphone levels from induced brain CYP2D metabolism of oxycodone may have resulted in

rapid MOR desensitization, and thereby decreased the ability of oxycodone to activate, and thereby

produce analgesia at, MORs.

We do not know the precise reason that inhibiting brain CYP2D increased oxycodone levels,

without altering oxymorphone levels, while inducing brain CYP2D increased oxymorphone levels,

without altering oxycodone levels. Our behavioural findings suggest that oxycodone (brain levels

increased with increased analgesia), but not oxymorphone (brain levels increased with decreased

Page 146: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

124

analgesia), was predominantly responsible for oxycodone-induced analgesia. However, after

inducing brain CYP2D, we observed no corresponding decrease in brain oxycodone levels with

decreased analgesia. Difficulties in compound detection may explain some of this discrepancy.

Oxymorphone levels were below the limit of quantitation (0.1 ng/ml) in samples collected from 0

– 60 min after oxycodone administration in 78% of vehicle pretreated animals (7/9), 86% of

propranolol pretreated animals (6/7), and 71% of nicotine and propranolol pretreated animals (5/7).

In comparison, oxymorphone was detected in all animals pretreated with nicotine (5/5), further

strengthening the conclusion that nicotine pretreatment induced the brain CYP2D metabolism of

oxycodone to oxymorphone. However, this fails to explain why there was no observed decrease

in brain oxycodone levels after inducing brain CYP2D. One potential explanation is that the

oxycodone (or oxymorphone) levels collected in nucleus accumbens did not reflect those in

regions mediating analgesia. The relative levels of oxycodone and oxymorphone and other

metabolites sampled from the nucleus accumbens did not differ from those sampled from the

cerebral ventricles, suggesting that oxycodone and its metabolite levels were homogenous across

brain regions. However, rat brain stem, which contains regions involved in opioid-modulation of

pain (or antinociception) transmission (e.g. PAG, RVM, dorsal root ganglia), expresses roughly

2.1-fold more CYP2D protein than rat striatum, which includes the nucleus accumbens (Ossipov

et al, 2010; Yue et al, 2008). More CYP2D protein in the brain stem may have produced an isolated

microenvironment of higher brain CYP2D activity, and therefore oxycodone metabolism,

accompanied by changes in oxycodone levels that were not detected in the nucleus accumbens or

cerebral ventricles.

5.2.2 The potential impact of brain CYP2D on other opioids

The opposing impacts of brain CYP2D on codeine (by formation of active metabolite) and

oxycodone (by removal of active parent) analgesia in the two acute administration models in this

thesis provide a basis for predicting the role of brain CYP2D in analgesia from other opioids. For

example, the opioid tramadol is metabolized primarily by CYP2D6 to O-desmethyltramadol (O-

DSMT, 50% of urinary recovery), which has a 300-fold greater affinity for the MOR than the

parent tramadol and is therefore believed to be responsible for tramadol’s analgesic effects (Grond

and Sablotzki, 2004; Wang et al, 2015). Consistent with this hypothesis, Malaysian pain-patients

genotyped as CYP2D6 UM and EMs had greater tramadol clearance (to O-DSMT) and MOR-

Page 147: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

125

related adverse-effect profiles than IMs (Gan et al, 2007). Furthermore, at 10 min after oral

tramadol administration in rats (time of approximate peak brain tramadol levels), the ratio of

tramadol/O-DSMT was 10 to 30-fold greater in brain than plasma, suggesting that, compared to

tramadol, O-DSMT has difficulty crossing the BBB (Tao et al, 2002). Tramadol, but not O-DSMT,

levels in rat correlated between cerebrospinal fluid and plasma after intraperitoneal tramadol

administration (Wang et al, 2015). The difference in MOR affinity between parent and metabolite,

the role of CYP2D metabolism, and the apparent differences in BBB permeability, suggest that

central CYP2D-mediated metabolism of tramadol to O-DSMT likely functions as an activation

pathway eliciting analgesia. This was assessed in vivo in rats; inhibiting brain CYP2D increased

tramadol t1/2 in brain and decreased tramadol analgesia and brain O-DSMT levels after

intraperitoneal tramadol administration, while inducing brain CYP2D decreased tramadol t1/2 in

brain and increased tramadol analgesia and brain O-DSMT levels (Wang et al, 2015). Thus, it

appears that the role of brain CYP2D in analgesia is similar for tramadol and codeine, functioning

as an activation pathway.

Hydrocodone, a semi-synthetic opioid compound with structural similarities to oxycodone, is

metabolized primarily through 6-keto-reduction to dihydrocodeine, but also through CYP2D6 to

hydromorphone (0-5% of urinary recovery), a compound with 33-fold greater MOR affinity than

hydrocodone (Chen et al, 1991a; Cone et al, 2013; Smith, 2011). Nevertheless, the influence of

CYP2D6 metabolism (e.g. genetic variation in CYPD6 activity) on hydrocodone response is

unclear. In two independent clinical cases, patients genotyped as CYP2D6 PMs were able to

achieve analgesia from hydrocodone, but not codeine, suggesting that the CYP2D6 pathway

influences analgesia from hydrocodone and codeine differently (Foster et al, 2007; Susce et al,

2006). This may be due to hydrocodone itself possessing activity at MORs, with an in vitro MOR

displacement Ki 16-fold less than morphine and 2.4-fold less than oxycodone, but 12.5-fold greater

than codeine (Chen et al, 1991a). However, in a prospective observational study, hydrocodone

effectiveness was decreased in individuals concurrently using CYP2D6 substrates and inhibitors

(Monte et al, 2014). While hydrocodone has been identified as a P-gp substrate in vivo,

hydromorphone transport across the BBB is unknown (Somogyi et al, 2007). Since hydrocodone

activates MORs, undergoes minor CYP2D-mediated metabolism to the more potent

hydromorphone, and its analgesia is not greatly influenced by peripheral CYP2D metabolism, the

Page 148: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

126

relative role of parent and metabolite in hydrocodone analgesia apparently resembles that of

oxycodone more closely than for codeine and tramadol. This suggests that brain CYP2D-mediated

metabolism of hydrocodone to hydromorphone may play an inactivation role in mediating

analgesia, similar to that observed for oxycodone.

Fentanyl, a synthetic opioid, is predominantly converted by CYP3A4-mediated N-dealkylation to

the nontoxic and inactive metabolite norfentanyl (76% of urinary excretion by intravenous

administration), and is not metabolized by CYP2D6 (Smith, 2011). Inhibiting CYP3A4 in human

liver microsomes in vitro with ketoconazole and erythromycin reduced fentanyl metabolism (by

90%), while inhibiting CYP2D6 with quinidine had no effect (Feierman and Lasker, 1996).

Therefore, neither CYP2D6 genetic polymorphism nor variable brain CYP2D6 metabolism would

be expected to play a role in altering fentanyl metabolism and analgesia. However, like morphine

and oxymorphone, which are not further metabolized by CYP2D6, fentanyl analgesia in vivo could

serve as a useful control for off-target effects of brain CYP2D inhibitor and/or inducer

pretreatments.

5.3 Brain CYP2D and chronic opioid administration

5.3.1 Absolute vs. proportional decrease in codeine analgesia

One of the novel findings from chapter 2 was that the rate of development of codeine and

morphine tolerance, or decrease in peak analgesic response across repeated administration,

correlated with acute peak analgesic response. We hypothesized that an increase in acute codeine

analgesia via induction of brain CYP2D would result in one of the following outcomes: A) either

a similar absolute decrease in analgesia across time with a reduced proportional decrease in

analgesia and therefore a longer time to develop tolerance (Figure 35, Option A compared to

reference group), or B) a similar proportional decrease in analgesia across time, and therefore an

increased absolute decrease in analgesia with similar time to develop tolerance (Figure 35, Option

B). In support of option B, we demonstrated in chapter 2 (Figure 18c) that inducing brain CYP2D

with nicotine pretreatment increased acute codeine response (43.1 vs. 26.9% MPE, p<0.01),

producing a similar proportional (28 vs. 33%), but greater absolute (10.0 vs. 3.8% MPE, p<0.03),

decrease in analgesia over the first four days of codeine administration (Figure 35, Option B). We

Page 149: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

127

are unaware of any study in the literature that has described analgesic tolerance in this manner,

therefore the consequences of this finding are unclear. Our results suggest that a greater acute

analgesic response predicts a greater decrease in response (and therefore perception of analgesia)

over time per dose of codeine. Consistently, when acute analgesia was increased by giving larger

doses of codeine, there was a similar relationship observed between acute analgesia and the rate

of tolerance (Figure 23a). The impact of this finding on repeated/chronic opioid use, such as

codeine or oxycodone, may be important in predicting adverse outcomes from developing

tolerance.

It is important to consider the impact of an increased rate of opioid tolerance on opioid-taking

behaviours. Based on the models in Figure 35 and our findings in chapter 2, we propose that

individuals who experience greater acute analgesia from an opioid (in our model, codeine or

morphine) would reach half-maximal response at the same time of opioid-taking as those with

lower acute analgesic effect (Figure 35, purple and grey dotted lines). Individuals who experience

greater acute analgesia, by having greater brain CYP2D activity or beginning opioid treatment at

(or illicitly taking) a higher opioid dose, would experience a greater decrease in response than

those receiving a lower dose, and would be potentially more likely to escalate dose and to a greater

extent. Tolerance to opioid-induced side effects like respiratory depression can cause an increase

in mortality, for example when individuals take the same dose of opioid in a different environment,

or when individuals relapse after an extended period of abstinence during which tolerance is lost

(Cahill et al, 2016). For example, rats injected with a high dose of heroin in an environment

associated with prior heroin injections (associated context cues) experienced less mortality

(32.4%) than rats injected in an environment not associated with prior heroin injections (64.3%,

different context cues) and rats naïve to heroin (96.4%) (Siegel et al, 1982).

Page 150: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

128

Figure 35: Initial hypotheses (option A and B, compared to reference in grey) of the influence of increased acute codeine analgesia on the rate of development of tolerance

Figure 35: Initial hypotheses (option A and B, compared to reference in grey) of the influence

of increased acute codeine analgesia on the rate of development of tolerance. Compared to the

reference group (grey) showing decreased analgesia across repeated codeine administration, we

hypothesized two outcomes (option A and B) for the impact of increasing acute codeine analgesia

(by inducing brain CYP2D or increasing codeine dose, shown by red arrow) on the rate of codeine

tolerance. In option A, increasing acute analgesia may produce a smaller proportional, but similar

absolute, decrease in analgesia compared to the reference group, resulting in a greater time to

tolerant or days to 50% response (blue dashed line) with daily codeine administration.

Alternatively, in option B, increasing acute analgesia may produce a similar proportional, but

greater absolute, decrease in analgesia compared to the reference group, resulting in the same time

to tolerant or days to 50% response (purple dashed line) with daily codeine administration.

In a longitudinal prospective study of patients receiving opioids for cancer pain, individuals treated

with lower (15-20 mg, n=14) compared to higher (30-60 mg, n=16) doses of morphine, increased

morphine dose across three weeks to 57.1 mg and 83.4 mg, respectively (2.9-fold and 1.9-fold

increases, Figure 36) (Corli et al, 2015). Similarly, individuals treated with lower doses of

oxycodone (15-30 mg, n=19), compared to higher (40-80 mg, n=25), increased oxycodone dose

across three weeks to 48.9 mg and 87.2 mg, respectively (1.8-fold for both) (Corli et al, 2015).

This suggests that initiating opioid treatment at both low and high doses can produce dose-

escalation. However, acute analgesic response to the initial doses used was difficult to ascertain

from the study; it’s unknown if individuals who received high acute doses of opioid were

experiencing greater analgesia (Corli et al, 2015). Assessing analgesic response after first dose

Page 151: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

129

through visual analogue scales or pain indices could be used to assess the relationship between

acute effect and dose escalation that we propose from our study.

Figure 36: Dose-escalation of morphine and oxycodone after 21 days of opioid treatment in individuals receiving treatment for chronic cancer pain

Figure 36: Dose-escalation of morphine and oxycodone after 21 days of opioid treatment in

individuals receiving treatment for chronic cancer pain. Patients in a longitudinal prospective

study receiving opioid treatment for cancer pain were split evenly into low and high initial-dose

groups. Starting doses on day 1 and final doses on day 21 were recorded and averaged. Individuals

initiating treatment on low-dose morphine escalated to 2.9-fold the dose ion day 21, compared to

1.9-fold in those initiating on high-dose morphine. Individuals initiating treatment on low-dose

oxycodone escalated to 1.8-fold the dose on day 21, compared to 1.8-fold in those initiating on

high-dose oxycodone. Adapted from Corli et al. 2015.

Beyond opioid-taking behaviour, the findings in chapter 2 raise the question of the significance

of a relative vs. absolute decrease in analgesia over time to the neurobiology of tolerance. First,

there are agonist-specific short- and long-term neural adaptations involved in opioid tolerance

(introduced in section 1.1.4.2) that could be investigated with respect to greater acute MOR agonist

exposure producing a greater relative neural response. Following receptor binding, MOR

phosphorylation by G-protein receptor kinases (GRKs) initiates desensitization and internalization

of the MOR, which involves the association of β-arrestin proteins uncoupling MOR from G-

proteins (Dumas and Pollack, 2008; Koch and Hollt, 2008). These processes correlate with

agonist-induced desensitization and decreased functional receptor response. Morphine does not

initiate GRK-mediated MOR phosphorylation or MOR endocytosis (Keith et al, 1996; Sternini et

Page 152: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

130

al, 1996; Zhang et al, 1998). Instead, morphine has been suggested to produce analgesic tolerance

as a result of compensatory mechanisms counteracting receptor function (Koch and Hollt, 2008).

While acute morphine exposure inhibits adenylyl cyclase activity, chronic exposure produces a

counter-regulatory increase in adenylyl cyclase and resulting cAMP levels (adenylyl cyclase

superactivation) (Nevo et al, 1998; Sharma et al, 1975a; Sharma et al, 1975b). In support of this,

subcutaneous morphine given BID to rats at a dose that produced analgesic tolerance (decreased

tail-flick response) and physical dependence (increased withdrawal score) induced coupling of

stimulatory G-protein (Gs) to MOR in rat striatum and spinal cord (Wang et al, 2005a). Alterations

in G-protein coupling were not due to changes in expression of MOR or G-proteins, and decreasing

analgesic tolerance and physical dependence by co-pretreatment with an ultra-low dose of

naloxone (non-selective OR antagonist) decreased Gs-coupling (Wang et al, 2005a). Furthermore,

chronic morphine induced glial-cell (i.e. astrocytes and microglia) activation, inflammation, and

analgesic tolerance were increased in animals given daily intrathecal morphine, and decreased by

co-pretreatment of an ultra-low dose of naltrexone (MOR antagonist) (Mattioli et al, 2010).

Assessing differences in Gs-MOR coupling (by immunoprecipitation) or gliosis (by

immunohistochemistry) in rats tolerant to codeine, after different acute analgesic responses (either

by induction of brain CYP2D-activation to morphine or by increased doses), may be useful in

associating quantifiable neural alterations with the correlation of acute analgesic response to rate

of tolerance seen in chapter 2.

Oxycodone produces potent analgesic tolerance; in rats with induced inflammatory pain given

intraperitoneal oxycodone BID for 8 days, there was a 10-fold rightward shift in the dose response

curve of oxycodone-induced mechanical hyperalgesia on day 9 compared to day 1 (Thorn et al,

2017). Since we demonstrated in chapter 2 that acute response to codeine impacted the rate of

codeine tolerance, and in chapter 3 that brain CYP2D-mediated metabolism of oxycodone impacts

acute analgesic response, the development of oxycodone tolerance may similarly develop as a

function of acute oxycodone analgesia. Decreasing acute oxycodone analgesia by inducing brain-

CYP2D metabolism of oxycodone to oxymorphone may result in a decreased rate of oxycodone

tolerance. Compared to morphine, which promotes MOR desensitization without internalization,

the mechanism of oxycodone tolerance and extent of oxycodone-induced desensitization and

internalization of MORs is unclear (Allouche et al, 2014). Oxycodone does not strongly promote

Page 153: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

131

MOR phosphorylation by c-Jun N-Terminal Kinase, or recruit GRK-arrestin pathways (GRK

induced β-arrestin coupling), nor does it evoke MOR internalization (demonstrated with rat MORs

in human embryonic kidney [HEK293] cell lines) (Koch et al, 2009; McPherson et al, 2010;

Williams et al, 2013). Therefore, similar to codeine and morphine-induced tolerance, oxycodone

tolerance may occur due to adaptive processes like adenylyl cyclase superactivation.

5.3.2 Implications of brain CYP2D in opioid abuse liability

As discussed in section 1.3.3, the abuse potential of opioid compounds depends on numerous

factors, including the speed of onset of drug response, duration of response, and net positive and

negative drug effects (Wightman et al, 2012). Like analgesia, the subjective effects of opioid use

(e.g. euphoria and elation) are mediated by action at the MORs, and therefore sources of variability

in analgesic response may also impact abuse liability. Specifically, analogous to the analgesic

effects, the initial reinforcing effects of codeine and oxycodone may be mediated by their CYP2D

metabolism within the brain prior to peripheral metabolites entering the CNS (discussed in sections

5.2.1.1 [codeine] and 5.2.1.2. [oxycodone]).

Pharmacokinetic assessments of opioid abuse potential often use a measured abuse quotient,

defined as the ratio of maximum plasma drug concentration (CMAX) over the time to maximum

concentration (TMAX) (Harris et al, 2014). A high abuse quotient indicates increased likelihood of

drug liking and has been used as a standard comparator for abuse-deterrent opioid formulations

(Perrino et al, 2013; Webster et al, 2016). While our studies did not determine the full kinetic

profile of codeine and oxycodone, we can adapt this measure using the peak analgesic effect

(EMAX) and time to peak analgesia in animals across brain CYP2D inhibitor and/or inducer

pretreatment groups. In chapter 1, the predicted codeine abuse quotient was greatest in rats

pretreated with the brain CYP2D specific inducer nicotine (5.02% MPE/min), and lowest in rats

pretreated with the brain CYP2D specific inhibitor propranolol (1.4% MPE/min; Figure 37). This

relationship was reversed in chapter 3, where the predicted oxycodone abuse quotient was greatest

in rats with inhibited brain CYP2D (3.4 %MPE/min), compared to induced brain CYP2D (1.5

%MPE/min; Figure 37). This suggests that, similar to the effect of brain CYP2D on analgesia,

individuals with greater brain CYP2D activity may be at greater risk for codeine abuse, due to the

Page 154: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

132

rapid activation of codeine to morphine, but at a lesser risk for oxycodone abuse, due to the rapid

metabolism to, and subsequent efflux of, oxymorphone.

Figure 37: Modified abuse quotient (EMAX/TMAX) for codeine and oxycodone after brain CYP2D inhibitor and/or inducer pretreatment

Figure 37: Modified abuse quotient (EMAX/TMAX) for codeine and oxycodone after brain

CYP2D inhibitor and/or inducer pretreatment. Rats pretreated (PT) with the brain CYP2D

specific inducer nicotine had a greater codeine abuse quotient, while rats pretreated with the brain

CYP2D specific inhibitor propranolol had a greater oxycodone abuse quotient. Abuse quotient was

calculated using EMAX and TMAX values obtained in McMillan and Tyndale 2015, and McMillan et

al. 2017.

The impact of brain CYP2D on codeine and oxycodone abuse can be determined through animal

models of abuse liability; rats does-dependently self-administer codeine and oxycodone

(Mavrikaki et al, 2017; O'Connor et al, 2011). However, elucidating this in human populations is

more difficult, owing to the lack of literature assessing a direct role of central metabolism on drug

response, as well as the difficulty in isolating the effect of xenobiotic inducers on brain, compared

to liver, CYP2D6. For example, as discussed in section 1.4.4.2, CYP2D6 levels are higher in the

post-mortem brains of smokers compared to non-smokers, while liver CYP2D6 levels do not differ

(Mann et al, 2008; Miksys et al, 2002; Miksys and Tyndale, 2004; Zanger and Schwab, 2013).

However, cigarette smoking is strongly associated with greater drug taking overall, including

opioids; in a sample of over 600 tertiary pain clinic patients, 82% of smokers used opioid

medications compared to 50% of non-smokers (Eckhardt et al, 1994; Mackey et al, 2016). The

increased prevalence of devices that deliver nicotine alone, like electronic cigarettes, particularly

Page 155: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

133

in adolescents, suggests a unique population of individuals who may experience nicotine-induction

of brain CYP2D6 and thereby altered CNS-acting CYP2D6 substrate drug response, like opioids

(Dutra and Glantz, 2014). Comparing e-cigarette users to non-users, genotyped for CYP2D6

activity, on their response to CNS-acting CYP2D6 substrates used clinically could be useful in

elucidating an effect of nicotine on brain CYP2D6 metabolism.

5.4 Functional impact of brain CYP2D6 metabolism of opioids in humans

5.4.1 Brain CYP2D6 and the opioid epidemic

The findings in this thesis can be considered broadly in the context of the current opioid epidemic,

primarily with regard to the potential for brain CYP2D6 metabolism of opioids to influence opioid

response in humans, and what outcomes would be most impacted by variation. As discussed in

section 1.2.2.2, human CYP2D6 is highly genetically polymorphic, resulting in stratification of

individuals into metabolism phenotypes with measurable differences in CYP2D6 drug metabolism

and resultant response (Zanger et al, 2004). The experimental paradigms used throughout this

thesis were developed to inhibit and induce brain CYP2D selectively, without impacting hepatic

CYP2D. We showed that there was no impact of brain CYP2D inhibitor and/or inducer

pretreatments on plasma morphine levels after i.p. codeine (chapter 1); on ex vivo hepatic CYP2D

activity or the plasma codeine CYP2D metabolic ratio (morphine to codeine) after p.o. codeine

(chapter 2); or on ex vivo hepatic CYP2D activity or the plasma oxycodone CYP2D metabolic

ratio (oxymorphone to oxycodone) after p.o. oxycodone (chapter 3). Due to genetic

polymorphism, however, humans possess a wide-range of CYP2D6 activity in the liver that may

translate to the brain. CYP2D6 PMs have observable differences in brain function and behaviour,

which may be due, in part, to altered CNS metabolism of endogenous signaling molecules

(Kirchheiner et al, 2011). Furthermore, in CYP2D6 PMs homozygous for the gene deletion

CYP2D6*4, there was no detectable CYP2D6 in liver or enzymatic activity in vivo, and no

CYP2D6 detected in brain (Miksys et al, 2002). This suggests that genetic variation in CYP2D6

(e.g. decreased function, duplication, etc.) may similarly impact brain CYP2D6 enzyme activity.

To elucidate the impact of brain CYP2D6 metabolism on opioid compounds clinically, the

combination of genetics, xenobiotic inducers, and other influences on brain metabolism must be

considered.

Page 156: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

134

Brain CYP2D6 metabolism in humans may explain some of the existing variability in opioid levels

and response within or between CYP2D6 genotypes (or phenotype groups). In vivo phenotyping

was used initially to categorize individuals into four distinct phenotype groups; due to the large

number of allele combinations per phenotype group, activity scores for each allele have been

employed to better represent CYP2D6 activity (Gaedigk and Coetsee, 2008; Gaedigk et al, 2008).

However, variation still exists in both peripheral metabolism kinetics and subjective drug response

within these CYP2D6 activity score groups, particularly in CYP2D6 IMs and EMs. For example,

in a cohort of women prescribed codeine post-operatively, variability in total pain scores at day 2

after codeine was highest in those classified as CYP2D6 IMs (coefficient of variation = 0.44) or

EMs (0.46) compared to PMs (0.32) and UMs (0.19), despite the EMs and IMs groups containing

the greatest number of individuals (VanderVaart et al, 2011). Some of this variation may be

explained by differences in peripheral codeine and morphine levels due to a range of both codeine

dosing by weight (range of 0.4-11.1 mg/kg in IMs and 0.8-10.8 mg/kg in EMs) and CYP2D6

diplotypes in each predicted phenotype (12 in IMs and 3 in EMs) (VanderVaart et al, 2011).

However, variation in the brain CYP2D6 metabolism of codeine (for example through exposure

to xenobiotic brain CYP2D6 inducers or inhibitors) may also account for some of this within-

phenotype variability in codeine response.

Our findings in chapters 1 and 3 strongly suggest an impact of brain CYP2D6 on acute opioid

response. Opioids are routinely prescribed in an acute setting, with emergency department visits

being a major source of prescriptions; from 2001 to 2010 in the U.S. the percentage of emergency

department visits that resulted in opioid prescriptions increased from 20.8% to 31.0% (Mazer-

Amirshahi et al, 2014; Volkow et al, 2011). One prospective study in 2015 in the U.S. found that

90.5% of opioid-naïve patients (581 of 642) were prescribed an opioid after common outpatient

general surgery procedures (Hill et al, 2017). Our data suggests that individuals exposed to brain

CYP2D6 inducers (e.g. nicotine or alcohol) who were then administered opioids acutely would

experience increased analgesia from codeine, and decreased analgesia from oxycodone. This may

also manifest as increased or decreased subjective effects of each opioid, potentially impacting

likelihood of abuse, as well as adverse events.

Page 157: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

135

5.4.2 Potential sex differences in the brain CYP2D6 metabolism of opioids

Investigating CNS-acting drug response and CYP2D metabolism represents an interesting

intersection with known sex differences between males and females. Population-based studies

show that women are more likely to experience, and more frequently report, chronic pain

symptoms than men (Lee and Ho, 2013). Based on the Canadian Alcohol and Drug Use Monitoring

Survey, opioid use is higher among Canadian women (18.3%) than men (15.5%), while

prescription opioid abuse in U.S. treatment centres was reported to be higher among women

(15.4%) than men (11.1%) (Canada, 2015; Green et al, 2009). Similarly, female rats show a greater

vulnerability to drug abuse than males in numerous paradigms (Anker and Carroll, 2011).

Analgesia, drug reward, and vulnerability to relapse additionally vary with the estrous cycle in

female rats, although findings are inconsistent. Ovariectomized female rats given exogenous

estradiol had increased acquisition of heroin self-administration and heroin-maintained

responding; however, there was no effect of estrous cycle in sham-operated rats (Roth et al, 2002).

In contrast, mean heroin infusions were lower in female rats in proestrus (high estradiol and

progesterone) than during estrus and meta/diestrus (Lacy et al, 2016). Little is known about

differences in acute response to codeine and oxycodone between males and females. In comparing

the antinociceptive potency (ED50) of multiple opioids in the warm water tail withdrawal assay,

male rats displayed greater antinociceptive potency (lower ED50) of hydrocodone (1.7-fold),

hydromorphone (1.6-fold), and morphine (2.8-fold) than female rats, but there was no difference

in ED50 of oxycodone and codeine (Peckham and Traynor, 2006). Furthermore, male rats self-

administer more oxycodone than females at FR1 schedules, but females self-administer more

oxycodone than males at higher doses, although the specific stage of estrous cycle was not

examined (Mavrikaki et al, 2017)

The influence of sex and gender on CYP2D6 activity is also conflicting, as CYP2D6 activity has

been reported as faster and slower in females compared to males (Soldin et al, 2011; Soldin and

Mattison, 2009). Among females, this may vary based on the estrous cycle; estrogen decreases

CYP2D6 activity in vitro, and may decrease CYP2D6 activity in humanized Tg-CYP2D6 mice in

vivo by enhancing SHP repression of HNF-4α activation of CYP2D6 (section 1.2.2.2.2) (Pan and

Jeong, 2015; Pepper et al, 1991). In a large cohort of CYP2D6 EMs, females had slightly lower

(1.2-fold) urinary dextromethorphan/dextrorphan ratios than males, indicative of increased

Page 158: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

136

CYP2D6 activity, while there was no effect of co-use of oral contraceptives (Hagg et al, 2001).

Furthermore, little is known regarding sex differences in brain CYP2D. Pain and opioid response

are sexually dimorphic; as hepatic CYP2D6 activity does not vary greatly between sexes, this

suggests an influence of sex and/or sex hormones elsewhere on opioid response. Sex differences

in brain CYP2D6 activity may account for some of this variability.

Our lab has begun to investigate sex differences in brain CYP2D activity in rats, along with the

corresponding impact on brain opioid metabolism and response, in vivo. Pilot data has shown that

females have lower ex vivo brain CYP2D activity than males (0.6-fold), with no difference in ex

vivo liver CYP2D activity (Figure 38). In support of this, female rats had greater brain oxycodone

levels and lower brain oxycodone CYP2D-metabolic ratios (oxymorphone/oxycodone) than males

assessed by in vivo brain microdialysis after oral oxycodone administration (Figure 38). While

these findings are preliminary, they suggest that decreased brain CYP2D activity in females may

reduce brain oxycodone metabolism in vivo, resulting in increased oxycodone analgesia. This is

in line with the finding that female rats self-administer more oxycodone than males, suggesting

central metabolism of oxycodone to oxymorphone is decreased, yielding greater subjective effects

of oxycodone; this is, however, inconsistent with the finding that males and female rats do not

exhibit different codeine and oxycodone antinociceptive ED50 (Mavrikaki et al, 2017; Peckham

and Traynor, 2006). Additional studies are in progress to elucidate the full impact of sex on

oxycodone analgesia, peripheral oxycodone levels and metabolism, as well as phase-specific

effects of the female estrous cycle.

Page 159: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

137

Figure 38: Pilot data on sex differences in ex vivo rat brain and liver CYP2D activity and central oxycodone metabolism in vivo

Figure 38: Pilot data on sex differences in ex vivo rat brain and liver CYP2D activity and

central oxycodone metabolism in vivo. A) Brain, but not liver, CYP2D activity was lower in

female rats compared to males. B) Brain oxycodone levels were higher, and C) the brain

oxycodone CYP2D metabolic ratio (oxymorphone/oxycodone) was lower, in females compared

to males.

5.4.3 Endogenous opioid pain thresholds

Endogenous morphine-like opioids, including endorphins and enkephalins, are released under

conditions such as surgical, pathogenic, psychological, and physical stress and may be involved in

the modulation of nociception and pain sensitivity through activation of ORs (Candiotti et al, 2009;

Guarna et al, 2002). For example, in a group of patients undergoing cardiac bypass surgery who

received no morphine for post-surgery pain, there was a 13.8-fold increase in plasma morphine

concentrations 24 hr post-surgery (80 pg/ml to 1106 pg/ml) (Brix-Christensen et al, 1997).

CYP2D6 may contribute to the biosynthesis of endogenous morphine (endorphins) in vivo.

Incubating human polymorphonuclear white blood cells with the endogenous morphine precursor

tyramine dose-dependently increased morphine concentrations in vitro (Zhu et al, 2005). Morphine

synthesis was diminished when white blood cells were coincubated with tyramine and the

CYP2D6 substrate bufuralol, and was blocked entirely when coincubated with tyramine and

quinidine (CYP2D6 inhibitor) (Zhu et al, 2005). The presence of CYP2D6 enzyme was supported

by RT-PCR, suggesting CYP2D6 was expressed in human white blood cells and capable of

synthesizing endogenous morphine from exogenous precursors (Zhu et al, 2005).

Interindividual differences in CYP2D6 metabolism (e.g. genetic polymorphism) may impact the

production of endogenous opioids, and therefore functional outcomes like pain thresholds.

Page 160: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

138

CYP2D6 PMs had higher sensitivity to pressure pain than EMs indicating less tolerance to tonic

pain; a higher fraction of PMs (32%), than EMs (18%), prematurely withdrew their hand during

the cold pressure test (Sindrup et al, 1993). In a cohort of females in the U.S. investigated for post-

surgical pain, CYP2D6 PMs experienced severe post-operative pain more frequently than all other

phenotype groups suggesting lower pain thresholds (Yang et al, 2012). Additionally, in a separate

group of females from the same cohort taking morphine (not a CYP2D6 substrate), CYP2D6 UMs

required less morphine than all other phenotype groups (Candiotti et al, 2009). While there is

relatively little literature on this phenomenon, differences in pain sensitivity among CYP2D6

genotype groups suggests CYP2D6 may impact endogenous nociception, possibly through

endogenous opioid formation.

As discussed briefly in section 1.4.4.3.2, CYP2D6 expressed in rat brain membranes can

metabolize precursors tyramine and 5-methyltryptamine to dopamine and serotonin, respectively,

in vitro, while in vivo brain microdialysis studies indicate that this formation pathway may be

functional in vivo (Bromek et al, 2010; Bromek et al, 2011; Haduch et al, 2015; Haduch et al,

2013). However, this was investigated after ablation of the classical dopamine and serotonin

formation pathways in vivo and with amplification of the role of brain CYP2D through exogenous

precursor administration; therefore, the extent of brain CYP2D’s impact on basal endogenous

metabolism is unknown (Bromek et al, 2011; Haduch et al, 2015). The suggested impact of brain

CYP2D6 on endogenous neurochemical regulation, including that found in literature on CYP2D6

genotype and pain thresholds, infers that brain CYP2D may contribute to endogenous opioid

homeostasis; individuals exposed to xenobiotic inducers of brain CYP2D (e.g. smokers and

alcoholics) may have higher pain thresholds due to increased production of endogenous opioids in

the CNS. However, in chapter 1 and 3 we saw no effect of our brain CYP2D specific inhibitor

and inducer pretreatments on baseline nociception (i.e. threshold or sensitivity to a noxious

stimulus) and morphine-induced analgesia. Furthermore, in two separate studies assessing

electrical pain stimulation thresholds and cold pressor pain tolerance, there was no observable

difference in pain sensitivity between smokers and non-smokers (Unrod et al, 2004; Waller et al,

1983). This may be due to peripheral ORs (e.g. in the skin) and peripherally formed endogenous

opioids (e.g. from white blood cells) contributing more to pain sensitivity and thresholds than CNS

ORs and centrally formed endogenous opioids (Kapitzke et al, 2005).

Page 161: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

139

5.5 Extensions of research

5.5.1 Drug-drug interactions with brain CYP2D

Having shown that brain CYP2D activity impacts codeine analgesia (by formation of the more

active metabolite morphine) and oxycodone analgesia (by metabolism of the parent oxycodone to

the metabolite oxymorphone), it is likely that drug-drug interactions with substrates or inhibitors

of brain CYP2D may impact codeine and oxycodone response in a clinically significant manner

(introduced in section 1.2.2.2.2) (Feng et al, 2017). Numerous randomized clinical trials have

shown that CNS-acting drugs that are CYP2D6 inhibitors alter peripheral metabolism and response

to opioids that are more dependent on metabolic activation by CYP2D6 for analgesic effect (e.g.

codeine and tramadol). Administration of the antidepressant paroxetine, a CYP2D6 inhibitor, to

CYP2D6 EMs decreased the total plasma concentrations of tramadol’s CYP2D-metabolite O-

desmethyltramadol, and decreased the hypoalgesic effect of tramadol in experimental pain models

(Laugesen et al, 2005). Similarly, in patients hospitalized for acute back pain, administration of

the neuroleptic levomepromazine, a CYP2D6 inhibitor, in CYP2D6 EMs decreased the CYP2D6-

mediated O-demethylation ratio of morphine to codeine, indicating the likelihood of insufficient

codeine pain relief (Vevelstad et al, 2009). Therefore, while there are documented changes in

plasma O-desmethyltramadol (from tramadol) and morphine (from codeine) levels in these studies,

indicative of decreased peripheral CYP2D6 activity, the influence of paroxetine and

levomepromazine on brain CYP2D6 activity, among other CNS-acting CYP2D6 inhibitors, may

also contribute to the changes in opioid pharmacodynamic effects and response.

Comparatively, drug-drug interactions of brain CYP2D-inhibitors and opioids may produce a

greater pharmacodynamic impact on opioids where the contribution of hepatic CYP2D6 to

analgesia is negligible or unknown (e.g. oxycodone and hydrocodone). In a randomized clinical

trial of healthy individuals, paroxetine reduced total oxymorphone plasma levels from oxycodone

(44% reduction in AUC), but did not alter plasma oxycodone levels or response (Gronlund et al,

2010). The individuals in this study were primarily (10 of 12 participants) CYP2D6 EMs; it is

possible that interference in brain CYP2D6 metabolism by drug-drug interactions were masked

due to the range of CYP2D6 genotypes and therefore CYP2D6 basal activity in CYP2D6 EMs. A

randomized control trial, stratifying individuals based on CYP2D6 genotype and oxycodone

Page 162: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

140

response prior to, and following, paroxetine treatment (or another CNS-acting CYP2D6-inhibitor),

may be useful for assessing genotype-specific influences of drug-drug interactions on the response

to more metabolically-complex opioids like oxycodone.

5.5.2 Imaging brain CYP2D6 in humans

The ability to determine the location, abundance, and activity of CYP2D6 enzyme in the brain in

vivo using non-invasive molecular imaging could provide valuable information about brain

CYP2D6. In general, positron emission tomography (PET) has been a viable avenue to pursue this,

as radiotracers can be designed for catabolism by CYP enzymes (Holland et al, 2013). For

example, in separate studies assessing radiotracer labelling specificity in patients with epilepsy,

there was an increase in the CYP3A4-mediated metabolism of radiotracers [11C]-deprenyl and

[11C]-verapamil, attributed to the potential induction of liver CYP3A4 by anticonvulsant

medications (Abrahim et al, 2008; Cumming et al, 1999). However, PET imaging of CYP enzymes

has been difficult due to trapping or accumulation of enzymatic products, as well as specificity of

radiotracer ligand (Holland et al, 2013). High affinity inhibitors are currently being developed to

serve as tracers, particularly for measuring enzymes mediating intracellular signal transduction in

the brain (Holland et al, 2013). For example, CYP19A1 is expressed in human brain, and has been

shown to catalyze the formation of estradiol from testosterone, as well as the formation of estriol

and estrone from other androgen precursors in vitro (Azcoitia et al, 2011). CYP19A1 can be

visualized in rat, monkey, and human brain with the radiolabeled inhibitor [11C]-vorozole (Biegon

et al, 2010; Takahashi et al, 2006).

We are currently unaware of any characterized substrates or inhibitors of CYP2D6 that could be

used in PET imaging studies of brain CYP2D6, creating the need for surrogate measures of brain

CYP2D6 activity. One potential technique for imaging brain CYP2D6 would be to use radioligand

displacement studies directed at the functional effect of a CYP2D6-mediated metabolite, such as

the CYP2D6 probe substrate dextromethorphan metabolism to dextrorphan. Dextromethorphan

blocks excitatory n-methyl-d-aspartate (NMDA) receptors in vitro; dextrorphan also binds to

NMDA but with greater affinity than dextromethorphan (Church et al, 1994; Parsons et al, 1995).

In monkeys trained to discriminate phenylcyclidine (an NMDA receptor antagonist) from saline,

dextrorphan was a greater reinforcer than dextromethorphan indicative of greater NMDA receptor

Page 163: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

141

activation (Nicholson et al, 1999). Furthermore, the positive reinforcing effects of

dextromethorphan were reduced in individuals pretreated with the CYP2D6 inhibitor quinidine,

suggesting CYP2D6 metabolism to dextrorphan may be responsible for dextromethorphan

response (Zawertailo et al, 2010). Assessing dextrorphan-induced NMDA receptor antagonism is

one potential technique for elucidating brain CYP2D activity in vivo. While there is no agreed

upon PET radiotracer for labelling NMDA receptors, there are numerous candidates (e.g.

fluoropiperidine derivatives) that display high selectivity and brain uptake (Kassenbrock et al,

2016). If brain CYP2D6 enzyme activity is increased in smokers (or individuals administered

nicotine), the brain CYP2D6 metabolism of dextromethorphan to dextrorphan would be increased

along with the resulting dextrorphan-induced NMDA receptor antagonism, presenting as a

decrease in CNS NMDA receptor binding and radiolabelling by the PET tracer.

Lastly, there has been progress made towards using functional magnetic resonance imaging (fMRI)

techniques to quantify brain perfusion as an indicator of CYP2D6 genotype. Brain perfusion at

rest is a well-characterized index of cerebral activity; variation in resting perfusion is associated

with specific personality traits, as well as genetic polymorphisms in proteins associated with these

traits (e.g. serotonin transporter) (Kirchheiner et al, 2011; Rao et al, 2007; Shulman et al, 1997).

CYP2D6 genotype may associate with personality phenotypes, due to the metabolism of

endogenous compounds (demonstrated in vitro) such as the neurotransmitter dopamine and the

endocannabinoid anandamide (Hiroi et al, 1998b; Snider et al, 2008). Relative to CYP2D6 EMs,

CYP2D6 PMs had 15% higher perfusion levels in the thalamus; fMRI detection of brain perfusion

provided relatively successful prediction of individuals as CYP2D6 UMs (sensitivity and

specificity ranging from 85–87%) and CYP2D6 PMs (71–79%) (Kirchheiner et al, 2011;

Napolitano et al, 2017). Brain perfusion in individuals exposed to xenobiotic inducers of brain

CYP2D6 (e.g. nicotine or ethanol) could be assessed as a proxy of brain CYP2D6 activity, where

decreased brain perfusion would be indicative of increased brain CYP2D6 activity. However, this

technique would also be sensitive to secondary changes associated with those xenobiotic

compounds; for example, smokers show decreased brain perfusion in brain regions associated with

maintenance of addictive disorders (Durazzo et al, 2015). Despite this, the effect of nicotine alone

(e.g. through nicotine replacement therapy, or electronic-cigarettes) on brain perfusion is

Page 164: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

142

unknown. Active investigation is underway to determine the optimal imaging technique for

translation of our preclinical findings on inducible brain CYP2D to humans.

5.5.3 Therapeutic targeting of brain CYP2D6 in humans

The potential to inhibit or induce brain CYP2D6 with, or independently of, liver CYP2D6 in

humans for therapeutic purposes is an interesting extension of our brain metabolism research. In

regards to this thesis specifically, targeted inhibition or induction of the brain CYP2D6 metabolism

of opioids like codeine or oxycodone could be used to 1) modulate acute analgesia, for example

by inhibiting brain CYP2D6 in CYP2D6 UMs prior to oxycodone treatment to increase analgesic

response, or to 2) decrease the rate of development of tolerance, for example by inhibiting brain

CYP2D6 in CYP2D6 UMs prior to codeine treatment to reduce acute analgesic response.

Increasing brain CYP2D6 activity in humans may be of therapeutic use in protecting against

neurotoxicity. As outlined in section 1.4.4.3.1, individuals with Parkinson’s disease have 40%

lower brain CYP2D6 levels than age-matched controls, and CYP2D6 PMs are at greater risk for

developing Parkinson’s disease (Elbaz et al, 2004; Mann et al, 2012; Mann and Tyndale, 2010).

As many neurotoxins are inactivated by CYP2D6 (e.g. MPTP, β-carbolines), brain CYP2D6

activity may function to protect against neurotoxicity, as seen in Parkinson’s disease (Miksys and

Tyndale, 2006; Zanger et al, 2004). In support of this, smokers are at a lower risk for Parkinson’s

disease which may be due, in part, to greater inactivation of neurotoxins via increased brain

CYP2D6 levels in smokers (mentioned in introduction section 1.4.4.3.1) (Alves et al, 2004;

Miksys and Tyndale, 2006).

The most direct method for inducing brain CYP2D6 specifically, as utilized in the animal model

paradigms in this study, would be to use xenobiotic inducers of brain CYP activity. As discussed

in section 1.4.4.2, while hepatic CYP2D6 levels in humans are primarily regulated by genetics,

brain CYP2D6 activity may be altered by exposure to select xenobiotics. Smokers and alcoholics

have higher CYP2D6 levels in the brain, while nicotine and ethanol increase rat brain CYP2D

expression and dose-dependently increase monkey brain CYP2D expression (Mann et al, 2008;

Miksys et al, 2002; Miksys and Tyndale, 2004; Miller et al, 2014; Warner and Gustafsson, 1994;

Yue et al, 2008). However, the individual influences of nicotine and ethanol on human brain

CYP2D6 in vivo have yet to be characterized, including associated nuances, such as the time-

Page 165: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

143

course of effect, the dose dependency of effect, the functional outcome of effect on drug

metabolism and response, etc.

Conversely, gene therapies, like the use of microRNA (miRNA), could be used for brain specific

down-regulation of CYP2D activity. miRNA are short non-coding RNA strands complementary

to targeted mRNAs, that function as components of the RNA-induced silencing complex leading

to translational inhibition or degradation of those specific mRNA (Esteller, 2011). As briefly

mentioned in 1.4.4.2, exogenous testosterone administration decreased CYP2D levels in rat brain;

testosterone increased levels of brain-specific miRNA (miR-101 and miR-128-2) in human

neurons (SH-SY5Y) and astrocytes (U251) but not hepatocytes (HepG2) in vitro, while

overexpression of those miRNA decreased CYP2D mRNA (Li et al, 2015a). Testosterone

decreased levels of the CYP2D metabolite O-desmethyltramadol in rat brain, but not plasma, after

i.p. tramadol administration, and decreased in tramadol analgesia, suggesting testosterone was

inhibiting brain CYP2D activity in vivo, likely by upregulation of miRNA (Li et al, 2015a).

Delivering miRNA to the brain is a challenge, as the BBB hinders accumulation of active

compounds in the CNS and therefore limits transfection efficiency. The two strategies used most

commonly are miRNA mimics to restore suppressed miRNA levels, or anti-miRNA to repress

overactive miRNA function (van Rooij and Kauppinen, 2014). In general, gene therapies using

RNA interference (like miRNA) lack stability due to degradation, therefore delivery systems are

used to both protect miRNA, and assist their entrance into the CNS (Chen et al, 2015). Some

examples include viral vectors, lipid-based carriers, gold nanoparticles, polymer-based carriers,

carbon-based carriers, and magnetic resonance-guided focused ultrasound (Wen, 2016). The

delivery of miRNA specific to brain CYP2D6 in humans could be utilized to therapeutically inhibit

brain CYP2D6, therefore increasing the brain levels of CYP2D-substrate parent compounds and

therapeutic effect or preventing the formation of active toxic metabolite(s). Specific to this thesis,

inhibiting brain CYP2D6 could function to regulate short and long-term effects to opioid

compounds, for example decreasing acute codeine analgesia and decreasing the rate of codeine

tolerance.

Page 166: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

144

5.5.4 The mechanism of inducible brain-specific CYP2D

The neural mechanism(s) behind the brain specific induction of CYP2D by nicotine pretreatment

is currently unknown. Nicotine treatment increased brain CYP2D expression in rats and monkeys,

neither of which were associated with changes in brain CYP2D mRNA levels, or changes in

hepatic CYP2D enzyme activity and mRNA levels (Mann et al, 2008; Miller et al, 2014; Yue et

al, 2008). Because nicotine did not increase brain CYP2D mRNA levels, this indicated that

CYP2D induction likely occurs via non-transcriptional events, such as increasing translational

efficiency (decreased splicing or increased translation), increasing enzyme stability, or decrease

enzyme degradation.

One potential mechanism behind the induction of brain CYP2D by nicotine is alteration of the

ubiquitin-mediated protein degradation of CYP2D in the brain specifically. The binding of

ubiquitin targets proteins for proteasome degradation and fragmentation, where alterations in this

pathway may produce functional changes in enzyme expression (Kane et al, 2004). For example,

the induction of rat liver CYP2E1 by ethanol is accompanied by decreased CYP2E1 protein

ubiquitination (Roberts et al, 1995). Ethanol induced CYP2E1 in rat liver microsomes without

changing mRNA levels; less radiolabeled CYP2E1 activity was lost from ethanol-induced

microsomes (50%) than control microsomes (93%) in the first 48 hr (Roberts et al, 1995). After

inducing protein degradation with carbon tetrachloride, there were fewer immunolabeled

ubiquitin-CYP2E1 complexes in liver microsomes from rats treated with ethanol than in control

livers microsomes (Roberts et al, 1995). Nicotine also impacts ubiquitination-related proteins in

rat brain, though this is in a region-specific manner. Ubiquitin, ubiquitin-conjugating enzymes,

proteasomal subunits, and ubiquitin-chaperone proteins were all upregulated by nicotine in the

prefrontal cortex, but downregulated in the medial basal hypothalamus (Kane et al, 2004). The

paradigm of short-term nicotine pretreatment used in this thesis may be preferentially down-

regulating ubiquitin-related proteins in the brain regions associated with greatest increases in

CYP2D protein activity (cerebellum, hippocampus, and striatum) (Yue et al, 2008). A study

similar to that which identified ethanol induction of CYP2E1 could be designed to look at this

directly, with brain membranes from rats pretreated with nicotine compared to vehicle, as well as

antibodies raised against rat CYP2D isoforms and ubiquitin.

Page 167: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

145

5.6 Conclusions

Our findings contribute to the existing literature on brain CYP metabolism as a novel source of

variability through brain specific drug metabolism and resulting drug response. Using paradigms

developed in our laboratory to inhibit (by direct intracerebroventricular injection of propranolol

and propafenone) and induce (by short-term pretreatment with nicotine) rat CYP2D enzyme

specifically in the brain, we investigated the impact of brain CYP2D metabolism in vivo on

codeine and oxycodone central metabolism and resulting effect. We found that inhibiting brain

CYP2D in rats decreased, while inducing brain CYP2D increased, brain morphine levels from

codeine and resulting codeine analgesia. We further found that the increase in acute codeine

analgesia after inducing brain CYP2D resulted in a more rapid rate of onset of tolerance across

repeated codeine administration. The rate of tolerance, or decrease in analgesia per dose, correlated

to the acute analgesic response following the first dose of codeine, which itself was dependent on

codeine dose or level of brain CYP2D activity. Lastly, we found that, in contrast to codeine

analgesia, oxycodone analgesia was increased by inhibiting brain CYP2D, and was decreased by

inducing brain CYP2D. Brain drug levels were assessed through in vivo microdialysis, and brain

oxycodone levels were increased after inhibiting brain CYP2D, while brain oxymorphone levels

were increased after inducing brain CYP2D, indicating that brain oxycodone levels were primarily

responsible for oxycodone analgesia.

Opioid analgesics, while effective for the intended treatment of pain, are often misused and abused

resulting in opioid-related morbidity and mortality. Interindividual variation in CNS-acting drug

response, specifically in opioid-induced pain relief, and risk for dependence, is not fully

understood. The brain CYP metabolism of opioids, as we have explicitly shown for brain CYP2D,

could explain some of this variation, and add to the known risk factors for failure of opioid

therapeutic effect, as well as consequences of chronic use like tolerance, transition to abuse, and

overdose.

The tissue-specific regulation of CYP2D may have a unique impact on drug metabolism and

response. Smokers, similar to rats and monkeys pretreated with nicotine, have greater CYP2D

levels in the brain, but not liver, compared to non-smokers; smokers may therefore experience

altered CNS-acting drug metabolism and drug response (e.g. increased activation of codeine to

Page 168: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

146

morphine, or increased metabolism, and therefore removal, of oxycodone to oxymorphone), not

detected in peripheral pharmacokinetics. Remaining questions regarding the role of brain CYP2D

in opioid metabolism include: 1) the impact of brain CYP2D metabolism of opioids on abuse

liability, 2) the mechanism(s) behind the induction/regulation of brain CYP2D in animals and brain

CYP2D6 in humans, 3) the development of useful clinical measures or imaging techniques for

predicting and detecting brain CYP2D6 expression, function, and impact in humans, and 4)

whether inhibition or induction of brain CYP2D6 in humans (either through targeted therapeutics

or drug-drug interactions) produces similar functional outcomes as those seen in rats in vivo.

Page 169: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

147

6. References

Abdel-Rahman SM, Leeder JS, Wilson JT, Gaedigk A, Gotschall RR, Medve R, et al (2002).

Concordance between tramadol and dextromethorphan parent/metabolite ratios: the influence of

CYP2D6 and non-CYP2D6 pathways on biotransformation. J Clin Pharmacol 42(1): 24-29.

Abler B, Hofer C, Viviani R (2008). Habitual emotion regulation strategies and baseline brain

perfusion. Neuroreport 19(1): 21-24.

Abrahim A, Luurtsema G, Bauer M, Karch R, Lubberink M, Pataraia E, et al (2008). Peripheral

metabolism of (R)-[11C]verapamil in epilepsy patients. Eur J Nucl Med Mol Imaging 35(1):

116-123.

Adler TK, Fujimoto JM, Way EL, Baker EM (1955). The metabolic fate of codeine in man. J

Pharmacol Exp Ther 114(3): 251-262.

Agundez JA, Jimenez-Jimenez FJ, Alonso-Navarro H, Garcia-Martin E (2014). Drug and

xenobiotic biotransformation in the blood-brain barrier: a neglected issue. Front Cell Neurosci 8:

335.

Aichhorn W, Weiss U, Marksteiner J, Kemmler G, Walch T, Zernig G, et al (2005). Influence of

age and gender on risperidone plasma concentrations. J Psychopharmacol 19(4): 395-401.

Aklillu E, Persson I, Bertilsson L, Johansson I, Rodrigues F, Ingelman-Sundberg M (1996).

Frequent distribution of ultrarapid metabolizers of debrisoquine in an ethiopian population

carrying duplicated and multiduplicated functional CYP2D6 alleles. J Pharmacol Exp Ther

278(1): 441-446.

Albores A, Ortega-Mantilla G, Sierra-Santoyo A, Cebrian ME, Munoz-Sanchez JL, Calderon-

Salinas JV, et al (2001). Cytochrome P450 2B (CYP2B)-mediated activation of methyl-

parathion in rat brain extracts. Toxicol Lett 124(1-3): 1-10.

Allouche S, Noble F, Marie N (2014). Opioid receptor desensitization: mechanisms and its link

to tolerance. Front Pharmacol 5: 280.

Alves G, Kurz M, Lie SA, Larsen JP (2004). Cigarette smoking in Parkinson's disease: influence

on disease progression. Mov Disord 19(9): 1087-1092.

Ananthan S (2006). Opioid ligands with mixed mu/delta opioid receptor interactions: an

emerging approach to novel analgesics. AAPS J 8(1): E118-125.

Andreassen TN, Klepstad P, Davies A, Bjordal K, Lundstrom S, Kaasa S, et al (2011).

Influences on the pharmacokinetics of oxycodone: a multicentre cross-sectional study in 439

adult cancer patients. Eur J Clin Pharmacol 67(5): 493-506.

Anker JJ, Carroll ME (2011). Females are more vulnerable to drug abuse than males: evidence

from preclinical studies and the role of ovarian hormones. Curr Top Behav Neurosci 8: 73-96.

Page 170: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

148

Azcoitia I, Yague JG, Garcia-Segura LM (2011). Estradiol synthesis within the human brain.

Neuroscience 191: 139-147.

Bachmakov I, Werner U, Endress B, Auge D, Fromm MF (2006). Characterization of beta-

adrenoceptor antagonists as substrates and inhibitors of the drug transporter P-glycoprotein.

Fundam Clin Pharmacol 20(3): 273-282.

Bailey CP, Llorente J, Gabra BH, Smith FL, Dewey WL, Kelly E, et al (2009). Role of protein

kinase C and mu-opioid receptor (MOPr) desensitization in tolerance to morphine in rat locus

coeruleus neurons. Eur J Neurosci 29(2): 307-318.

Barnett ML, Olenski AR, Jena AB (2017). Opioid-Prescribing Patterns of Emergency Physicians

and Risk of Long-Term Use. N Engl J Med 376(7): 663-673.

Basbaum AI, Bautista DM, Scherrer G, Julius D (2009). Cellular and molecular mechanisms of

pain. Cell 139(2): 267-284.

Basbaum AI, Fields HL (1984). Endogenous pain control systems: brainstem spinal pathways

and endorphin circuitry. Annu Rev Neurosci 7: 309-338.

Baum LO, Strobel HW (1997). Regulation of expression of cytochrome P-450 2D mRNA in rat

brain with steroid hormones. Brain Res 765(1): 67-73.

Befort K, Filliol D, Decaillot FM, Gaveriaux-Ruff C, Hoehe MR, Kieffer BL (2001). A single

nucleotide polymorphic mutation in the human mu-opioid receptor severely impairs receptor

signaling. J Biol Chem 276(5): 3130-3137.

Behbehani MM, Fields HL (1979). Evidence that an excitatory connection between the

periaqueductal gray and nucleus raphe magnus mediates stimulation produced analgesia. Brain

Res 170(1): 85-93.

Benowitz NL (1999). Nicotine addiction. Prim Care 26(3): 611-631.

Benowitz NL, Peng M, Jacob P, 3rd (2003). Effects of cigarette smoking and carbon monoxide

on chlorzoxazone and caffeine metabolism. Clin Pharmacol Ther 74(5): 468-474.

Berg CJ, Wen H, Cummings JR, Ahluwalia JS, Druss BG (2013). Depression and substance

abuse and dependency in relation to current smoking status and frequency of smoking among

nondaily and daily smokers. Am J Addict 22(6): 581-589.

Bergh AF, Strobel HW (1996). Anatomical distribution of NADPH-cytochrome P450 reductase

and cytochrome P4502D forms in rat brain: effects of xenobiotics and sex steroids. Mol Cell

Biochem 162(1): 31-41.

Berrendero F, Kieffer BL, Maldonado R (2002). Attenuation of nicotine-induced

antinociception, rewarding effects, and dependence in mu-opioid receptor knock-out mice. J

Neurosci 22(24): 10935-10940.

Page 171: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

149

Bertelsen KM, Venkatakrishnan K, Von Moltke LL, Obach RS, Greenblatt DJ (2003). Apparent

mechanism-based inhibition of human CYP2D6 in vitro by paroxetine: comparison with

fluoxetine and quinidine. Drug Metab Dispos 31(3): 289-293.

Bettiol SS, Rose TC, Hughes CJ, Smith LA (2015). Alcohol Consumption and Parkinson's

Disease Risk: A Review of Recent Findings. J Parkinsons Dis 5(3): 425-442.

Beyenburg S, Mitchell AJ, Schmidt D, Elger CE, Reuber M (2005). Anxiety in patients with

epilepsy: systematic review and suggestions for clinical management. Epilepsy Behav 7(2): 161-

171.

Biegon A, Kim SW, Alexoff DL, Jayne M, Carter P, Hubbard B, et al (2010). Unique

distribution of aromatase in the human brain: in vivo studies with PET and [N-methyl-

11C]vorozole. Synapse 64(11): 801-807.

Borg KO, Carlsson E, Hoffmann KJ, Jonsson TE, Thorin H, Wallin B (1975). Metabolism of

metoprolol-(3-h) in man, the dog and the rat. Acta Pharmacol Toxicol (Copenh) 36(Suppl 5):

125-135.

Bostrom E, Hammarlund-Udenaes M, Simonsson US (2008). Blood-brain barrier transport helps

to explain discrepancies in in vivo potency between oxycodone and morphine. Anesthesiology

108(3): 495-505.

Bostrom E, Simonsson US, Hammarlund-Udenaes M (2005). Oxycodone pharmacokinetics and

pharmacodynamics in the rat in the presence of the P-glycoprotein inhibitor PSC833. J Pharm

Sci 94(5): 1060-1066.

Bostrom E, Simonsson US, Hammarlund-Udenaes M (2006). In vivo blood-brain barrier

transport of oxycodone in the rat: indications for active influx and implications for

pharmacokinetics/pharmacodynamics. Drug Metab Dispos 34(9): 1624-1631.

Boussadia B, Ghosh C, Plaud C, Pascussi JM, de Bock F, Rousset MC, et al (2014). Effect of

status epilepticus and antiepileptic drugs on CYP2E1 brain expression. Neuroscience 281: 124-

134.

Bouw MR, Gardmark M, Hammarlund-Udenaes M (2000). Pharmacokinetic-pharmacodynamic

modelling of morphine transport across the blood-brain barrier as a cause of the antinociceptive

effect delay in rats--a microdialysis study. Pharm Res 17(10): 1220-1227.

Bozarth MA, Wise RA (1981). Intracranial self-administration of morphine into the ventral

tegmental area in rats. Life Sci 28(5): 551-555.

Branch RA, Adedoyin A, Frye RF, Wilson JW, Romkes M (2000). In vivo modulation of CYP

enzymes by quinidine and rifampin. Clin Pharmacol Ther 68(4): 401-411.

Britt MD, Wise RA (1983). Ventral tegmental site of opiate reward: antagonism by a hydrophilic

opiate receptor blocker. Brain Res 258(1): 105-108.

Page 172: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

150

Britto MR, Wedlund PJ (1992). Cytochrome P-450 in the brain. Potential evolutionary and

therapeutic relevance of localization of drug-metabolizing enzymes. Drug Metab Dispos 20(3):

446-450.

Brix-Christensen V, Tonnesen E, Sanchez RG, Bilfinger TV, Stefano GB (1997). Endogenous

morphine levels increase following cardiac surgery as part of the antiinflammatory response? Int

J Cardiol 62(3): 191-197.

Brockmoller J, Kirchheiner J, Schmider J, Walter S, Sachse C, Muller-Oerlinghausen B, et al

(2002). The impact of the CYP2D6 polymorphism on haloperidol pharmacokinetics and on the

outcome of haloperidol treatment. Clin Pharmacol Ther 72(4): 438-452.

Brodsky M, Elliott K, Hynansky A, Jenab S, Inturrisi CE (1995). Quantitation of mu-opioid

receptor (MOR-1) mRNA in selected regions of the rat CNS. Neuroreport 6(5): 725-729.

Bromek E, Haduch A, Daniel WA (2010). The ability of cytochrome P450 2D isoforms to

synthesize dopamine in the brain: An in vitro study. Eur J Pharmacol 626(2-3): 171-178.

Bromek E, Haduch A, Golembiowska K, Daniel WA (2011). Cytochrome P450 mediates

dopamine formation in the brain in vivo. J Neurochem 118(5): 806-815.

Brook K, Bennett J, Desai SP (2017). The Chemical History of Morphine: An 8000-year

Journey, from Resin to de-novo Synthesis. J Anesth Hist 3(2): 50-55.

Brtko J, Dvorak Z (2011). Role of retinoids, rexinoids and thyroid hormone in the expression of

cytochrome p450 enzymes. Curr Drug Metab 12(2): 71-88.

Buratti FM, Volpe MT, Meneguz A, Vittozzi L, Testai E (2003). CYP-specific bioactivation of

four organophosphorothioate pesticides by human liver microsomes. Toxicol Appl Pharmacol

186(3): 143-154.

Bushnell MC, Ceko M, Low LA (2013). Cognitive and emotional control of pain and its

disruption in chronic pain. Nat Rev Neurosci 14(7): 502-511.

Cahill CM, Walwyn W, Taylor AMW, Pradhan AAA, Evans CJ (2016). Allostatic Mechanisms

of Opioid Tolerance Beyond Desensitization and Downregulation. Trends Pharmacol Sci 37(11):

963-976.

Cai R, Crane E, Poneleit K, Paulozzi L (2010). Emergency department visits involving

nonmedical use of selected prescription drugs in the United States, 2004-2008. J Pain Palliat

Care Pharmacother 24(3): 293-297.

Cairns W, Smith CA, McLaren AW, Wolf CR (1996). Characterization of the human

cytochrome P4502D6 promoter. A potential role for antagonistic interactions between members

of the nuclear receptor family. J Biol Chem 271(41): 25269-25276.

Campa D, Gioia A, Tomei A, Poli P, Barale R (2008). Association of ABCB1/MDR1 and

OPRM1 gene polymorphisms with morphine pain relief. Clin Pharmacol Ther 83(4): 559-566.

Page 173: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

151

Canada H (2015). Canadian Alcohol and Drug Use Monitoring Survey: Summary of Results for

2012.

Candiotti KA, Yang Z, Rodriguez Y, Crescimone A, Sanchez GC, Takacs P, et al (2009). The

impact of CYP2D6 genetic polymorphisms on postoperative morphine consumption. Pain Med

10(5): 799-805.

Caron E, Rioux N, Nicolas O, Lebel-Talbot H, Hamelin BA (2005a). Quantification of the

expression and inducibility of 12 rat cytochrome P450 isoforms by quantitative RT-PCR. J

Biochem Mol Toxicol 19(6): 368-378.

Caron E, Rioux N, Nicolas O, Lebel-Talbot H, Hamelin BA (2005b). Quantification of the

expression and inducibility of 12 rat cytochrome P450 isoforms by quantitative RT-PCR. J

Biochem Mol Toxicol 19(6): 368-378.

Carstens E, Anderson KA, Simons CT, Carstens MI, Jinks SL (2001). Analgesia induced by

chronic nicotine infusion in rats: differences by gender and pain test. Psychopharmacology

(Berl) 157(1): 40-45.

Castagne V, Moser PC, Porsolt RD (2009). Preclinical behavioral models for predicting

antipsychotic activity. Adv Pharmacol 57: 381-418.

Celver J, Xu M, Jin W, Lowe J, Chavkin C (2004). Distinct domains of the mu-opioid receptor

control uncoupling and internalization. Mol Pharmacol 65(3): 528-537.

Chakrabarti S, Rivera M, Yan SZ, Tang WJ, Gintzler AR (1998a). Chronic morphine augments

G(beta)(gamma)/Gs(alpha) stimulation of adenylyl cyclase: relevance to opioid tolerance. Mol

Pharmacol 54(4): 655-662.

Chakrabarti S, Wang L, Tang WJ, Gintzler AR (1998b). Chronic morphine augments adenylyl

cyclase phosphorylation: relevance to altered signaling during tolerance/dependence. Mol

Pharmacol 54(6): 949-953.

Chen Y, Zhao H, Tan Z, Zhang C, Fu X (2015). Bottleneck limitations for microRNA-based

therapeutics from bench to the bedside. Pharmazie 70(3): 147-154.

Chen ZR, Irvine RJ, Bochner F, Somogyi AA (1990). Morphine formation from codeine in rat

brain: a possible mechanism of codeine analgesia. Life Sci 46(15): 1067-1074.

Chen ZR, Irvine RJ, Somogyi AA, Bochner F (1991a). Mu receptor binding of some commonly

used opioids and their metabolites. Life Sci 48(22): 2165-2171.

Chen ZR, Somogyi AA, Reynolds G, Bochner F (1991b). Disposition and metabolism of codeine

after single and chronic doses in one poor and seven extensive metabolisers. Br J Clin

Pharmacol 31(4): 381-390.

Cheng J, Zhen Y, Miksys S, Beyoglu D, Krausz KW, Tyndale RF, et al (2013). Potential role of

CYP2D6 in the central nervous system. Xenobiotica 43(11): 973-984.

Page 174: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

152

Chiang HL, Chia YY, Lin HS, Chen CH (2016). The Implications of Tobacco Smoking on Acute

Postoperative Pain: A Prospective Observational Study. Pain Res Manag 2016: 9432493.

Christrup LL (1997). Morphine metabolites. Acta Anaesthesiol Scand 41(1 Pt 2): 116-122.

Chuang SS, Helvig C, Taimi M, Ramshaw HA, Collop AH, Amad M, et al (2004). CYP2U1, a

novel human thymus- and brain-specific cytochrome P450, catalyzes omega- and (omega-1)-

hydroxylation of fatty acids. J Biol Chem 279(8): 6305-6314.

Church J, Sawyer D, McLarnon JG (1994). Interactions of dextromethorphan with the N-methyl-

D-aspartate receptor-channel complex: single channel recordings. Brain Res 666(2): 189-194.

Cleary J, Mikus G, Somogyi A, Bochner F (1994). The influence of pharmacogenetics on opioid

analgesia: studies with codeine and oxycodone in the Sprague-Dawley/Dark Agouti rat model. J

Pharmacol Exp Ther 271(3): 1528-1534.

Coleman T, Spellman EF, Rostami-Hodjegan A, Lennard MS, Tucker GT (2000). The 1'-

hydroxylation of Rac-bufuralol by rat brain microsomes. Drug Metab Dispos 28(9): 1094-1099.

Collett BJ (1998). Opioid tolerance: the clinical perspective. Br J Anaesth 81(1): 58-68.

Comer SD, Sullivan MA, Vosburg SK, Kowalczyk WJ, Houser J (2010). Abuse liability of

oxycodone as a function of pain and drug use history. Drug Alcohol Depend 109(1-3): 130-138.

Compton WM, Jones CM, Baldwin GT (2016). Relationship between Nonmedical Prescription-

Opioid Use and Heroin Use. N Engl J Med 374(2): 154-163.

Cone EJ, Heltsley R, Black DL, Mitchell JM, Lodico CP, Flegel RR (2013). Prescription

opioids. II. Metabolism and excretion patterns of hydrocodone in urine following controlled

single-dose administration. J Anal Toxicol 37(8): 486-494.

Conroy JL, Fang C, Gu J, Zeitlin SO, Yang W, Yang J, et al (2010). Opioids activate brain

analgesic circuits through cytochrome P450/epoxygenase signaling. Nat Neurosci 13(3): 284-

286.

Cook DJ, Finnigan JD, Cook K, Black GW, Charnock SJ (2016). Cytochromes P450: History,

Classes, Catalytic Mechanism, and Industrial Application. Adv Protein Chem Struct Biol 105:

105-126.

Cooper RJ (2013). Over-the-counter medicine abuse - a review of the literature. J Subst Use

18(2): 82-107.

Corli O, Roberto A, Greco MT, Montanari M (2015). Assessing the response to opioids in cancer

patients: a methodological proposal and the results. Support Care Cancer 23(7): 1867-1873.

Costa LG, McDonald BE, Murphy SD, Omenn GS, Richter RJ, Motulsky AG, et al (1990).

Serum paraoxonase and its influence on paraoxon and chlorpyrifos-oxon toxicity in rats. Toxicol

Appl Pharmacol 103(1): 66-76.

Page 175: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

153

Court MH, Duan SX, Hesse LM, Venkatakrishnan K, Greenblatt DJ (2001). Cytochrome P-450

2B6 is responsible for interindividual variability of propofol hydroxylation by human liver

microsomes. Anesthesiology 94(1): 110-119.

Cowan DT, Allan LG, Libretto SE, Griffiths P (2001). Opioid drugs: a comparative survey of

therapeutic and "street" use. Pain Med 2(3): 193-203.

Creed MC, Nobrega JN (2013). Neurobiological basis of dyskinetic effects induced by

antipsychotics: the contribution of animal models. Curr Med Chem 20(3): 389-396.

Cumming P, Yokoi F, Chen A, Deep P, Dagher A, Reutens D, et al (1999). Pharmacokinetics of

radiotracers in human plasma during positron emission tomography. Synapse 34(2): 124-134.

Cunningham CW, Mercer SL, Hassan HE, Traynor JR, Eddington ND, Coop A (2008). Opioids

and efflux transporters. Part 2: P-glycoprotein substrate activity of 3- and 6-substituted morphine

analogs. J Med Chem 51(7): 2316-2320.

D'Amour FE, Smith DL (1941). A method for determining loss of pain sensation. JPET 72(1):

74-79.

Dahl ML, Johansson I, Bertilsson L, Ingelman-Sundberg M, Sjoqvist F (1995). Ultrarapid

hydroxylation of debrisoquine in a Swedish population. Analysis of the molecular genetic basis.

J Pharmacol Exp Ther 274(1): 516-520.

Damaj MI, Meyer EM, Martin BR (2000). The antinociceptive effects of alpha7 nicotinic

agonists in an acute pain model. Neuropharmacol 39(13): 2785-2791.

Daniel WA, Haduch A, Wojcikowski J (2005). Inhibition of rat liver CYP2D in vitro and after 1-

day and long-term exposure to neuroleptics in vivo-possible involvement of different

mechanisms. Eur Neuropsychopharmacol 15(1): 103-110.

Dart RC, Severtson SG, Bucher-Bartelson B (2015). Trends in opioid analgesic abuse and

mortality in the United States. N Engl J Med 372(16): 1573-1574.

Dauchy S, Dutheil F, Weaver RJ, Chassoux F, Daumas-Duport C, Couraud PO, et al (2008).

ABC transporters, cytochromes P450 and their main transcription factors: expression at the

human blood-brain barrier. J Neurochem 107(6): 1518-1528.

Decleves X, Jacob A, Yousif S, Shawahna R, Potin S, Scherrmann JM (2011). Interplay of drug

metabolizing CYP450 enzymes and ABC transporters in the blood-brain barrier. Curr Drug

Metab 12(8): 732-741.

Desmeules J, Gascon MP, Dayer P, Magistris M (1991). Impact of environmental and genetic

factors on codeine analgesia. Eur J Clin Pharmacol 41(1): 23-26.

DeVos SL, Miller TM (2013). Direct intraventricular delivery of drugs to the rodent central

nervous system. J Vis Exp(75): e50326.

Page 176: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

154

Dewey WL, Harris LS (1971). Antinociceptive activity of the narcotic antagonists analogues and

antagonistic activity of the narcotic analgesics in rodents. J Pharmacol Exp Ther 179: 652-659.

Dhatt RK, Gudehithlu KP, Wemlinger TA, Tejwani GA, Neff NH, Hadjiconstantinou M (1995).

Preproenkephalin mRNA and methionine-enkephalin content are increased in mouse striatum

after treatment with nicotine. J Neurochem 64(4): 1878-1883.

Ding X, Kaminsky LS (2003). Human extrahepatic cytochromes P450: function in xenobiotic

metabolism and tissue-selective chemical toxicity in the respiratory and gastrointestinal tracts.

Annu Rev Pharmacol Toxicol 43: 149-173.

Dodgen TM, Hochfeld WE, Fickl H, Asfaha SM, Durandt C, Rheeder P, et al (2013).

Introduction of the AmpliChip CYP450 Test to a South African cohort: a platform comparative

prospective cohort study. BMC Med Genet 14: 20.

Dong H, Lu SJ, Zhang R, Liu DD, Zhang YZ, Song CY (2015). Effect of the CYP2D6 gene

polymorphism on postoperative analgesia of tramadol in Han nationality nephrectomy patients.

Eur J Clin Pharmacol 71(6): 681-686.

Dostrovsky JO, Deakin JF (1977). Periaqueductal grey lesions reduce morphine analgesia in the

rat. Neurosci Lett 4(2): 99-103.

Dowell D, Haegerich TM, Chou R (2016). CDC Guideline for Prescribing Opioids for Chronic

Pain - United States, 2016. MMWR Recomm Rep 65(1): 1-49.

Dumas EO, Pollack GM (2008). Opioid tolerance development: a

pharmacokinetic/pharmacodynamic perspective. AAPS J 10(4): 537-551.

Durazzo TC, Meyerhoff DJ, Murray DE (2015). Comparison of Regional Brain Perfusion Levels

in Chronically Smoking and Non-Smoking Adults. Int J Environ Res Public Health 12(7): 8198-

8213.

Dutheil F, Dauchy S, Diry M, Sazdovitch V, Cloarec O, Mellottee L, et al (2009). Xenobiotic-

metabolizing enzymes and transporters in the normal human brain: regional and cellular mapping

as a basis for putative roles in cerebral function. Drug Metab Dispos 37(7): 1528-1538.

Dutheil F, Jacob A, Dauchy S, Beaune P, Scherrmann JM, Decleves X, et al (2010). ABC

transporters and cytochromes P450 in the human central nervous system: influence on brain

pharmacokinetics and contribution to neurodegenerative disorders. Expert Opin Drug Metab

Toxicol 6(10): 1161-1174.

Dutra LM, Glantz SA (2014). Electronic cigarettes and conventional cigarette use among U.S.

adolescents: a cross-sectional study. JAMA Pediatr 168(7): 610-617.

Dvorak Z, Pavek P (2010). Regulation of drug-metabolizing cytochrome P450 enzymes by

glucocorticoids. Drug Metab Rev 42(4): 621-635.

Page 177: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

155

Eckhardt L, Woodruff SI, Elder JP (1994). A longitudinal analysis of adolescent smoking and its

correlates. J Sch Health 64(2): 67-72.

Eddy NB, Leimbach D (1953). Synthetic analgesics. II. Dithienylbutenyl- and

dithienylbutylamines. J Pharmacol Exp Ther 107(3): 385-393.

Edwards RJ, Price RJ, Watts PS, Renwick AB, Tredger JM, Boobis AR, et al (2003). Induction

of cytochrome P450 enzymes in cultured precision-cut human liver slices. Drug Metab Dispos

31(3): 282-288.

Eichelbaum M, Baur MP, Dengler HJ, Osikowska-Evers BO, Tieves G, Zekorn C, et al (1987).

Chromosomal assignment of human cytochrome P-450 (debrisoquine/sparteine type) to

chromosome 22. Br J Clin Pharmacol 23(4): 455-458.

Elbaz A, Levecque C, Clavel J, Vidal JS, Richard F, Amouyel P, et al (2004). CYP2D6

polymorphism, pesticide exposure, and Parkinson's disease. Ann Neurol 55(3): 430-434.

Elshenawy OH, Shoieb SM, Mohamed A, El-Kadi AO (2017). Clinical Implications of 20-

Hydroxyeicosatetraenoic Acid in the Kidney, Liver, Lung and Brain: An Emerging Therapeutic

Target. Pharmaceutics 9(1).

Esteller M (2011). Non-coding RNAs in human disease. Nat Rev Genet 12(12): 861-874.

Fan R, Schrott LM, Snelling S, Ndi J, Arnold T, Korneeva NL (2015). Chronic oxycodone

induces integrated stress response in rat brain. BMC Neurosci 16: 58.

Farin FM, Omiecinski CJ (1993). Regiospecific expression of cytochrome P-450s and

microsomal epoxide hydrolase in human brain tissue. J Toxicol Environ Health 40(2-3): 317-

335.

Farsalinos KE, Spyrou A, Stefopoulos C, Tsimopoulou K, Kourkoveli P, Tsiapras D, et al

(2015). Nicotine absorption from electronic cigarette use: comparison between experienced

consumers (vapers) and naive users (smokers). Sci Rep 5: 11269.

Feierman DE, Lasker JM (1996). Metabolism of fentanyl, a synthetic opioid analgesic, by human

liver microsomes. Role of CYP3A4. Drug Metab Dispos 24(9): 932-939.

Femenia T, Gomez-Galan M, Lindskog M, Magara S (2012). Dysfunctional hippocampal

activity affects emotion and cognition in mood disorders. Brain Res 1476: 58-70.

Feng XQ, Zhu LL, Zhou Q (2017). Opioid analgesics-related pharmacokinetic drug interactions:

from the perspectives of evidence based on randomized controlled trials and clinical risk

management. J Pain Res 10: 1225-1239.

Feng Y, He X, Yang Y, Chao D, Lazarus LH, Xia Y (2012). Current research on opioid receptor

function. Curr Drug Targets 13(2): 230-246.

Page 178: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

156

Ferguson CS, Miksys S, Palmour R, Tyndale RF (2011). Independent and combined effects of

ethanol self-administration and nicotine treatment on hepatic CYP2E1 in African green

monkeys. Drug Metab Dispos 39(12): 2233-2241.

Ferguson CS, Miksys S, Palmour RM, Tyndale RF (2012). Differential effects of nicotine

treatment and ethanol self-administration on CYP2A6, CYP2B6 and nicotine pharmacokinetics

in African green monkeys. J Pharmacol Exp Ther 343(3): 628-637.

Ferguson CS, Miksys S, Palmour RM, Tyndale RF (2013). Ethanol self-administration and

nicotine treatment induce brain levels of CYP2B6 and CYP2E1 in African green monkeys.

Neuropharmacol 72: 74-81.

Ferguson CS, Tyndale RF (2011). Cytochrome P450 enzymes in the brain: emerging evidence of

biological significance. Trends Pharmacol Sci 32(12): 708-714.

Ferguson SS, Barak LS, Zhang J, Caron MG (1996). G-protein-coupled receptor regulation: role

of G-protein-coupled receptor kinases and arrestins. Can J Physiol Pharmacol 74(10): 1095-

1110.

Fields HL, Margolis EB (2015). Understanding opioid reward. Trends Neurosci 38(4): 217-225.

Fischer B, Rehm J (2017). Revisiting the 'paradigm shift' in opioid use: Developments and

implications 10 years later. Drug Alcohol Rev.

Fischer B, Rehm J, Patra J, Cruz MF (2006). Changes in illicit opioid use across Canada. CMAJ

175(11): 1385.

Fishman J, Hahn EF, Norton BI (1976). N-demethylation of morphine in rat brain is localised in

sites with high opiate receptor content. Nature 261(5555): 64-65.

Florence CS, Zhou C, Luo F, Xu L (2016). The Economic Burden of Prescription Opioid

Overdose, Abuse, and Dependence in the United States, 2013. Med Care 54(10): 901-906.

Flores-Pérez J, Flores-Pérez C, Juárez-Olguín H, Lares-Asseff I, Sosa-Macías M (2004).

Determination of dextromethorphan and dextrorphan in human urine by high performance-liquid

chromatography for pharmacogenetic investigations. Chromatographia 59: 481-485.

Foster A, Mobley E, Wang Z (2007). Complicated pain management in a CYP450 2D6 poor

metabolizer. Pain Pract 7(4): 352-356.

Frenk SM, Porter KS, Paulozzi LJ (2015). Prescription opioid analgesic use among adults:

United States, 1999-2012. NCHS Data Brief(189): 1-8.

Furlan AD, Sandoval JA, Mailis-Gagnon A, Tunks E (2006). Opioids for chronic noncancer

pain: a meta-analysis of effectiveness and side effects. CMAJ 174(11): 1589-1594.

Gaedigk A (2013). Complexities of CYP2D6 gene analysis and interpretation. Int Rev Psychiatry

25(5): 534-553.

Page 179: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

157

Gaedigk A, Bradford LD, Alander SW, Leeder JS (2006). CYP2D6*36 gene arrangements

within the cyp2d6 locus: association of CYP2D6*36 with poor metabolizer status. Drug Metab

Dispos 34(4): 563-569.

Gaedigk A, Coetsee C (2008). The CYP2D6 gene locus in South African Coloureds: unique

allele distributions, novel alleles and gene arrangements. Eur J Clin Pharmacol 64(5): 465-475.

Gaedigk A, Simon SD, Pearce RE, Bradford LD, Kennedy MJ, Leeder JS (2008). The CYP2D6

activity score: translating genotype information into a qualitative measure of phenotype. Clin

Pharmacol Ther 83(2): 234-242.

Gan SH, Ismail R, Wan Adnan WA, Zulmi W (2007). Impact of CYP2D6 genetic polymorphism

on tramadol pharmacokinetics and pharmacodynamics. Mol Diagn Ther 11(3): 171-181.

Garcia-Suastegui WA, Ramos-Chavez LA, Rubio-Osornio M, Calvillo-Velasco M, Atzin-

Mendez JA, Guevara J, et al (2017). The Role of CYP2E1 in the Drug Metabolism or

Bioactivation in the Brain. Oxid Med Cell Longev 2017: 4680732.

Garcia KL, Coen K, Miksys S, Le AD, Tyndale RF (2015). Effect of Brain CYP2B Inhibition on

Brain Nicotine Levels and Nicotine Self-Administration. Neuropsychopharmacol 40(8): 1910-

1918.

Garcia KL, Le AD, Tyndale RF (2016). Brain CYP2B induction can decrease nicotine levels in

the brain. Addict Biol.

Geletko SM, Erickson AD (2000). Decreased methadone effect after ritonavir initiation.

Pharmacotherapy 20(1): 93-94.

Girdler SS, Maixner W, Naftel HA, Stewart PW, Moretz RL, Light KC (2005). Cigarette

smoking, stress-induced analgesia and pain perception in men and women. Pain 114(3): 372-

385.

Golab-Janowska M, Honczarenko K, Gawronska-Szklarz B, Potemkowski A (2007). CYP2D6

gene polymorphism as a probable risk factor for Alzheimer's disease and Parkinson's disease

with dementia. Neurol Neurochir Pol 41(2): 113-121.

Goldstein A, Sheehan P (1969). Tolerance to opioid narcotics. I. Tolerance to the "running fit"

caused by levorphanol in the mouse. J Pharmacol Exp Ther 169(2): 175-184.

Gonzalez FJ, Vilbois F, Hardwick JP, McBride OW, Nebert DW, Gelboin HV, et al (1988).

Human debrisoquine 4-hydroxylase (P450IID1): cDNA and deduced amino acid sequence and

assignment of the CYP2D locus to chromosome 22. Genomics 2(2): 174-179.

Gonzalez I, Penas-Lledo EM, Perez B, Dorado P, Alvarez M, LLerena A (2008). Relation

between CYP2D6 phenotype and genotype and personality in healthy volunteers.

Pharmacogenomics 9(7): 833-840.

Page 180: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

158

Grace PM, Maier SF, Watkins LR (2015). Opioid-induced central immune signaling:

implications for opioid analgesia. Headache 55(4): 475-489.

Green TC, Grimes Serrano JM, Licari A, Budman SH, Butler SF (2009). Women who abuse

prescription opioids: findings from the Addiction Severity Index-Multimedia Version Connect

prescription opioid database. Drug Alcohol Depend 103(1-2): 65-73.

Grobe N, Kutchan TM, Zenk MH (2012). Rat CYP2D2, not 2D1, is functionally conserved with

human CYP2D6 in endogenous morphine formation. FEBS Lett 586(13): 1749-1753.

Grond S, Sablotzki A (2004). Clinical pharmacology of tramadol. Clin Pharmacokinet 43(13):

879-923.

Gronlund J, Saari TI, Hagelberg NM, Neuvonen PJ, Olkkola KT, Laine K (2010). Exposure to

oral oxycodone is increased by concomitant inhibition of CYP2D6 and 3A4 pathways, but not by

inhibition of CYP2D6 alone. Br J Clin Pharmacol 70(1): 78-87.

Grossman ML, Basbaum AI, Fields HL (1982). Afferent and efferent connections of the rat tail

flick reflex (a model used to analyze pain control mechanisms). J Comp Neurol 206(1): 9-16.

Guarna M, Bianchi E, Bartolini A, Ghelardini C, Galeotti N, Bracci L, et al (2002). Endogenous

morphine modulates acute thermonociception in mice. J Neurochem 80(2): 271-277.

Guy GP, Jr., Zhang K, Bohm MK, Losby J, Lewis B, Young R, et al (2017). Vital Signs:

Changes in Opioid Prescribing in the United States, 2006-2015. MMWR Morb Mortal Wkly Rep

66(26): 697-704.

Haduch A, Bromek E, Daniel WA (2011). The effect of psychotropic drugs on cytochrome P450

2D (CYP2D) in rat brain. Eur J Pharmacol 651(1-3): 51-58.

Haduch A, Bromek E, Kot M, Kaminska K, Golembiowska K, Daniel WA (2015). The

cytochrome P450 2D-mediated formation of serotonin from 5-methoxytryptamine in the brain in

vivo: a microdialysis study. J Neurochem 133(1): 83-92.

Haduch A, Bromek E, Sadakierska-Chudy A, Wojcikowski J, Daniel WA (2013). The catalytic

competence of cytochrome P450 in the synthesis of serotonin from 5-methoxytryptamine in the

brain: an in vitro study. Pharmacol Res 67(1): 53-59.

Haduch A, Bromek E, Wojcikowski J, Golembiowska K, Daniel WA (2016). Melatonin

Supports CYP2D-Mediated Serotonin Synthesis in the Brain. Drug Metab Dispos 44(3): 445-

452.

Haduch A, Wojcikowski J, Daniel WA (2004). Effects of chronic treatment with classic and

newer antidepressants and neuroleptics on the activity and level of CYP2D in the rat brain. Pol J

Pharmacol 56(6): 857-862.

Hagg S, Spigset O, Dahlqvist R (2001). Influence of gender and oral contraceptives on CYP2D6

and CYP2C19 activity in healthy volunteers. Br J Clin Pharmacol 51(2): 169-173.

Page 181: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

159

Hamelin BA, Bouayad A, Methot J, Jobin J, Desgagnes P, Poirier P, et al (2000). Significant

interaction between the nonprescription antihistamine diphenhydramine and the CYP2D6

substrate metoprolol in healthy men with high or low CYP2D6 activity. Clin Pharmacol Ther

67(5): 466-477.

Hammarlund-Udenaes M, Friden M, Syvanen S, Gupta A (2008). On the rate and extent of drug

delivery to the brain. Pharm Res 25(8): 1737-1750.

Han B, Compton WM, Jones CM, Cai R (2015). Nonmedical Prescription Opioid Use and Use

Disorders Among Adults Aged 18 Through 64 Years in the United States, 2003-2013. JAMA

314(14): 1468-1478.

Hannemann F, Bichet A, Ewen KM, Bernhardt R (2007). Cytochrome P450 systems--biological

variations of electron transport chains. Biochim Biophys Acta 1770(3): 330-344.

Hansson T, Tindberg N, Ingelman-Sundberg M, Kohler C (1990). Regional distribution of

ethanol-inducible cytochrome P450 IIE1 in the rat central nervous system. Neuroscience 34(2):

451-463.

Harris GC, Wimmer M, Byrne R, Aston-Jones G (2004). Glutamate-associated plasticity in the

ventral tegmental area is necessary for conditioning environmental stimuli with morphine.

Neuroscience 129(3): 841-847.

Harris SC, Perrino PJ, Smith I, Shram MJ, Colucci SV, Bartlett C, et al (2014). Abuse potential,

pharmacokinetics, pharmacodynamics, and safety of intranasally administered crushed

oxycodone HCl abuse-deterrent controlled-release tablets in recreational opioid users. J Clin

Pharmacol 54(4): 468-477.

He F, Jiang Y, Li L (2016). The effect of naloxone treatment on opioid-induced side effects: A

meta-analysis of randomized and controlled trails. Medicine (Baltimore) 95(37): e4729.

Hedenmalm K, Sundgren M, Granberg K, Spigset O, Dahlqvist R (1997). Urinary excretion of

codeine, ethylmorphine, and their metabolites: relation to the CYP2D6 activity. Ther Drug Monit

19(6): 643-649.

Hedlund E, Gustafsson JA, Warner M (2001). Cytochrome P450 in the brain; a review. Curr

Drug Metab 2(3): 245-263.

Hedlund E, Wyss A, Kainu T, Backlund M, Kohler C, Pelto-Huikko M, et al (1996).

Cytochrome P4502D4 in the brain: specific neuronal regulation by clozapine and toluene. Mol

Pharmacol 50(2): 342-350.

Heelon MW, Meade LB (1999). Methadone withdrawal when starting an antiretroviral regimen

including nevirapine. Pharmacotherapy 19(4): 471-472.

Heiskanen T, Olkkola KT, Kalso E (1998). Effects of blocking CYP2D6 on the

pharmacokinetics and pharmacodynamics of oxycodone. Clin Pharmacol Ther 64(6): 603-611.

Page 182: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

160

Hendrickson HP, Gurley BJ, Wessinger WD (2003). Determination of dextromethorphan and its

metabolites in rat serum by liquid-liquid extraction and liquid chromatography with fluorescence

detection. J Chromatogr B Analyt Technol Biomed Life Sci 788(2): 261-268.

Hesse LM, He P, Krishnaswamy S, Hao Q, Hogan K, von Moltke LL, et al (2004).

Pharmacogenetic determinants of interindividual variability in bupropion hydroxylation by

cytochrome P4502B6 in human liver microsomes. Pharmacogenetics 14(4): 225-238.

Hildebrandt MG, Steyerberg EW, Stage KB, Passchier J, Kragh-Soerensen P, Danish University

Antidepressant G (2003). Are gender differences important for the clinical effects of

antidepressants? Am J Psychiatry 160(9): 1643-1650.

Hill MV, McMahon ML, Stucke RS, Barth RJ, Jr. (2017). Wide Variation and Excessive Dosage

of Opioid Prescriptions for Common General Surgical Procedures. Ann Surg 265(4): 709-714.

Hiroi T, Chow T, Imaoka S, Funae Y (2002). Catalytic specificity of CYP2D isoforms in rat and

human. Drug Metab Dispos 30(9): 970-976.

Hiroi T, Imaoka S, Chow T, Funae Y (1998a). Tissue distributions of CYP2D1, 2D2, 2D3 and

2D4 mRNA in rats detected by RT-PCR. Biochim Biophys Acta 1380(3): 305-312.

Hiroi T, Imaoka S, Funae Y (1998b). Dopamine formation from tyramine by CYP2D6. Biochem

Biophys Res Commun 249(3): 838-843.

Hoffman JM, Haidar CE, Wilkinson MR, Crews KR, Baker DK, Kornegay NM, et al (2014).

PG4KDS: a model for the clinical implementation of pre-emptive pharmacogenetics. Am J Med

Genet C Semin Med Genet 166C(1): 45-55.

Holland JP, Cumming P, Vasdev N (2013). PET radiopharmaceuticals for probing enzymes in

the brain. Am J Nucl Med Mol Imaging 3(3): 194-216.

Honkakoski P, Negishi M (2000). Regulation of cytochrome P450 (CYP) genes by nuclear

receptors. Biochem J 347(Pt 2): 321-337.

Houde RWW, S.L., Beaver WT (1966). Evaluation of analgesics in patients with cancer pain.

Lasagna L, ed. International encyclopedia of pharmacology and therapeutics. Pergamon Press:

Oxford, pp 59-98.

Hough LB, Nalwalk JW, Cleary RA, Phillips JG, Fang C, Yang W, et al (2014a). Deficits in

neuronal cytochrome P450 activity attenuate opioid analgesia but not opioid side effects.

European journal of pharmacology 740: 255-262.

Hough LB, Nalwalk JW, Yang J, Conroy JL, VanAlstine MA, Yang W, et al (2011). Brain P450

epoxygenase activity is required for the antinociceptive effects of improgan, a nonopioid

analgesic. Pain 152(4): 878-887.

Hough LB, Nalwalk JW, Yang W, Ding X (2014b). Significance of neuronal cytochrome P450

activity in opioid-mediated stress-induced analgesia. Brain Res 1578: 30-37.

Page 183: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

161

Hough LB, Nalwalk JW, Yang W, Ding X (2015). Neuronal cytochrome P450 activity and

opioid analgesia: relevant sites and mechanisms. Brain research 1616: 10-18.

Howard LA, Micu AL, Sellers EM, Tyndale RF (2001). Low doses of nicotine and ethanol

induce CYP2E1 and chlorzoxazone metabolism in rat liver. J Pharmacol Exp Ther 299(2): 542-

550.

Howard LA, Miksys S, Hoffmann E, Mash D, Tyndale RF (2003). Brain CYP2E1 is induced by

nicotine and ethanol in rat and is higher in smokers and alcoholics. Br J Pharmacol 138(7):

1376-1386.

Hutchinson MR, Menelaou A, Foster DJ, Coller JK, Somogyi AA (2004). CYP2D6 and

CYP3A4 involvement in the primary oxidative metabolism of hydrocodone by human liver

microsomes. Br J Clin Pharmacol 57(3): 287-297.

Iliff JJ, Jia J, Nelson J, Goyagi T, Klaus J, Alkayed NJ (2010). Epoxyeicosanoid signaling in

CNS function and disease. Prostaglandins Other Lipid Mediat 91(3-4): 68-84.

Ingelman-Sundberg M (2005). Genetic polymorphisms of cytochrome P450 2D6 (CYP2D6):

clinical consequences, evolutionary aspects and functional diversity. Pharmacogenomics J 5(1):

6-13.

Ingelman-Sundberg M, Sim SC, Gomez A, Rodriguez-Antona C (2007). Influence of

cytochrome P450 polymorphisms on drug therapies: pharmacogenetic, pharmacoepigenetic and

clinical aspects. Pharmacol Ther 116(3): 496-526.

Innocenti F, Liu W, Fackenthal D, Ramirez J, Chen P, Ye X, et al (2008). Single nucleotide

polymorphism discovery and functional assessment of variation in the UDP-

glucuronosyltransferase 2B7 gene. Pharmacogenet Genomics 18(8): 683-697.

Isojarvi JI, Tauboll E, Herzog AG (2005). Effect of antiepileptic drugs on reproductive endocrine

function in individuals with epilepsy. CNS Drugs 19(3): 207-223.

Janson AM, Fuxe K, Goldstein M (1992). Differential effects of acute and chronic nicotine

treatment on MPTP-(1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) induced degeneration of

nigrostriatal dopamine neurons in the black mouse. Clin Investig 70(3-4): 232-238.

Jeninga EH, Gurnell M, Kalkhoven E (2009). Functional implications of genetic variation in

human PPARgamma. Trends Endocrinol Metab 20(8): 380-387.

Johannes CB, Le TK, Zhou X, Johnston JA, Dworkin RH (2010). The prevalence of chronic pain

in United States adults: results of an Internet-based survey. J Pain 11(11): 1230-1239.

Johansson I, Oscarson M, Yue QY, Bertilsson L, Sjoqvist F, Ingelman-Sundberg M (1994).

Genetic analysis of the Chinese cytochrome P4502D locus: characterization of variant CYP2D6

genes present in subjects with diminished capacity for debrisoquine hydroxylation. Mol

Pharmacol 46(3): 452-459.

Page 184: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

162

Johansson I, Yue QY, Dahl ML, Heim M, Sawe J, Bertilsson L, et al (1991). Genetic analysis of

the interethnic difference between Chinese and Caucasians in the polymorphic metabolism of

debrisoquine and codeine. Eur J Clin Pharmacol 40(6): 553-556.

Johnson EA, Oldfield S, Braksator E, Gonzalez-Cuello A, Couch D, Hall KJ, et al (2006).

Agonist-selective mechanisms of mu-opioid receptor desensitization in human embryonic kidney

293 cells. Mol Pharmacol 70(2): 676-685.

Johnson JL, Rolan PE, Johnson ME, Bobrovskaya L, Williams DB, Johnson K, et al (2014).

Codeine-induced hyperalgesia and allodynia: investigating the role of glial activation. Transl

Psychiatry 4: e482.

Jolivalt C, Minn A, Vincent-Viry M, Galteau MM, Siest G (1995). Dextromethorphan O-

demethylase activity in rat brain microsomes. Neurosci Lett 187(1): 65-68.

Jones CM (2013). Heroin use and heroin use risk behaviors among nonmedical users of

prescription opioid pain relievers - United States, 2002-2004 and 2008-2010. Drug Alcohol

Depend 132(1-2): 95-100.

Jones CM, Logan J, Gladden RM, Bohm MK (2015). Vital Signs: Demographic and Substance

Use Trends Among Heroin Users - United States, 2002-2013. MMWR Morb Mortal Wkly Rep

64(26): 719-725.

Joshi M, Tyndale RF (2006a). Induction and recovery time course of rat brain CYP2E1 after

nicotine treatment. Drug Metab Dispos 34(4): 647-652.

Joshi M, Tyndale RF (2006b). Regional and cellular distribution of CYP2E1 in monkey brain

and its induction by chronic nicotine. Neuropharmacol 50(5): 568-575.

Jover R, Bort R, Gomez-Lechon MJ, Castell JV (1998). Re-expression of C/EBP alpha induces

CYP2B6, CYP2C9 and CYP2D6 genes in HepG2 cells. FEBS Lett 431(2): 227-230.

Jover R, Bort R, Gomez-Lechon MJ, Castell JV (2001). Cytochrome P450 regulation by

hepatocyte nuclear factor 4 in human hepatocytes: a study using adenovirus-mediated antisense

targeting. Hepatology 33(3): 668-675.

Jukic MM, Opel N, Strom J, Carrillo-Roa T, Miksys S, Novalen M, et al (2016). Elevated

CYP2C19 expression is associated with depressive symptoms and hippocampal homeostasis

impairment. Mol Psychiatry.

Kaiko RF, Benziger DP, Fitzmartin RD, Burke BE, Reder RF, Goldenheim PD (1996).

Pharmacokinetic-pharmacodynamic relationships of controlled-release oxycodone. Clin

Pharmacol Ther 59(1): 52-61.

Kane JK, Konu O, Ma JZ, Li MD (2004). Nicotine coregulates multiple pathways involved in

protein modification/degradation in rat brain. Brain Res Mol Brain Res 132(2): 181-191.

Kanjhan R (1995). Opioids and pain. Clin Exp Pharmacol Physiol 22(6-7): 397-403.

Page 185: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

163

Kapitzke D, Vetter I, Cabot PJ (2005). Endogenous opioid analgesia in peripheral tissues and the

clinical implications for pain control. Ther Clin Risk Manag 1(4): 279-297.

Kaplan HL, Busto UE, Baylon GJ, Cheung SW, Otton SV, Somer G, et al (1997). Inhibition of

cytochrome P450 2D6 metabolism of hydrocodone to hydromorphone does not importantly

affect abuse liability. J Pharmacol Exp Ther 281(1): 103-108.

Kassenbrock A, Vasdev N, Liang SH (2016). Selected PET Radioligands for Ion Channel Linked

Neuroreceptor Imaging: Focus on GABA, NMDA and nACh Receptors. Curr Top Med Chem

16(16): 1830-1842.

Kathiramalainathan K, Kaplan HL, Romach MK, Busto UE, Li NY, Sawe J, et al (2000).

Inhibition of cytochrome P450 2D6 modifies codeine abuse liability. J Clin Psychopharmacol

20(4): 435-444.

Katritch V, Cherezov V, Stevens RC (2013). Structure-function of the G protein-coupled

receptor superfamily. Annu Rev Pharmacol Toxicol 53: 531-556.

Keith DE, Murray SR, Zaki PA, Chu PC, Lissin DV, Kang L, et al (1996). Morphine activates

opioid receptors without causing their rapid internalization. J Biol Chem 271(32): 19021-19024.

Kelley AE, Stinus L, Iversen SD (1980). Interactions between D-ala-met-enkephalin, A10

dopaminergic neurones, and spontaneous behaviour in the rat. Behav Brain Res 1(1): 3-24.

Kerry NL, Somogyi AA, Mikus G, Bochner F (1993). Primary and secondary oxidative

metabolism of dextromethorphan. In vitro studies with female Sprague-Dawley and Dark Agouti

rat liver microsomes. Biochem Pharmacol 45(4): 833-839.

Khokhar JY, Miksys SL, Tyndale RF (2010). Rat brain CYP2B induction by nicotine is

persistent and does not involve nicotinic acetylcholine receptors. Brain Res 1348: 1-9.

Khokhar JY, Tyndale RF (2011). Drug metabolism within the brain changes drug response:

selective manipulation of brain CYP2B alters propofol effects. Neuropsychopharmacol 36(3):

692-700.

Khokhar JY, Tyndale RF (2012). Rat brain CYP2B-enzymatic activation of chlorpyrifos to the

oxon mediates cholinergic neurotoxicity. Toxicol Sci 126(2): 325-335.

Khokhar JY, Tyndale RF (2014). Intracerebroventricularly and systemically delivered inhibitor

of brain CYP2B (C8-Xanthate), even following chlorpyrifos exposure, reduces chlorpyrifos

activation and toxicity in male rats. Toxicol Sci 140(1): 49-60.

Killer N, Hock M, Gehlhaus M, Capetian P, Knoth R, Pantazis G, et al (2009). Modulation of

androgen and estrogen receptor expression by antiepileptic drugs and steroids in hippocampus of

patients with temporal lobe epilepsy. Epilepsia 50(8): 1875-1890.

Page 186: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

164

Kirchheiner J, Heesch C, Bauer S, Meisel C, Seringer A, Goldammer M, et al (2004). Impact of

the ultrarapid metabolizer genotype of cytochrome P450 2D6 on metoprolol pharmacokinetics

and pharmacodynamics. Clin Pharmacol Ther 76(4): 302-312.

Kirchheiner J, Schmidt H, Tzvetkov M, Keulen JT, Lotsch J, Roots I, et al (2007).

Pharmacokinetics of codeine and its metabolite morphine in ultra-rapid metabolizers due to

CYP2D6 duplication. Pharmacogenomics J 7(4): 257-265.

Kirchheiner J, Seeringer A, Godoy AL, Ohmle B, Maier C, Beschoner P, et al (2011). CYP2D6

in the brain: genotype effects on resting brain perfusion. Mol Psychiatry 16(3): 237, 333-241.

Kiyotani K, Mushiroda T, Sasa M, Bando Y, Sumitomo I, Hosono N, et al (2008). Impact of

CYP2D6*10 on recurrence-free survival in breast cancer patients receiving adjuvant tamoxifen

therapy. Cancer Sci 99(5): 995-999.

Klimas R, Witticke D, El Fallah S, Mikus G (2013). Contribution of oxycodone and its

metabolites to the overall analgesic effect after oxycodone administration. Expert Opin Drug

Metab Toxicol 9(5): 517-528.

Koch T, Hollt V (2008). Role of receptor internalization in opioid tolerance and dependence.

Pharmacol Ther 117(2): 199-206.

Koch T, Kroslak T, Averbeck M, Mayer P, Schroder H, Raulf E, et al (2000). Allelic variation

S268P of the human mu-opioid receptor affects both desensitization and G protein coupling. Mol

Pharmacol 58(2): 328-334.

Koch T, Seifert A, Wu DF, Rankovic M, Kraus J, Borner C, et al (2009). mu-opioid receptor-

stimulated synthesis of reactive oxygen species is mediated via phospholipase D2. J Neurochem

110(4): 1288-1296.

Koenigs LL, Trager WF (1998). Mechanism-based inactivation of cytochrome P450 2B1 by 8-

methoxypsoralen and several other furanocoumarins. Biochemistry 37(38): 13184-13193.

Kohlrausch FB, Gama CS, Lobato MI, Belmonte-de-Abreu P, Gesteira A, Barros F, et al (2009).

Molecular diversity at the CYP2D6 locus in healthy and schizophrenic southern Brazilians.

Pharmacogenomics 10(9): 1457-1466.

Komori M (1993). A novel P450 expressed at the high level in rat brain. Biochem Biophys Res

Commun 196(2): 721-728.

Koyano S, Saito Y, Fukushima-Uesaka H, Ishida S, Ozawa S, Kamatani N, et al (2005).

Functional analysis of six human aryl hydrocarbon receptor variants in a Japanese population.

Drug Metab Dispos 33(8): 1254-1260.

Krishna DR, Klotz U (1994). Extrahepatic metabolism of drugs in humans. Clin Pharmacokinet

26(2): 144-160.

Page 187: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

165

Kubota T, Chiba K, Ishizaki T (1996). Genotyping of S-mephenytoin 4'-hydroxylation in an

extended Japanese population. Clin Pharmacol Ther 60(6): 661-666.

Lacy RT, Strickland JC, Feinstein MA, Robinson AM, Smith MA (2016). The effects of sex,

estrous cycle, and social contact on cocaine and heroin self-administration in rats.

Psychopharmacology (Berl) 233(17): 3201-3210.

Lake S, Milloy MJ, Dong H, Hayashi K, Wood E, Kerr T, et al (2016). Initiation into

prescription opioid injection and associated trends in heroin use among people who use illicit

drugs. Drug Alcohol Depend 169: 73-79.

Lalovic B, Kharasch E, Hoffer C, Risler L, Liu-Chen LY, Shen DD (2006). Pharmacokinetics

and pharmacodynamics of oral oxycodone in healthy human subjects: role of circulating active

metabolites. Clin Pharmacol Ther 79(5): 461-479.

Lalovic B, Phillips B, Risler LL, Howald W, Shen DD (2004). Quantitative contribution of

CYP2D6 and CYP3A to oxycodone metabolism in human liver and intestinal microsomes. Drug

Metab Dispos 32(4): 447-454.

Lamba J, Lamba V, Schuetz E (2005). Genetic variants of PXR (NR1I2) and CAR (NR1I3) and

their implications in drug metabolism and pharmacogenetics. Curr Drug Metab 6(4): 369-383.

Lamba JK, Lamba V, Yasuda K, Lin YS, Assem M, Thompson E, et al (2004a). Expression of

constitutive androstane receptor splice variants in human tissues and their functional

consequences. J Pharmacol Exp Ther 311(2): 811-821.

Lamba V, Yasuda K, Lamba JK, Assem M, Davila J, Strom S, et al (2004b). PXR (NR1I2):

splice variants in human tissues, including brain, and identification of neurosteroids and nicotine

as PXR activators. Toxicol Appl Pharmacol 199(3): 251-265.

Laugesen S, Enggaard TP, Pedersen RS, Sindrup SH, Brosen K (2005). Paroxetine, a

cytochrome P450 2D6 inhibitor, diminishes the stereoselective O-demethylation and reduces the

hypoalgesic effect of tramadol. Clin Pharmacol Ther 77(4): 312-323.

Law PY, Loh HH (1999). Regulation of opioid receptor activities. J Pharmacol Exp Ther 289(2):

607-624.

Le Bars D, Gozariu M, Cadden SW (2001). Animal models of nociception. Pharmacol Rev

53(4): 597-652.

Le Merrer J, Becker JA, Befort K, Kieffer BL (2009). Reward processing by the opioid system in

the brain. Physiol Rev 89(4): 1379-1412.

Lee AJ, Cai MX, Thomas PE, Conney AH, Zhu BT (2003). Characterization of the oxidative

metabolites of 17beta-estradiol and estrone formed by 15 selectively expressed human

cytochrome p450 isoforms. Endocrinology 144(8): 3382-3398.

Page 188: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

166

Lee AM, Joshi M, Yue J, Tyndale RF (2006a). Phenobarbital induces monkey brain CYP2E1

protein but not hepatic CYP2E1, in vitro or in vivo chlorzoxazone metabolism. Eur J Pharmacol

552(1-3): 151-158.

Lee AM, Miksys S, Palmour R, Tyndale RF (2006b). CYP2B6 is expressed in African Green

monkey brain and is induced by chronic nicotine treatment. Neuropharmacology 50(4): 441-450.

Lee AM, Miksys S, Tyndale RF (2006c). Phenobarbital increases monkey in vivo nicotine

disposition and induces liver and brain CYP2B6 protein. Br J Pharmacol 148(6): 786-794.

Lee AM, Yue J, Tyndale RF (2006d). In vivo and in vitro characterization of chlorzoxazone

metabolism and hepatic CYP2E1 levels in African Green monkeys: induction by chronic

nicotine treatment. Drug Metab Dispos 34(9): 1508-1515.

Lee CW, Ho IK (2013). Sex differences in opioid analgesia and addiction: interactions among

opioid receptors and estrogen receptors. Mol Pain 9: 45.

Lee D, Chronister CW, Broussard WA, Utley-Bobak SR, Schultz DL, Vega RS, et al (2016).

Illicit Fentanyl-Related Fatalities in Florida: Toxicological Findings. J Anal Toxicol 40(8): 588-

594.

Lelas S, Wegert S, Otton SV, Sellers EM, France CP (1999). Inhibitors of cytochrome P450

differentially modify discriminative-stimulus and antinociceptive effects of hydrocodone and

hydromorphone in rhesus monkeys. Drug Alcohol Depend 54(3): 239-249.

Lerman C, Shields PG, Wileyto EP, Audrain J, Pinto A, Hawk L, et al (2002). Pharmacogenetic

investigation of smoking cessation treatment. Pharmacogenetics 12(8): 627-634.

Li J, Xie M, Wang X, Ouyang X, Wan Y, Dong G, et al (2015a). Sex hormones regulate cerebral

drug metabolism via brain miRNAs: down-regulation of brain CYP2D by androgens reduces the

analgesic effects of tramadol. Br J Pharmacol 172(19): 4639-4654.

Li MH, Suchland KL, Ingram SL (2015b). GABAergic transmission and enhanced modulation

by opioids and endocannabinoids in adult rat rostral ventromedial medulla. J Physiol 593(1):

217-230.

Lim HS, Ju Lee H, Seok Lee K, Sook Lee E, Jang IJ, Ro J (2007). Clinical implications of

CYP2D6 genotypes predictive of tamoxifen pharmacokinetics in metastatic breast cancer. J Clin

Oncol 25(25): 3837-3845.

Lim JS, Chen XA, Singh O, Yap YS, Ng RC, Wong NS, et al (2011). Impact of CYP2D6,

CYP3A5, CYP2C9 and CYP2C19 polymorphisms on tamoxifen pharmacokinetics in Asian

breast cancer patients. Br J Clin Pharmacol 71(5): 737-750.

Lin JH, Lu AY (1998). Inhibition and induction of cytochrome P450 and the clinical

implications. Clin Pharmacokinet 35(5): 361-390.

Page 189: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

167

Lin LY, Kumagai Y, Cho AK (1992). Enzymatic and chemical demethylenation of

(methylenedioxy)amphetamine and (methylenedioxy)methamphetamine by rat brain

microsomes. Chem Res Toxicol 5(3): 401-406.

Linares OA, Daly D, Stefanovski D, Boston RC (2014). The CYP2D6 gene determines

oxycodone's phenotype-specific addictive potential: implications for addiction prevention and

treatment. Med Hypotheses 82(3): 390-394.

Liu M, Zhu Q, Wu J, Yu X, Hu M, Xie X, et al (2017). Glutamate affects the production of

epoxyeicosanoids within the brain: The up-regulation of brain CYP2J through the MAPK-CREB

signaling pathway. Toxicology 381: 31-38.

Llerena A, Dorado P, Ramirez R, Gonzalez I, Alvarez M, Penas-Lledo EM, et al (2012).

CYP2D6 genotype and debrisoquine hydroxylation phenotype in Cubans and Nicaraguans.

Pharmacogenomics J 12(2): 176-183.

Lockwood JT, Remington G (2015). Emerging drugs for antipsychotic-induced tardive

dyskinesia: investigational drugs in Phase II and Phase III clinical trials. Expert Opin Emerg

Drugs 20(3): 407-421.

Lohman D, Schleifer R, Amon JJ (2010). Access to pain treatment as a human right. BMC Med

8: 8.

Lotsch J, Skarke C, Liefhold J, Geisslinger G (2004). Genetic predictors of the clinical response

to opioid analgesics: clinical utility and future perspectives. Clin Pharmacokinet 43(14): 983-

1013.

Loyd DR, Murphy AZ (2009). The role of the periaqueductal gray in the modulation of pain in

males and females: are the anatomy and physiology really that different? Neural Plast 2009:

462879.

Lundqvist E, Johansson I, Ingelman-Sundberg M (1999). Genetic mechanisms for duplication

and multiduplication of the human CYP2D6 gene and methods for detection of duplicated

CYP2D6 genes. Gene 226(2): 327-338.

Luscher C, Slesinger PA (2010). Emerging roles for G protein-gated inwardly rectifying

potassium (GIRK) channels in health and disease. Nat Rev Neurosci 11(5): 301-315.

Ma QP, Han JS (1991). Naloxone blocks the release of opioid peptides in periaqueductal gray

and N. accumbens induced by intra-amygdaloid injection of morphine. Peptides 12(6): 1235-

1238.

Mackey I, Sharifzadeh Y, Sturgeon J, Mackey S (2016). Cigarette smoking is a predictor of

opioid use in a tertiary care pain clinic sample: a Collaborative Health Outcomes Information

Registry (CHOIR) study. The Journal of Pain 17(4): S15.

Mague SD, Blendy JA (2010). OPRM1 SNP (A118G): involvement in disease development,

treatment response, and animal models. Drug Alcohol Depend 108(3): 172-182.

Page 190: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

168

Manchikanti L (2008). Health care reform in the United States: radical surgery needed now more

than ever. Pain Physician 11(1): 13-42.

Manda VK, Mittapalli RK, Bohn KA, Adkins CE, Lockman PR (2010). Nicotine and cotinine

increases the brain penetration of saquinavir in rat. J Neurochem 115(6): 1495-1507.

Mann A, Miksys S, Lee A, Mash DC, Tyndale RF (2008). Induction of the drug metabolizing

enzyme CYP2D in monkey brain by chronic nicotine treatment. Neuropharmacol 55(7): 1147-

1155.

Mann A, Miksys SL, Gaedigk A, Kish SJ, Mash DC, Tyndale RF (2012). The neuroprotective

enzyme CYP2D6 increases in the brain with age and is lower in Parkinson's disease patients.

Neurobiol Aging 33(9): 2160-2171.

Mann A, Tyndale RF (2010). Cytochrome P450 2D6 enzyme neuroprotects against 1-methyl-4-

phenylpyridinium toxicity in SH-SY5Y neuronal cells. Eur J Neurosci 31(7): 1185-1193.

Mansour A, Fox CA, Akil H, Watson SJ (1995). Opioid-receptor mRNA expression in the rat

CNS: anatomical and functional implications. Trends Neurosci 18(1): 22-29.

Margolis EB, Hjelmstad GO, Bonci A, Fields HL (2005). Both kappa and mu opioid agonists

inhibit glutamatergic input to ventral tegmental area neurons. J Neurophysiol 93(6): 3086-3093.

Margolis EB, Toy B, Himmels P, Morales M, Fields HL (2012). Identification of rat ventral

tegmental area GABAergic neurons. PLoS One 7(7): e42365.

Matoh N, Tanaka S, Takehashi M, Banasik M, Stedeford T, Masliah E, et al (2003).

Overexpression of CYP2D6 attenuates the toxicity of MPP+ in actively dividing and

differentiated PC12 cells. Gene Expr 11(3-4): 117-124.

Mattioli TA, Milne B, Cahill CM (2010). Ultra-low dose naltrexone attenuates chronic

morphine-induced gliosis in rats. Mol Pain 6: 22.

Mavrikaki M, Pravetoni M, Page S, Potter D, Chartoff E (2017). Oxycodone self-administration

in male and female rats. Psychopharmacology (Berl) 234(6): 977-987.

Mazer-Amirshahi M, Mullins PM, Rasooly I, van den Anker J, Pines JM (2014). Rising opioid

prescribing in adult U.S. emergency department visits: 2001-2010. Acad Emerg Med 21(3): 236-

243.

McFadyen C, Melvin WT, Murray GI (1998). Regional distribution of individual forms of

cytochrome P450 mRNA in normal adult human brain. Biochem Pharmacol 55(6): 825-830.

McLaughlin LA, Paine MJ, Kemp CA, Marechal JD, Flanagan JU, Ward CJ, et al (2005). Why

is quinidine an inhibitor of cytochrome P450 2D6? The role of key active-site residues in

quinidine binding. J Biol Chem 280(46): 38617-38624.

Page 191: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

169

McMillan DM, Tyndale RF (2015). Nicotine Increases Codeine Analgesia Through the

Induction of Brain CYP2D and Central Activation of Codeine to Morphine.

Neuropsychopharmacol 40(7): 1804-1812.

McMillan DM, Tyndale RF (2017a). CYP-mediated drug metabolism in the brain impacts drug

response. Pharmacol Ther.

McMillan DM, Tyndale RF (2017b). Inducing rat brain CYP2D with nicotine increases the rate

of codeine tolerance; predicting the rate of tolerance from acute analgesic response. Biochem

Pharmacol.

McPherson J, Rivero G, Baptist M, Llorente J, Al-Sabah S, Krasel C, et al (2010). mu-opioid

receptors: correlation of agonist efficacy for signalling with ability to activate internalization.

Mol Pharmacol 78(4): 756-766.

Mercer SL, Coop A (2011). Opioid analgesics and P-glycoprotein efflux transporters: a potential

systems-level contribution to analgesic tolerance. Curr Top Med Chem 11(9): 1157-1164.

Metcalf MD, Rosicky AD, Hassan HE, Eddington ND, Coop A, Cunningham CW, et al (2014).

Opioids and efflux transporters. Part 4: influence of N-substitution on P-glycoprotein substrate

activity of noroxymorphone analogues. Bioorg Med Chem Lett 24(15): 3592-3595.

Meyer RP, Gehlhaus M, Knoth R, Volk B (2007). Expression and function of cytochrome p450

in brain drug metabolism. Curr Drug Metab 8(4): 297-306.

Meyer RP, Hagemeyer CE, Knoth R, Kaufmann MR, Volk B (2006). Anti-epileptic drug

phenytoin enhances androgen metabolism and androgen receptor expression in murine

hippocampus. J Neurochem 96(2): 460-472.

Michels R, Marzuk PM (1993). Progress in psychiatry (1). N Engl J Med 329(8): 552-560.

Micu AL, Miksys S, Sellers EM, Koop DR, Tyndale RF (2003). Rat hepatic CYP2E1 is induced

by very low nicotine doses: an investigation of induction, time course, dose response, and

mechanism. J Pharmacol Exp Ther 306(3): 941-947.

Miech R, Johnston L, O'Malley PM, Keyes KM, Heard K (2015). Prescription Opioids in

Adolescence and Future Opioid Misuse. Pediatrics 136(5): e1169-1177.

Miksys S, Hoffmann E, Tyndale RF (2000a). Regional and cellular induction of nicotine-

metabolizing CYP2B1 in rat brain by chronic nicotine treatment. Biochem Pharmacol 59(12):

1501-1511.

Miksys S, Lerman C, Shields PG, Mash DC, Tyndale RF (2003). Smoking, alcoholism and

genetic polymorphisms alter CYP2B6 levels in human brain. Neuropharmacol 45(1): 122-132.

Miksys S, Rao Y, Hoffmann E, Mash DC, Tyndale RF (2002). Regional and cellular expression

of CYP2D6 in human brain: higher levels in alcoholics. J Neurochem 82(6): 1376-1387.

Page 192: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

170

Miksys S, Rao Y, Sellers EM, Kwan M, Mendis D, Tyndale RF (2000b). Regional and cellular

distribution of CYP2D subfamily members in rat brain. Xenobiotica 30(6): 547-564.

Miksys S, Tyndale RF (2004). The unique regulation of brain cytochrome P450 2 (CYP2) family

enzymes by drugs and genetics. Drug Metab Rev 36(2): 313-333.

Miksys S, Tyndale RF (2006). Nicotine induces brain CYP enzymes: relevance to Parkinson's

disease. J Neural Transm Suppl(70): 177-180.

Miksys S, Tyndale RF (2009). Brain drug-metabolizing cytochrome P450 enzymes are active in

vivo, demonstrated by mechanism-based enzyme inhibition. Neuropsychopharmacol 34(3): 634-

640.

Miksys S, Tyndale RF (2013). Cytochrome P450-mediated drug metabolism in the brain. J

Psychiatry Neurosci 38(3): 152-163.

Miksys S, Wadji FB, Tolledo EC, Remington G, Nobrega JN, Tyndale RF (2017). Rat brain

CYP2D enzymatic metabolism alters acute and chronic haloperidol side-effects by different

mechanisms. Prog Neuropsychopharmacol Biol Psychiatry 78: 140-148.

Miksys SL, Cheung C, Gonzalez FJ, Tyndale RF (2005). Human CYP2D6 and mouse CYP2Ds:

organ distribution in a humanized mouse model. Drug Metab Dispos 33(10): 1495-1502.

Miksys SL, Tyndale RF (2002). Drug-metabolizing cytochrome P450s in the brain. J Psychiatry

Neurosci 27(6): 406-415.

Mikus G, Trausch B, Rodewald C, Hofmann U, Richter K, Gramatte T, et al (1997). Effect of

codeine on gastrointestinal motility in relation to CYP2D6 phenotype. Clin Pharmacol Ther

61(4): 459-466.

Miller LG (1990). Cigarettes and drug therapy: pharmacokinetic and pharmacodynamic

considerations. Clin Pharm 9(2): 125-135.

Miller RT, Miksys S, Hoffmann E, Tyndale RF (2014). Ethanol self-administration and nicotine

treatment increase brain levels of CYP2D in African green monkeys. Br J Pharmacol 171(12):

3077-3088.

Mo SL, Zhou ZW, Yang LP, Wei MQ, Zhou SF (2009). New insights into the structural features

and functional relevance of human cytochrome P450 2C9. Part I. Curr Drug Metab 10(10):

1075-1126.

Montane Jaime LK, Lalla A, Steimer W, Gaedigk A (2013). Characterization of the CYP2D6

gene locus and metabolic activity in Indo- and Afro-Trinidadians: discovery of novel allelic

variants. Pharmacogenomics 14(3): 261-276.

Monte AA, Heard KJ, Campbell J, Hamamura D, Weinshilboum RM, Vasiliou V (2014). The

effect of CYP2D6 drug-drug interactions on hydrocodone effectiveness. Acad Emerg Med 21(8):

879-885.

Page 193: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

171

Moreno S, Farioli-Vecchioli S, Ceru MP (2004). Immunolocalization of peroxisome proliferator-

activated receptors and retinoid X receptors in the adult rat CNS. Neuroscience 123(1): 131-145.

Mrkusich EM, Kivell BM, Miller JH, Day DJ (2004). Abundant expression of mu and delta

opioid receptor mRNA and protein in the cerebellum of the fetal, neonatal, and adult rat. Brain

Res Dev Brain Res 148(2): 213-222.

Na S, Li J, Zhang H, Li Y, Yang Z, Zhong Y, et al (2017). The induction of cytochrome P450

2E1 by ethanol leads to the loss of synaptic proteins via PPARalpha down-regulation. Toxicology

385: 18-27.

Napolitano G, Stingl JC, Schmid M, Viviani R (2017). Predicting CYP2D6 phenotype from

resting brain perfusion images by gradient boosting. Psychiatry Res 259: 16-24.

Narimatsu S, Arai T, Masubuchi Y, Horie T, Hosokawa M, Ueno K, et al (2001). Inactivation of

rat cytochrome P450 2D enzyme by a further metabolite of 4-hydroxypropranolol, the major and

active metabolite of propranolol. Biol Pharm Bull 24(9): 988-994.

Narita M, Khotib J, Suzuki M, Ozaki S, Yajima Y, Suzuki T (2003). Heterologous mu-opioid

receptor adaptation by repeated stimulation of kappa-opioid receptor: up-regulation of G-protein

activation and antinociception. J Neurochem 85(5): 1171-1179.

Nelson AJ, Thur KE, Horsley RR, Spicer C, Marsden CA, Cassaday HJ (2011). Reduced

dopamine function within the medial shell of the nucleus accumbens enhances latent inhibition.

Pharmacol Biochem Behav 98(1): 1-7.

Nelson DR (2006). Cytochrome P450 nomenclature, 2004. Methods Mol Biol 320: 1-10.

Nevo I, Avidor-Reiss T, Levy R, Bayewitch M, Heldman E, Vogel Z (1998). Regulation of

adenylyl cyclase isozymes on acute and chronic activation of inhibitory receptors. Mol

Pharmacol 54(2): 419-426.

Nicholson KL, Hayes BA, Balster RL (1999). Evaluation of the reinforcing properties and

phencyclidine-like discriminative stimulus effects of dextromethorphan and dextrorphan in rats

and rhesus monkeys. Psychopharmacology (Berl) 146(1): 49-59.

Nieminen TH, Hagelberg NM, Saari TI, Neuvonen M, Neuvonen PJ, Laine K, et al (2010).

Grapefruit juice enhances the exposure to oral oxycodone. Basic Clin Pharmacol Toxicol 107(4):

782-788.

Nishimura M, Naito S, Yokoi T (2004). Tissue-specific mRNA expression profiles of human

nuclear receptor subfamilies. Drug Metab Pharmacokinet 19(2): 135-149.

Niwa T, Hiroi T, Tsuzuki D, Yamamoto S, Narimatsu S, Fukuda T, et al (2004). Effect of

genetic polymorphism on the metabolism of endogenous neuroactive substances, progesterone

and p-tyramine, catalyzed by CYP2D6. Brain Res Mol Brain Res 129(1-2): 117-123.

Page 194: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

172

Norris PJ, Hardwick JP, Emson PC (1996). Regional distribution of cytochrome P450 2D1 in the

rat central nervous system. J Comp Neurol 366(2): 244-258.

NSDUH (2016). 2015 National Survey on Drug Use and Health, Rockville, MD. Substance

Abuse and Mental Health Services Administration.

NSDUH (2017). 2016 National Survey on Drug Use and Health, Rockville, MD. Substance

Abuse and Mental Health Services Administration.

O'Connor EC, Chapman K, Butler P, Mead AN (2011). The predictive validity of the rat self-

administration model for abuse liability. Neurosci Biobehav Rev 35(3): 912-938.

Oguri K, Hanioka N, Yoshimura H (1990). Species differences in metabolism of codeine:

urinary excretion of codeine glucuronide, morphine-3-glucuronide and morphine-6-glucuronide

in mice, rats, guinea pigs and rabbits. Xenobiotica 20(7): 683-688.

Oldendorf WH, Hyman S, Braun L, Oldendorf SZ (1972). Blood-brain barrier: penetration of

morphine, codeine, heroin, and methadone after carotid injection. Science 178(4064): 984-986.

Ossipov MH, Dussor GO, Porreca F (2010). Central modulation of pain. J Clin Invest 120(11):

3779-3787.

Pan X, Jeong H (2015). Estrogen-Induced Cholestasis Leads to Repressed CYP2D6 Expression

in CYP2D6-Humanized Mice. Mol Pharmacol 88(1): 106-112.

Pan X, Ning M, Jeong H (2017). Transcriptional Regulation of CYP2D6 Expression. Drug

Metab Dispos 45(1): 42-48.

Parain K, Hapdey C, Rousselet E, Marchand V, Dumery B, Hirsch EC (2003). Cigarette smoke

and nicotine protect dopaminergic neurons against the 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyridine Parkinsonian toxin. Brain Res 984(1-2): 224-232.

Pardridge WM (2007). shRNA and siRNA delivery to the brain. Adv Drug Deliv Rev 59(2-3):

141-152.

Parsons CG, Quack G, Bresink I, Baran L, Przegalinski E, Kostowski W, et al (1995).

Comparison of the potency, kinetics and voltage-dependency of a series of uncompetitive

NMDA receptor antagonists in vitro with anticonvulsive and motor impairment activity in vivo.

Neuropharmacol 34(10): 1239-1258.

Pathan H, Williams J (2012). Basic opioid pharmacology: an update. Br J Pain 6(1): 11-16.

Paul SM, Axelrod J, Diliberto EJ, Jr. (1977). Catechol estrogen-forming enzyme of brain:

demonstration of a cytochrome p450 monooxygenase. Endocrinology 101(5): 1604-1610.

Peckham EM, Traynor JR (2006). Comparison of the antinociceptive response to morphine and

morphine-like compounds in male and female Sprague-Dawley rats. J Pharmacol Exp Ther

316(3): 1195-1201.

Page 195: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

173

Penas-Lledo EM, Llerena A (2014). CYP2D6 variation, behaviour and psychopathology:

implications for pharmacogenomics-guided clinical trials. Br J Clin Pharmacol 77(4): 673-683.

Peng J, Sarkar S, Chang SL (2012). Opioid receptor expression in human brain and peripheral

tissues using absolute quantitative real-time RT-PCR. Drug Alcohol Depend 124(3): 223-228.

Pepper JM, Lennard MS, Tucker GT, Woods HF (1991). Effect of steroids on the cytochrome

P4502D6-catalysed metabolism of metoprolol. Pharmacogenetics 1(2): 119-122.

Perrino PJ, Colucci SV, Apseloff G, Harris SC (2013). Pharmacokinetics, tolerability, and safety

of intranasal administration of reformulated OxyContin((R)) tablets compared with original

OxyContin ((R)) tablets in healthy adults. Clin Drug Investig 33(6): 441-449.

Persson A, Sim SC, Virding S, Onishchenko N, Schulte G, Ingelman-Sundberg M (2014).

Decreased hippocampal volume and increased anxiety in a transgenic mouse model expressing

the human CYP2C19 gene. Mol Psychiatry 19(6): 733-741.

Pert CB, Snyder SH (1973). Properties of opiate-receptor binding in rat brain. Proc Natl Acad

Sci U S A 70(8): 2243-2247.

Petersen SL, Curran MA, Marconi SA, Carpenter CD, Lubbers LS, McAbee MD (2000).

Distribution of mRNAs encoding the arylhydrocarbon receptor, arylhydrocarbon receptor

nuclear translocator, and arylhydrocarbon receptor nuclear translocator-2 in the rat brain and

brainstem. J Comp Neurol 427(3): 428-439.

Pezalla EJ, Rosen D, Erensen JG, Haddox JD, Mayne TJ (2017). Secular trends in opioid

prescribing in the USA. J Pain Res 10: 383-387.

Prakash A, Markham A (2000). Metoprolol: a review of its use in chronic heart failure. Drugs

60(3): 647-678.

Pratt PF, Medhora M, Harder DR (2004). Mechanisms regulating cerebral blood flow as

therapeutic targets. Curr Opin Investig Drugs 5(9): 952-956.

Qin S, Shen L, Zhang A, Xie J, Shen W, Chen L, et al (2008). Systematic polymorphism

analysis of the CYP2D6 gene in four different geographical Han populations in mainland China.

Genomics 92(3): 152-158.

Quik M, Huang LZ, Parameswaran N, Bordia T, Campos C, Perez XA (2009). Multiple roles for

nicotine in Parkinson's disease. Biochem Pharmacol 78(7): 677-685.

Quik M, Parameswaran N, McCallum SE, Bordia T, Bao S, McCormack A, et al (2006). Chronic

oral nicotine treatment protects against striatal degeneration in MPTP-treated primates. J

Neurochem 98(6): 1866-1875.

Rao H, Gillihan SJ, Wang J, Korczykowski M, Sankoorikal GM, Kaercher KA, et al (2007).

Genetic variation in serotonin transporter alters resting brain function in healthy individuals. Biol

Psychiatry 62(6): 600-606.

Page 196: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

174

Richter KP, Ahluwalia JS (2000). A case for addressing cigarette use in methadone and other

opioid treatment programs. J Addict Dis 19(4): 35-52.

Rifkind AB (2006). CYP1A in TCDD toxicity and in physiology-with particular reference to

CYP dependent arachidonic acid metabolism and other endogenous substrates. Drug metabolism

reviews 38(1-2): 291-335.

Riley J, Eisenberg E, Muller-Schwefe G, Drewes AM, Arendt-Nielsen L (2008). Oxycodone: a

review of its use in the management of pain. Curr Med Res Opin 24(1): 175-192.

Roberts BJ, Song BJ, Soh Y, Park SS, Shoaf SE (1995). Ethanol induces CYP2E1 by protein

stabilization. Role of ubiquitin conjugation in the rapid degradation of CYP2E1. J Biol Chem

270(50): 29632-29635.

Roques BP, Fournie-Zaluski MC, Wurm M (2012). Inhibiting the breakdown of endogenous

opioids and cannabinoids to alleviate pain. Nat Rev Drug Discov 11(4): 292-310.

Rosenblum A, Marsch LA, Joseph H, Portenoy RK (2008). Opioids and the treatment of chronic

pain: controversies, current status, and future directions. Exp Clin Psychopharmacol 16(5): 405-

416.

Roth ME, Casimir AG, Carroll ME (2002). Influence of estrogen in the acquisition of

intravenously self-administered heroin in female rats. Pharmacol Biochem Behav 72(1-2): 313-

318.

Roussin A, Bouyssi A, Pouche L, Pourcel L, Lapeyre-Mestre M (2013). Misuse and dependence

on non-prescription codeine analgesics or sedative H1 antihistamines by adults: a cross-sectional

investigation in France. PLoS One 8(10): e76499.

Rowland K, Yeo WW, Ellis SW, Chadwick IG, Haq I, Lennard MS, et al (1994). Inhibition of

CYP2D6 activity by treatment with propranolol and the role of 4-hydroxy propranolol. Br J Clin

Pharmacol 38(1): 9-14.

Rowland P, Blaney FE, Smyth MG, Jones JJ, Leydon VR, Oxbrow AK, et al (2006). Crystal

structure of human cytochrome P450 2D6. J Biol Chem 281(11): 7614-7622.

Rudd RA, Seth P, David F, Scholl L (2016). Increases in Drug and Opioid-Involved Overdose

Deaths - United States, 2010-2015. MMWR Morb Mortal Wkly Rep 65(5051): 1445-1452.

Sadiq MW, Bostrom E, Keizer R, Bjorkman S, Hammarlund-Udenaes M (2013). Oxymorphone

active uptake at the blood-brain barrier and population modeling of its pharmacokinetic-

pharmacodynamic relationship. J Pharm Sci 102(9): 3320-3331.

Saito M, Yasui-Furukori N, Nakagami T, Furukori H, Kaneko S (2005). Dose-dependent

interaction of paroxetine with risperidone in schizophrenic patients. J Clin Psychopharmacol

25(6): 527-532.

Page 197: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

175

Sasame HA, Ames MM, Nelson SD (1977). Cytochrome P-450 and NADPH cytochrome c

reductase in rat brain: formation of catechols and reactive catechol metabolites. Biochem Biophys

Res Commun 78(3): 919-926.

Schilter B, Andersen MR, Acharya C, Omiecinski CJ (2000). Activation of cytochrome P450

gene expression in the rat brain by phenobarbital-like inducers. J Pharmacol Exp Ther 294(3):

916-922.

Schilter B, Omiecinski CJ (1993). Regional distribution and expression modulation of

cytochrome P-450 and epoxide hydrolase mRNAs in the rat brain. Mol Pharmacol 44(5): 990-

996.

Schmidt BL, Tambeli CH, Gear RW, Levine JD (2001). Nicotine withdrawal hyperalgesia and

opioid-mediated analgesia depend on nicotine receptors in nucleus accumbens. Neuroscience

106(1): 129-136.

Schmidt H, Vormfelde SV, Walchner-Bonjean M, Klinder K, Freudenthaler S, Gleiter CH, et al

(2003). The role of active metabolites in dihydrocodeine effects. Int J Clin Pharmacol Ther

41(3): 95-106.

Schoedel KA, Sellers EM, Tyndale RF (2001). Induction of CYP2B1/2 and nicotine metabolism

by ethanol in rat liver but not rat brain. Biochem Pharmacol 62(8): 1025-1036.

Schopflocher D, Taenzer P, Jovey R (2011). The prevalence of chronic pain in Canada. Pain Res

Manag 16(6): 445-450.

Schroth W, Goetz MP, Hamann U, Fasching PA, Schmidt M, Winter S, et al (2009). Association

between CYP2D6 polymorphisms and outcomes among women with early stage breast cancer

treated with tamoxifen. JAMA 302(13): 1429-1436.

Sharma SK, Klee WA, Nirenberg M (1975a). Dual regulation of adenylate cyclase accounts for

narcotic dependence and tolerance. Proc Natl Acad Sci U S A 72(8): 3092-3096.

Sharma SK, Nirenberg M, Klee WA (1975b). Morphine receptors as regulators of adenylate

cyclase activity. Proc Natl Acad Sci U S A 72(2): 590-594.

Shulman GL, Fiez JA, Corbetta M, Buckner RL, Miezin FM, Raichle ME, et al (1997). Common

Blood Flow Changes across Visual Tasks: II. Decreases in Cerebral Cortex. J Cogn Neurosci

9(5): 648-663.

Siegel S, Hinson RE, Krank MD, McCully J (1982). Heroin "overdose" death: contribution of

drug-associated environmental cues. Science 216(4544): 436-437.

Siegle I, Fritz P, Eckhardt K, Zanger UM, Eichelbaum M (2001). Cellular localization and

regional distribution of CYP2D6 mRNA and protein expression in human brain.

Pharmacogenetics 11(3): 237-245.

Page 198: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

176

Sim SC, Nordin L, Andersson TM, Virding S, Olsson M, Pedersen NL, et al (2010). Association

between CYP2C19 polymorphism and depressive symptoms. Am J Med Genet B Neuropsychiatr

Genet 153B(6): 1160-1166.

Sindrup SH, Arendt-Nielsen L, Brosen K, Bjerring P, Angelo HR, Eriksen B, et al (1992). The

effect of quinidine on the analgesic effect of codeine. Eur J Clin Pharmacol 42(6): 587-591.

Sindrup SH, Brosen K, Bjerring P, Arendt-Nielsen L, Larsen U, Angelo HR, et al (1990).

Codeine increases pain thresholds to copper vapor laser stimuli in extensive but not poor

metabolizers of sparteine. Clin Pharmacol Ther 48(6): 686-693.

Sindrup SH, Poulsen L, Brosen K, Arendt-Nielsen L, Gram LF (1993). Are poor metabolisers of

sparteine/debrisoquine less pain tolerant than extensive metabolisers? Pain 53(3): 335-339.

Siu EC, Wildenauer DB, Tyndale RF (2006). Nicotine self-administration in mice is associated

with rates of nicotine inactivation by CYP2A5. Psychopharmacology (Berl) 184(3-4): 401-408.

Skurtveit S, Furu K, Selmer R, Handal M, Tverdal A (2010). Nicotine dependence predicts

repeated use of prescribed opioids. Prospective population-based cohort study. Ann Epidemiol

20(12): 890-897.

Smith HS (2009). Opioid metabolism. Mayo Clin Proc 84(7): 613-624.

Smith HS (2011). The metabolism of opioid agents and the clinical impact of their active

metabolites. Clin J Pain 27(9): 824-838.

Snider NT, Sikora MJ, Sridar C, Feuerstein TJ, Rae JM, Hollenberg PF (2008). The

endocannabinoid anandamide is a substrate for the human polymorphic cytochrome P450 2D6. J

Pharmacol Exp Ther 327(2): 538-545.

Soldin OP, Chung SH, Mattison DR (2011). Sex differences in drug disposition. J Biomed

Biotechnol 2011: 187103.

Soldin OP, Mattison DR (2009). Sex differences in pharmacokinetics and pharmacodynamics.

Clin Pharmacokinet 48(3): 143-157.

Somogyi AA, Barratt DT, Coller JK (2007). Pharmacogenetics of opioids. Clin Pharmacol Ther

81(3): 429-444.

Spector AA (2009). Arachidonic acid cytochrome P450 epoxygenase pathway. J Lipid Res 50

Suppl: S52-56.

Stamer UM, Musshoff F, Kobilay M, Madea B, Hoeft A, Stuber F (2007). Concentrations of

tramadol and O-desmethyltramadol enantiomers in different CYP2D6 genotypes. Clin

Pharmacol Ther 82(1): 41-47.

Stamer UM, Zhang L, Book M, Lehmann LE, Stuber F, Musshoff F (2013). CYP2D6 genotype

dependent oxycodone metabolism in postoperative patients. PLoS One 8(3): e60239.

Page 199: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

177

Stamidis H, Young GA (1992). Mu-delta opioid interactions. I: The delta peptide, DPDPE,

increases morphine-induced EEG and EEG spectral power. Peptides 13(4): 749-753.

Stamou M, Wu X, Kania-Korwel I, Lehmler HJ, Lein PJ (2014). Cytochrome p450 mRNA

expression in the rodent brain: species-, sex-, and region-dependent differences. Drug Metab

Dispos 42(2): 239-244.

Stark K, Dostalek M, Guengerich FP (2008). Expression and purification of orphan cytochrome

P450 4X1 and oxidation of anandamide. FEBS J 275(14): 3706-3717.

Starr CJ, Sawaki L, Wittenberg GF, Burdette JH, Oshiro Y, Quevedo AS, et al (2009). Roles of

the insular cortex in the modulation of pain: insights from brain lesions. J Neurosci 29(9): 2684-

2694.

Stein C (1995). The control of pain in peripheral tissue by opioids. N Engl J Med 332(25): 1685-

1690.

Stein C (2016). Opioid Receptors. Annu Rev Med 67: 433-451.

Stein C, Machelska H (2011). Modulation of peripheral sensory neurons by the immune system:

implications for pain therapy. Pharmacol Rev 63(4): 860-881.

Sternini C, Spann M, Anton B, Keith DE, Jr., Bunnett NW, von Zastrow M, et al (1996).

Agonist-selective endocytosis of mu opioid receptor by neurons in vivo. Proc Natl Acad Sci U S

A 93(17): 9241-9246.

Stevens JC, Marsh SA, Zaya MJ, Regina KJ, Divakaran K, Le M, et al (2008). Developmental

changes in human liver CYP2D6 expression. Drug Metab Dispos 36(8): 1587-1593.

Stingl JC, Brockmoller J, Viviani R (2013). Genetic variability of drug-metabolizing enzymes:

the dual impact on psychiatric therapy and regulation of brain function. Mol Psychiatry 18(3):

273-287.

Stingl JC, Esslinger C, Tost H, Bilek E, Kirsch P, Ohmle B, et al (2012). Genetic variation in

CYP2D6 impacts neural activation during cognitive tasks in humans. Neuroimage 59(3): 2818-

2823.

Sultatos LG (1994). Mammalian toxicology of organophosphorus pesticides. J Toxicol Environ

Health 43(3): 271-289.

Susce MT, Murray-Carmichael E, de Leon J (2006). Response to hydrocodone, codeine and

oxycodone in a CYP2D6 poor metabolizer. Prog Neuropsychopharmacol Biol Psychiatry 30(7):

1356-1358.

Takahashi K, Bergstrom M, Frandberg P, Vesstrom EL, Watanabe Y, Langstrom B (2006).

Imaging of aromatase distribution in rat and rhesus monkey brains with [11C]vorozole. Nucl

Med Biol 33(5): 599-605.

Page 200: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

178

Tao Q, Stone DJ, Borenstein MR, Codd EE, Coogan TP, Desai-Krieger D, et al (2002).

Differential tramadol and O-desmethyl metabolite levels in brain vs. plasma of mice and rats

administered tramadol hydrochloride orally. J Clin Pharm Ther 27(2): 99-106.

Teh LK, Bertilsson L (2012). Pharmacogenomics of CYP2D6: molecular genetics, interethnic

differences and clinical importance. Drug Metab Pharmacokinet 27(1): 55-67.

Tetel MJ (2009). Nuclear receptor coactivators: essential players for steroid hormone action in

the brain and in behaviour. J Neuroendocrinol 21(4): 229-237.

Tetrault JM, Butner JL (2015). Non-Medical Prescription Opioid Use and Prescription Opioid

Use Disorder: A Review. Yale J Biol Med 88(3): 227-233.

Thompson SJ, Koszdin K, Bernards CM (2000). Opiate-induced analgesia is increased and

prolonged in mice lacking P-glycoprotein. Anesthesiology 92(5): 1392-1399.

Thorn CF, Klein TE, Altman RB (2009). Codeine and morphine pathway. Pharmacogenet

Genomics 19(7): 556-558.

Thorn DA, Zhang Y, Li JX (2017). Tolerance and cross-tolerance to the antinociceptive effects

of oxycodone and the imidazoline I2 receptor agonist phenyzoline in adult male rats.

Psychopharmacology (Berl) 234(12): 1871-1880.

Tindberg N, Ingelman-Sundberg M (1996). Expression, catalytic activity, and inducibility of

cytochrome P450 2E1 (CYP2E1) in the rat central nervous system. J Neurochem 67(5): 2066-

2073.

Ting-A-Kee R, van der Kooy D (2012). The neurobiology of opiate motivation. Cold Spring

Harb Perspect Med 2(10).

Toselli F, de Waziers I, Dutheil M, Vincent M, Wilce PA, Dodd PR, et al (2015). Gene

expression profiling of cytochromes P450, ABC transporters and their principal transcription

factors in the amygdala and prefrontal cortex of alcoholics, smokers and drug-free controls by

qRT-PCR. Xenobiotica 45(12): 1129-1137.

Toselli F, Dodd PR, Gillam EM (2016). Emerging roles for brain drug-metabolizing cytochrome

P450 enzymes in neuropsychiatric conditions and responses to drugs. Drug Metab Rev 48(3):

379-404.

Trang T, Al-Hasani R, Salvemini D, Salter MW, Gutstein H, Cahill CM (2015). Pain and

Poppies: The Good, the Bad, and the Ugly of Opioid Analgesics. J Neurosci 35(41): 13879-

13888.

Treluyer JM, Jacqz-Aigrain E, Alvarez F, Cresteil T (1991). Expression of CYP2D6 in

developing human liver. Eur J Biochem 202(2): 583-588.

Tripathi HL, Martin BR, Aceto MD (1982). Nicotine-induced antinociception in rats and mice:

correlation with nicotine brain levels. J Pharmacol Exp Ther 221(1): 91-96.

Page 201: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

179

Tsutsumi M, Lasker JM, Shimizu M, Rosman AS, Lieber CS (1989). The intralobular

distribution of ethanol-inducible P450IIE1 in rat and human liver. Hepatology 10(4): 437-446.

Tunblad K, Jonsson EN, Hammarlund-Udenaes M (2003). Morphine blood-brain barrier

transport is influenced by probenecid co-administration. Pharm Res 20(4): 618-623.

Tyndale RF, Droll KP, Sellers EM (1997). Genetically deficient CYP2D6 metabolism provides

protection against oral opiate dependence. Pharmacogenetics 7(5): 375-379.

Tyndale RF, Li Y, Li NY, Messina E, Miksys S, Sellers EM (1999). Characterization of

cytochrome P-450 2D1 activity in rat brain: high-affinity kinetics for dextromethorphan. Drug

Metab Dispos 27(8): 924-930.

Unrod M, Kassel JD, Robinson M (2004). Effects of smoking, distraction, and gender on pain

perception. Behav Med 30(3): 133-139.

Upadhya SC, Tirumalai PS, Boyd MR, Mori T, Ravindranath V (2000). Cytochrome P4502E

(CYP2E) in brain: constitutive expression, induction by ethanol and localization by fluorescence

in situ hybridization. Arch Biochem Biophys 373(1): 23-34.

van Rooij E, Kauppinen S (2014). Development of microRNA therapeutics is coming of age.

EMBO Mol Med 6(7): 851-864.

Vance JM, Ali S, Bradley WG, Singer C, Di Monte DA (2010). Gene-environment interactions

in Parkinson's disease and other forms of parkinsonism. Neurotoxicology 31(5): 598-602.

VanderVaart S, Berger H, Sistonen J, Madadi P, Matok I, Gijsen VM, et al (2011). CYP2D6

polymorphisms and codeine analgesia in postpartum pain management: a pilot study. Ther Drug

Monit 33(4): 425-432.

Velez de Mendizabal N, Jones DR, Jahn A, Bies RR, Brown JW (2015). Nicotine and cotinine

exposure from electronic cigarettes: a population approach. Clin Pharmacokinet 54(6): 615-626.

Venhorst J, ter Laak AM, Commandeur JN, Funae Y, Hiroi T, Vermeulen NP (2003). Homology

modeling of rat and human cytochrome P450 2D (CYP2D) isoforms and computational

rationalization of experimental ligand-binding specificities. J Med Chem 46(1): 74-86.

Verrotti A, Loiacono G, Laus M, Coppola G, Chiarelli F, Tiboni GM (2011). Hormonal and

reproductive disturbances in epileptic male patients: emerging issues. Reprod Toxicol 31(4): 519-

527.

Vevelstad M, Pettersen S, Tallaksen C, Brors O (2009). O-demethylation of codeine to morphine

inhibited by low-dose levomepromazine. Eur J Clin Pharmacol 65(8): 795-801.

Vihavainen T, Relander TR, Leiviska R, Airavaara M, Tuominen RK, Ahtee L, et al (2008).

Chronic nicotine modifies the effects of morphine on extracellular striatal dopamine and ventral

tegmental GABA. J Neurochem 107(3): 844-854.

Page 202: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

180

Virk MS, Williams JT (2008). Agonist-specific regulation of mu-opioid receptor desensitization

and recovery from desensitization. Mol Pharmacol 73(4): 1301-1308.

Voirol P, Jonzier-Perey M, Porchet F, Reymond MJ, Janzer RC, Bouras C, et al (2000).

Cytochrome P-450 activities in human and rat brain microsomes. Brain Res 855(2): 235-243.

Volk B, Hettmannsperger U, Papp T, Amelizad Z, Oesch F, Knoth R (1991). Mapping of

phenytoin-inducible cytochrome P450 immunoreactivity in the mouse central nervous system.

Neuroscience 42(1): 215-235.

Volkow ND, McLellan AT (2016). Opioid Abuse in Chronic Pain--Misconceptions and

Mitigation Strategies. N Engl J Med 374(13): 1253-1263.

Volkow ND, McLellan TA, Cotto JH, Karithanom M, Weiss SR (2011). Characteristics of

opioid prescriptions in 2009. JAMA 305(13): 1299-1301.

Wagner B, Wolf GK, Ulmar G (1988). Does Smoking Reduce the Risk of Neuroleptic

Parkinsonoids. Pharmacopsychiatry 21(6): 302-303.

Waldhoer M, Bartlett SE, Whistler JL (2004). Opioid receptors. Annu Rev Biochem 73: 953-990.

Waller D, Schalling D, Levander S, Edman G (1983). Smoking, pain tolerance, and

physiological activation. Psychopharmacology (Berl) 79(2-3): 193-198.

Walther B, Ghersi-Egea JF, Minn A, Siest G (1986). Subcellular distribution of cytochrome P-

450 in the brain. Brain Res 375(2): 338-344.

Wandel C, Kim R, Wood M, Wood A (2002). Interaction of morphine, fentanyl, sufentanil,

alfentanil, and loperamide with the efflux drug transporter P-glycoprotein. Anesthesiology 96(4):

913-920.

Wang A, Savas U, Hsu MH, Stout CD, Johnson EF (2012). Crystal structure of human

cytochrome P450 2D6 with prinomastat bound. J Biol Chem 287(14): 10834-10843.

Wang HB, Zhao B, Zhong YQ, Li KC, Li ZY, Wang Q, et al (2010). Coexpression of delta- and

mu-opioid receptors in nociceptive sensory neurons. Proc Natl Acad Sci U S A 107(29): 13117-

13122.

Wang HY, Friedman E, Olmstead MC, Burns LH (2005a). Ultra-low-dose naloxone suppresses

opioid tolerance, dependence and associated changes in mu opioid receptor-G protein coupling

and Gbetagamma signaling. Neuroscience 135(1): 247-261.

Wang JS, Markowitz JS, Donovan JL, Devane CL (2005b). P-glycoprotein does not actively

transport nicotine and cotinine. Addict Biol 10(2): 127-129.

Wang Q, Han X, Li J, Gao X, Wang Y, Liu M, et al (2015). Regulation of cerebral CYP2D alters

tramadol metabolism in the brain: interactions of tramadol with propranolol and nicotine.

Xenobiotica 45(4): 335-344.

Page 203: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

181

Wang RW, Newton DJ, Scheri TD, Lu AY (1997). Human cytochrome P450 3A4-catalyzed

testosterone 6 beta-hydroxylation and erythromycin N-demethylation. Competition during

catalysis. Drug Metab Dispos 25(4): 502-507.

Wang X, Li J, Dong G, Yue J (2014). The endogenous substrates of brain CYP2D. Eur J

Pharmacol 724: 211-218.

Wang Y, Mitchell J, Moriyama K, Kim KJ, Sharma M, Xie GX, et al (2005c). Age-dependent

morphine tolerance development in the rat. Anesth Analg 100(6): 1733-1739.

Warner M, Gustafsson JA (1994). Effect of ethanol on cytochrome P450 in the rat brain. Proc

Natl Acad Sci U S A 91(3): 1019-1023.

Waxman DJ, Holloway MG (2009). Sex differences in the expression of hepatic drug

metabolizing enzymes. Mol Pharmacol 76(2): 215-228.

Waxman DJ, O'Connor C (2006). Growth hormone regulation of sex-dependent liver gene

expression. Mol Endocrinol 20(11): 2613-2629.

Webb JA, Rostami-Hodjegan A, Abdul-Manap R, Hofmann U, Mikus G, Kamali F (2001).

Contribution of dihydrocodeine and dihydromorphine to analgesia following dihydrocodeine

administration in man: a PK-PD modelling analysis. Br J Clin Pharmacol 52(1): 35-43.

Webster L (2009). Update on abuse-resistant and abuse-deterrent approaches to opioid

formulations. Pain Med 10 Suppl 2: S124-133.

Webster LR, Kopecky EA, Smith MD, Fleming AB (2016). A Randomized, Double-Blind,

Double-Dummy Study to Evaluate the Intranasal Human Abuse Potential and Pharmacokinetics

of a Novel Extended-Release Abuse-Deterrent Formulation of Oxycodone. Pain Med 17(6):

1112-1130.

Weingarten TN, Erie EA, Shi Y, Schroeder DR, Abel MD, Warner DO (2011). Influence of

tobacco use on postoperative opiate analgesia requirements in patients undergoing coronary

artery bypass graft surgery. Signa Vitae 6(2): 72-77.

Wen MM (2016). Getting miRNA Therapeutics into the Target Cells for Neurodegenerative

Diseases: A Mini-Review. Front Mol Neurosci 9: 129.

Whistler JL, Chuang HH, Chu P, Jan LY, von Zastrow M (1999). Functional dissociation of mu

opioid receptor signaling and endocytosis: implications for the biology of opiate tolerance and

addiction. Neuron 23(4): 737-746.

Wightman R, Perrone J, Portelli I, Nelson L (2012). Likeability and abuse liability of commonly

prescribed opioids. J Med Toxicol 8(4): 335-340.

Williams JT, Christie MJ, Manzoni O (2001). Cellular and synaptic adaptations mediating opioid

dependence. Physiol Rev 81(1): 299-343.

Page 204: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

182

Williams JT, Ingram SL, Henderson G, Chavkin C, von Zastrow M, Schulz S, et al (2013).

Regulation of mu-opioid receptors: desensitization, phosphorylation, internalization, and

tolerance. Pharmacol Rev 65(1): 223-254.

Wilson-Poe AR, Moron JA (2017). The dynamic interaction between pain and opioid misuse. Br

J Pharmacol.

Wise RA, Rompre PP (1989). Brain dopamine and reward. Annu Rev Psychol 40: 191-225.

Wisniewski AM, Purdy CH, Blondell RD (2008). The epidemiologic association between opioid

prescribing, non-medical use, and emergency department visits. J Addict Dis 27(1): 1-11.

Wittert G, Hope P, Pyle D (1996). Tissue distribution of opioid receptor gene expression in the

rat. Biochem Biophys Res Commun 218(3): 877-881.

Wojcikowski J, Golembiowska K, Daniel WA (2007). The regulation of liver cytochrome p450

by the brain dopaminergic system. Curr Drug Metab 8(6): 631-638.

Wright GE, Niehaus DJ, Drogemoller BI, Koen L, Gaedigk A, Warnich L (2010). Elucidation of

CYP2D6 genetic diversity in a unique African population: implications for the future application

of pharmacogenetics in the Xhosa population. Ann Hum Genet 74(4): 340-350.

Wu D, Otton SV, Inaba T, Kalow W, Sellers EM (1997). Interactions of amphetamine analogs

with human liver CYP2D6. Biochem Pharmacol 53(11): 1605-1612.

Wuttke H, Rau T, Heide R, Bergmann K, Bohm M, Weil J, et al (2002). Increased frequency of

cytochrome P450 2D6 poor metabolizers among patients with metoprolol-associated adverse

effects. Clin Pharmacol Ther 72(4): 429-437.

Wyss A, Gustafsson JA, Warner M (1995). Cytochromes P450 of the 2D subfamily in rat brain.

Mol Pharmacol 47(6): 1148-1155.

Xia Y, Driscoll JR, Wilbrecht L, Margolis EB, Fields HL, Hjelmstad GO (2011). Nucleus

accumbens medium spiny neurons target non-dopaminergic neurons in the ventral tegmental

area. J Neurosci 31(21): 7811-7816.

Xiao L, Xie X, Zhai Y (2010). Functional crosstalk of CAR-LXR and ROR-LXR in drug

metabolism and lipid metabolism. Adv Drug Deliv Rev 62(13): 1316-1321.

Xie R, Hammarlund-Udenaes M (1998). Blood-brain barrier equilibration of codeine in rats

studied with microdialysis. Pharm Res 15(4): 570-575.

Xu BQ, Aasmundstad TA, Bjorneboe A, Christophersen AS, Morland J (1995). Ethylmorphine

O-deethylation in isolated rat hepatocytes. Involvement of codeine O-demethylation enzyme

systems. Biochem Pharmacol 49(4): 453-460.

Page 205: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

183

Xu Y, Sun Y, Yao L, Shi L, Wu Y, Ouyang T, et al (2008). Association between CYP2D6 *10

genotype and survival of breast cancer patients receiving tamoxifen treatment. Ann Oncol 19(8):

1423-1429.

Yaksh TL, Rudy TA (1978). Narcotic analgestics: CNS sites and mechanisms of action as

revealed by intracerebral injection techniques. Pain 4(4): 299-359.

Yanev SG, Kent UM, Roberts ES, Ballou DP, Hollenberg PF (2000). Mechanistic studies of

cytochrome P4502B1 inactivation by xanthates. Arch Biochem Biophys 378(1): 157-166.

Yang X, Zhang B, Molony C, Chudin E, Hao K, Zhu J, et al (2010). Systematic genetic and

genomic analysis of cytochrome P450 enzyme activities in human liver. Genome Res 20(8):

1020-1036.

Yang Z, Yang Z, Arheart KL, Morris R, Zhang Y, Rodriguez Y, et al (2012). CYP2D6 poor

metabolizer genotype and smoking predict severe postoperative pain in female patients on arrival

to the recovery room. Pain Med 13(4): 604-609.

Yiannakopoulou E (2015). Pharmacogenomics and Opioid Analgesics: Clinical Implications. Int

J Genomics 2015: 368979.

Yu AM, Idle JR, Byrd LG, Krausz KW, Kupfer A, Gonzalez FJ (2003). Regeneration of

serotonin from 5-methoxytryptamine by polymorphic human CYP2D6. Pharmacogenetics 13(3):

173-181.

Yue J, Khokhar J, Miksys S, Tyndale RF (2009). Differential induction of ethanol-metabolizing

CYP2E1 and nicotine-metabolizing CYP2B1/2 in rat liver by chronic nicotine treatment and

voluntary ethanol intake. Eur J Pharmacol 609(1-3): 88-95.

Yue J, Miksys S, Hoffmann E, Tyndale RF (2008). Chronic nicotine treatment induces rat

CYP2D in the brain but not in the liver: an investigation of induction and time course. J

Psychiatry Neurosci 33(1): 54-63.

Zacny JP, Apfelbaum SM, Perkins KA (2013). Modulating roles of smoking status and sex on

oxycodone-induced nausea and drug liking. Exp Clin Psychopharmacol 21(2): 103-111.

Zahari Z, Ismail R (2014). Influence of Cytochrome P450, Family 2, Subfamily D, Polypeptide 6

(CYP2D6) polymorphisms on pain sensitivity and clinical response to weak opioid analgesics.

Drug Metab Pharmacokinet 29(1): 29-43.

Zanger UM, Raimundo S, Eichelbaum M (2004). Cytochrome P450 2D6: overview and update

on pharmacology, genetics, biochemistry. Naunyn Schmiedebergs Arch Pharmacol 369(1): 23-

37.

Zanger UM, Schwab M (2013). Cytochrome P450 enzymes in drug metabolism: regulation of

gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther 138(1):

103-141.

Page 206: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

184

Zarrindast MR, Faraji N, Rostami P, Sahraei H, Ghoshouni H (2003). Cross-tolerance between

morphine- and nicotine-induced conditioned place preference in mice. Pharmacol Biochem

Behav 74(2): 363-369.

Zawertailo LA, Tyndale RF, Busto U, Sellers EM (2010). Effect of metabolic blockade on the

psychoactive effects of dextromethorphan. Hum Psychopharmacol 25(1): 71-79.

Zhang H, Falck JR, Roman RJ, Harder DR, Koehler RC, Yang ZJ (2017). Upregulation of 20-

HETE Synthetic Cytochrome P450 Isoforms by Oxygen-Glucose Deprivation in Cortical

Neurons. Cell Mol Neurobiol.

Zhang J, Ferguson SS, Barak LS, Bodduluri SR, Laporte SA, Law PY, et al (1998). Role for G

protein-coupled receptor kinase in agonist-specific regulation of mu-opioid receptor

responsiveness. Proc Natl Acad Sci U S A 95(12): 7157-7162.

Zhong Y, Dong G, Luo H, Cao J, Wang C, Wu J, et al (2012). Induction of brain CYP2E1 by

chronic ethanol treatment and related oxidative stress in hippocampus, cerebellum, and

brainstem. Toxicology 302(2-3): 275-284.

Zhou K, Khokhar JY, Zhao B, Tyndale RF (2013). First demonstration that brain CYP2D-

mediated opiate metabolic activation alters analgesia in vivo. Biochem Pharmacol 85(12): 1848-

1855.

Zhu W, Cadet P, Baggerman G, Mantione KJ, Stefano GB (2005). Human white blood cells

synthesize morphine: CYP2D6 modulation. J Immunol 175(11): 7357-7362.

Zollner C, Stein C (2007). Opioids. Handb Exp Pharmacol(177): 31-63.

Zubieta JK, Smith YR, Bueller JA, Xu Y, Kilbourn MR, Jewett DM, et al (2001). Regional mu

opioid receptor regulation of sensory and affective dimensions of pain. Science 293(5528): 311-

315.

Page 207: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

185

7. Appendices

Appendix A: CYP-mediated drug metabolism in the brain impacts drug response

Douglas M. McMillan and Rachel F. Tyndale

Reprinted with permission from Elsevier: Pharmacology and Therapeutics

184: 189-200. © 2018 Elsevier. All rights reserved.

Abstract

The functional role of cytochrome P450 (CYP) enzymes in the brain is an exciting and evolving

field of research. CYPs are present and active in the brain, with heterogeneous patterns of

expression, activity, and sensitivity to modulation across cell types, regions, and species. Despite

total brain CYP expression being a fraction of hepatic CYP expression, the expanding literature of

in vitro and in vivo experiments has provided evidence that brain CYPs can impact acute and

chronic drug response, susceptibility to damage by neurotoxins, and are associated with altered

personality, behaviour, and risk of neurological disease. They may also play a role in endogenous

neurotransmitter and neurosteroid homeostasis. This review goes through the characterization of

brain CYPs across species, the patterns of susceptibility of brain CYPs to exogenous induction,

and recent preclinical evidence of the potential role of brain CYPs in vivo (e.g. CYP2D), along

with the development of experiment paradigms that allow modulation of brain CYP activity

without affecting CYP activity in the liver. Understanding brain CYP function, and changes

therein, may provide unique strategies for the development of CNS-acting therapeutics

metabolized locally in the brain, as well as therapeutics to target brain CYPs directly.

Page 208: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

186

1. Introduction to brain CYP enzymes

Cytochrome P450 enzymes (CYPs) are a major source of variability in pharmacokinetics of

exogenous and endogenous compounds. CYPs are regulated in a variety of ways including genetic

variation, and xenobiotic and endogenous (cytokines and hormones) induction and repression

(Zanger and Schwab, 2013). While drug metabolism occurs predominantly in the liver, CYP-

mediated drug metabolism within the brain has emerged as another source of variation in drug

response (Miksys and Tyndale, 2013). Total brain CYP content is low relative to liver

(approximately 0.5–2%) and is therefore unlikely to influence systemic drug and metabolite levels

of CYP substrates (Hedlund et al, 2001). However, brain CYP-mediated metabolism of centrally

acting compounds can impact local brain levels and resulting therapeutic effects, independent of

peripheral metabolism and systemic drug levels. The potential impact of variable brain CYP

activity is substantive, with demonstrated effects on behaviour, neurotoxicity, and drug response

(e.g. analgesics, anesthetics, antidepressants, antipsychotics) (Miksys and Tyndale, 2013; Toselli

et al, 2016; Wang et al, 2014). The focus of this review will be the evolving understanding of the

functional impact of brain CYP families 1–4, including newly developed animal models of

increased and decreased brain CYP activity, as well as new findings in brain CYP-mediated

metabolism of exogenous and endogenous compounds. Outside of this scope there have been

substantial advancements in the research areas of additional CYPs within the brain (e.g. CYP17,

CYP27, and CYP46), however we have focused on CYP1–4 due to their role in the metabolism of

a broad range of clinically relevant central nervous system (CNS)-acting compounds, and the

contrasting regulation of these CYPs in brain relative to that in liver.

2. Experimental in vivo tools for investigating the role of brain CYPs

Early evidence of CYP activity in the brain came in the 1970s (Fishman et al, 1976; Paul et al,

1977; Sasame et al, 1977), which was followed by research into subcellular localization, activity,

and substrate specificity compared to liver CYP enzymes (Miksys and Tyndale, 2004). Substantial

interest exists in brain CYPs due to the potentially unique impact of CYP-mediated metabolism

within the brain (e.g. on the blood-brain barrier (BBB)). This is despite CYP expression being

much lower in the brain than in liver, and also no prior knowledge of the existence of sufficient

levels and locations of cofactors and coenzymes required for CYP-mediated metabolism (Hedlund

et al, 2001). Early detection of CYP activity ex vivo was limited due to low enzyme levels and

Page 209: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

187

rapid degradation; for example, nearly 50% of brain CYP2D activity was lost within a week of

freezing (Tyndale et al, 1999). Nevertheless, reactions catalyzed by CYPs ex vivo have occurred

using brain membranes; for example, the CYP2D oxidation of dextromethorphan to dextrorphan

by human and rat brain microsomes (Tyndale et al, 1999; Voirol et al, 2000). Animal models have

been fundamental in elucidating the impact of brain CYP-mediated metabolism on centrally-acting

drug metabolism and response. The ability to measure drug levels directly in the brain (e.g. by in

vivo microdialysis), specific and selective manipulation of brain CYP activity with pharmacologic

inhibitors and inducers (modeled in Figure 1), molecular inhibition techniques, and transgenic

animal models have provided in vivo evidence of the impact of CYPs expressed in the brain.

2.1. Models of decreased brain CYP activity

One approach to selective brain inhibition is direct intracerebroventricular (ICV) administration

of CYP inhibitors which have no impact on hepatic metabolism confirmed in vivo and ex vivo.

The unique advantage of mechanism-based inhibitors is that they require CYP-mediated activation

prior to their metabolites binding to, and inactivating, the same enzyme; binding of the metabolite

to the enzyme demonstrates that the enzyme within the brain is functional in situ (Miksys and

Tyndale, 2009). The in vivo activity of brain CYPs was first demonstrated in rats through the ICV

injection of the CYP2B mechanism-based inhibitors 8-methoxypsoralen and C8-xanthate (Miksys

and Tyndale, 2009). [3H]-8-methoxypsoralen was injected into rat frontal cortex where it was

enzymatically activated by, and subsequently radiolabelled, CYP2B (Miksys and Tyndale, 2009).

ICV injection of C8-xanthate into one side of the frontal cortex inhibited brain CYP2B on that side

alone, as indicated by a reduction in CYP2B radiolabelling by [3H]-8-methoxypsoralen and

therefore a decrease in CYP2B activity (Miksys and Tyndale, 2009). Further proof of the specific

enzyme involved was provided by the immunoprecipitation of the radiolabelled protein with

antibodies to CYP2B (Miksys and Tyndale, 2009). This demonstrated both the in vivo activity of

rat brain CYP2B, and specificity of binding of the mechanism-based inhibitors.

Similarly, propranolol is a mechanism-based inhibitor of CYP2D; propranolol is first

hydroxylated by CYP2D to a reactive metabolite that binds the active site of the enzyme,

inactivating it (Narimatsu et al, 2001). ICV propranolol inhibits brain, but not liver, CYP2D

activity in rats, as determined by decreased ex vivo oxidation of the CYP2D substrate codeine by

rat brain, but not hepatic, membranes (Zhou et al, 2013).

Page 210: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

188

Figure 1: Model of the expected outcome of altered in vivo brain CYP2D activity on CNS-

acting drug response, where the CYP2D-formed metabolite is more active than the parent

compound. Control brain CYP2D metabolism of parent drug to metabolite results in baseline drug

effect (a). Inhibiting brain CYP2D specifically (e.g. ICV administration of a mechanism-based

inhibitor; INH) reduces metabolite production in brain, and decreases the resulting drug effect (b).

In contrast, brain-specific induction of CYP2D (e.g. chronic nicotine administration) increases

metabolite production in brain, and increases the resulting drug effect (c). ICV inhibitor delivery

subsequent to brain CYP2D induction attenuates both the increase in metabolite production in

brain and the increased drug effect (d).

Gene silencing through RNA interference has emerged as a promising technique for reducing CYP

protein levels in vivo, although its use is limited due to difficulty resulting from inadequate

transport of nucleic acids across CNS membranes (Pardridge, 2007). MicroRNAs (miRNAs)

function as RNA-induced silencing complex guides, mediating translational inhibition and

degradation of targeted genes and proteins (Esteller, 2011). Brain-enriched miRNA has been

implicated in organ-specific regulation of brain CYP2D; exogenous testosterone (given

Page 211: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

189

subcutaneously) decreases rat brain, but not liver, CYP2D levels (Li et al, 2015a). In culture,

testosterone increases brain-specific miR-101 and miR-128-2 in human brain-derived cell lines

(SH-SY5Y and U251); overexpression of miR-101 and miR-128-2 decreased CYP2D mRNA

levels in these cells (Li et al, 2015a).

More general approaches include whole brain-CYP knockout animal models (e.g. brain neuron-

specific deletion of the Cpr gene encoding cytochrome-P450 reductase, required for CYP

function; brain-Cpr-null) (Conroy et al, 2010). Various general CYP inhibitors (e.g. miconazole

and fluconazole) can also be used to assess metabolism by brain CYPs (Hough et al, 2011).

2.2. Models of increased brain CYP activity

CYPs in the brain appear to be especially sensitive to induction following xenobiotic exposure.

Brain CYP expression can be altered in addition to, or independent of, an effect on liver CYP

activity (e.g. phenobarbital increases monkey CYP2B expression in both the liver and brain, but

increases CYP2E expression in brain, but not liver (Lee et al, 2006a; Lee et al, 2006c)) (Table 1).

Chronic nicotine treatment induces rat and monkey brain, but not liver, CYP2B and CYP2D

enzymes, but induces CYP2E enzyme in both the brain and liver (Ferguson et al, 2011; Ferguson

et al, 2012, 2013; Howard et al, 2001; Joshi and Tyndale, 2006a, b; Khokhar et al, 2010; Lee et

al, 2006b; Lee et al, 2006d; Mann et al, 2008; Micu et al, 2003; Miksys et al, 2000b; Miller et al,

2014; Yue et al, 2008).

Page 212: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

190

Table 1. Summary of the effects of nicotine and ethanol on liver and brain CYP expression

across species.

Similarly, ethanol has distinct effects on brain and liver CYP expression. In rats, ethanol increases

CYP2D levels in the brain, CYP2B levels in the liver, and CYP2E levels in both brain and liver

(Howard et al, 2003; Schoedel et al, 2001; Warner and Gustafsson, 1994; Yue et al, 2009). In

monkeys, ethanol induced brain, but not liver, CYP2D while inducing brain and liver CYP2B and

CYP2E levels (Ferguson et al, 2011; Ferguson et al, 2012, 2013; Miller et al, 2014). In human

autopsy tissue, prior smoking (chronic nicotine exposure) and alcohol (ethanol) consumption have

similar effects on brain and liver CYP expression as chronic nicotine and alcohol in rats and

monkeys. Smokers and alcoholics have higher brain levels of CYP2B6 and CYP2E1compared to

non-smoking non-alcoholics, however due to high co-consumption, it is unknown if this is a result

of independent effects (or other variables) of the two compounds (Howard et al, 2003; Miksys et

al, 2003; Zhong et al, 2012). Smokers have higher hepatic CYP2E1, with no change in hepatic

CYP2B6 protein levels or activity, compared to non-smokers, while alcoholics (or drinkers) have

increased hepatic CYP2B6 and CYP2E1 activity (Benowitz et al, 2003; Hesse et al, 2004;

Tsutsumi et al, 1989). Human brain CYP2D6 levels are higher in smokers and alcoholics,

compared to non-smokers and non-alcoholics, with no change in liver CYP2D activity (Mann et

al, 2008; Miksys et al, 2002; Miksys and Tyndale, 2004). The mechanisms and patterns of brain

CYP induction by xenobiotic exposure are not well characterized; details on the region-specificity,

and potential mechanisms, of xenobiotic induction of brain CYPs are discussed later in the paper.

The sensitivity of brain CYPs to induction has been utilized in vivo in proof-of-concept studies of

Page 213: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

191

the role of brain CYP-mediated metabolism of exogenous compounds (discussed in the next

section).

Humanized transgenic animal models of overexpression of brain CYPs are another tool for

examining metabolism by brain CYPs. A human CYP2D6 transgenic (Tg-2D6) mouse model was

generated with human CYP2D6 protein expressed in the liver and brain; compared to wild-type,

Tg-2D6 mice have greater basal serotonin levels (which can be formed in vitro by CYP2D) and

lower harmaline induced tremors (metabolized by CYP2D), suggestive of increased brain CYP2D

activity (Cheng et al, 2013).

3. Evidence of a functional role for brain CYPs

The development of preclinical animal models, whereby brain-specific CYP expression and

activity can be altered, facilitates investigation into the impact of brain CYP-mediated metabolism

in drug efficacy, neurotoxicity, and endogenous compound metabolism. Using animals to model

the potential variation in human brain CYP-mediated metabolism has provided insight into

behavioural consequences of elevated and reduced brain CYP expression and activity in vivo.

3.1. The role of brain CYPs in response to exogenous compounds

The response to centrally acting drugs is variable and not always predicted by plasma drug levels.

CNS-drug response may reflect brain drug levels, modulated, at least in part, by local metabolism

by CYPs. Propofol is a sedative-anesthetic that is inactivated by CYP2B hydroxylation (Court et

al, 2001). Selectively inhibiting (with ICV C8-xanthate) and inducing (with short-term peripheral

injections of nicotine) rat brain CYP2B yielded increased and decreased, respectively, brain

propofol concentrations and propofol responses (measured by sleep duration) (Khokhar and

Tyndale, 2011). The effects of nicotine pretreatment were reversed by ICV C8-xanthate

pretreatment, suggesting that the pretreatments were acting by similar mechanisms of action,

namely via manipulation of brain CYP2B (through induction and inhibition, respectively)

(Khokhar and Tyndale, 2011). Propofol-induced sleep time correlated with brain, but not plasma,

propofol levels (Khokhar and Tyndale, 2011). This pretreatment paradigm allows alteration of

brain CYP2B activity while maintaining normal CYP2B function in the liver, as tested both in

vivo (via systemic drug and metabolite levels) and ex vivo (Khokhar and Tyndale, 2011).

Page 214: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

192

Nicotine, an inducer of rat brain CYP2B when given as a short-term pretreatment, is metabolized

by brain CYP2B in rats (versus CYP2A which is the main hepatic nicotine-metabolizing enzyme

in mice and humans) (Garcia et al, 2015). In similarly designed studies, inhibiting and inducing

rat-brain CYP2B resulted in an increase and decrease, respectively, of brain, but not plasma,

nicotine levels (Garcia et al, 2015; Garcia et al, 2016). Brain nicotine levels were assessed over

time by microdialysis, demonstrating the usefulness of in vivo brain microdialysis in assessing

changing drug and metabolite brain levels in rat behavioural models of altered brain CYP-mediated

metabolism. Inhibiting brain CYP2B (with ICV C8-xanthate, resulting in higher brain nicotine

levels) resulted in a leftward shift of the dose-acquisition curve for nicotine self-administration; a

greater percentage of animals reached acquisition-criteria at lower nicotine doses (Garcia et al,

2015). It also resulted in greater responding when tested on a progressive-ratio, thought to reflect

increased nicotine-reinforcement (Garcia et al, 2015). Human smokers who are

genetically CYP2B6 slow metabolizers report greater nicotine craving scores during abstinence

and are more likely to relapse during nicotine withdrawal, likely due to neuroadaptations in

response to higher brain nicotine levels, suggesting a potential role for brain CYP2B6 in nicotine

metabolism and long-term effects in smokers (Lerman et al, 2002).

Almost all oral opioid drugs (e.g. hydrocodone) have activity at the mu-opioid receptor, and are

enzymatically activated by CYP2D to more potent metabolites (e.g. hydromorphone) (Smith,

2011). CYP2D6 metabolizes the prodrug codeine to morphine, a step required for codeine-

induced analgesia; human CYP2D6 poor metabolizers (no CYP2D6 activity), and individuals

given CYP2D6 inhibitors systemically, produce no/less morphine, and experience no/less

analgesia, from codeine (Chen et al, 1991b; Sindrup et al, 1992). Inhibiting rat brain, but not liver,

CYP2D (with ICV propranolol) decreased brain, but not plasma, morphine levels and decreased

codeine analgesia after peripheral codeine administration (Zhou et al, 2013). Further, inducing rat

brain CYP2D with short term nicotine pretreatments increased brain, but not plasma, morphine

levels and increased codeine analgesia; the change in codeine analgesia after nicotine pretreatment

induction of brain CYP2D was subsequently reversed by inhibiting brain CYP2D with

ICV propranolol mechanism-based inhibitor pretreatment (Figure 2) (McMillan and Tyndale,

2015). Codeine analgesia correlated with brain, but not plasma, morphine levels after codeine

Page 215: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

193

administration (McMillan and Tyndale, 2015). Neither inhibiting nor inducing brain CYP2D had

any effect on drug levels or response following systemic administration of morphine (instead of

codeine); morphine is not a CYP2D substrate (McMillan and Tyndale, 2015). The selective impact

of these manipulations on codeine, but not morphine, metabolism and response, indicated that the

pretreatments acted through altering rat brain CYP2D metabolism, rather than off-site actions of

the pretreatments (McMillan and Tyndale, 2015). This acute effect of brain metabolism was

replicated with the analgesic tramadol, another opioid drug metabolically activated by CYP2D.

Selective rat brain CYP2D inhibition (with ICV propranolol) decreased brain tramadol

metabolism, measured as a decrease in brain levels of the CYP2D metabolite O-

desmethyltramadol (O-DSMT) and a prolongation of tramadol's elimination t1/2 (Wang et al,

2015). Inducing rat brain CYP2D with nicotine increased tramadol analgesia, O-DSMT brain

concentration, and shortened the Tmax of tramadol effect (Wang et al, 2015). Inhibiting and

inducing rat brain CYP2D did not alter the peripheral pharmacokinetics of tramadol in the plasma,

measured as the relative extent of conversion of tramadol to O-DSMT (Wang et al, 2015).

Furthermore, the increase in tramadol analgesia after nicotine pretreatment was attenuated by

inhibiting brain CYP2D (Wang et al, 2015). Beyond acute response, inducing rat brain CYP2D

with nicotine increased the rate of tolerance to codeine's analgesic effects; inducing brain CYP2D

increased initial codeine analgesia, and resulted in a more rapid decrease in analgesia across daily

codeine administration (McMillan and Tyndale, 2017b). The rate of tolerance (decrease in peak

analgesia per day) correlated to initial response, suggesting that changes in initial codeine

analgesia (e.g. via inhibiting or inducing brain CYP2D metabolism to morphine) would predict

changes in tolerance (McMillan and Tyndale, 2017b). Thus, people with increased brain CYP2D

activity (genetically or as a result of CNS CYP2D induction) may experience greater acute opioid-

induced analgesia from codeine or tramadol due to increased central opioid activation, as well as

a more rapid onset of analgesic tolerance, suggesting a potentially increased impetus for dose

escalation. These data also suggest that brain CYP2D may contribute to metabolism and analgesia

of other opioids (e.g. hydrocodone, oxycodone), as well as to other consequences of opioid use

(e.g. side-effects, tolerance, dependence, and withdrawal).

Page 216: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

194

Figure 2: Manipulating in vivo rat brain CYP2D activity alters central metabolism of

codeine to morphine and resulting codeine analgesia. Rat brain CYP2D was inhibited by ICV

pretreatment with propranolol (PL, CYP2D mechanism-based inhibitor given 24 h ahead of

codeine), induced with chronic sub-cutaneous pretreatment with nicotine, or induced and

subsequently inhibited prior to codeine administration (illustrated in a). Codeine is a prodrug that

exhibits little-to-no activity at the μ-opioid receptor and must first be activated by CYP2D to

morphine to produce analgesia. Compared to control animals (grey lines and bars; no inhibitor or

inducer), inhibiting brain CYP2D (grey lines and bars with red border) decreased analgesia at

multiple time points after codeine (b: %MPE, maximum possible effect) presenting as a decrease

in area under the analgesia-time curve surrounding peak analgesia (c: AUC0–30), and decreased

brain morphine levels after codeine administration (d). Inducing brain CYP2D (purple bars and

lines) increased %MPE (b), analgesic AUC0–30 (c), and brain morphine levels (d) after codeine

administration. Inhibiting the induced brain CYP2D (co-pretreatment with both brain CYP2D

inhibitor and inducer; purple bars and lines with red border) attenuated the induced %MPE (b),

analgesic AUC0–30 (c), and brain morphine levels (d) after codeine administration. The full study

is described in McMillan and Tyndale (2015). *p < 0.05 vs control; #p < 0.05, ##p < 0.01 vs

inhibitor plus inducer group. (For interpretation of the references to colour in this figure legend,

the reader is referred to the web version of this article.)

Page 217: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

195

Neuronal CYP epoxygenases may also play a significant, although indirect, role in exogenous and

endogenous produced analgesia. Analgesia in mice, mediated by opioid-receptor activation,

requires CYP-mediated epoxygenation of arachidonic acid (AA) to epoxyeicosatrienoic acids

(EETs), which themselves may activate analgesic circuits via unknown mechanisms (Conroy et

al, 2010; Spector, 2009). The production of EETs from AA is mediated by numerous CYP

isoforms, namely human CYP2C and CYP2J, as well as the orphan enzyme human CYP4X1 (Iliff

et al, 2010; Stark et al, 2008). Decreased mouse brain CYP activity, via transgenic knockout

(brain-Cpr-null) or pharmacologic inhibition (miconazole and fluconazole), attenuated morphine

(opioid) and improgan (non-opioid) analgesia (Conroy et al, 2010; Hough et al, 2011). Morphine-

induced side effects (respiratory depression, locomotor stimulation, and inhibition of intestinal

motility) were not affected by decreased brain CYP epoxygenase activity, suggesting that the

mechanism of these side-effects is dissociated from that of analgesia (Hough et al, 2014a; Hough

et al, 2015). A similar reduction of morphine analgesia was observed in mice treated with selective

CYP epoxygenase inhibitors (MW06-25 and MS-PPOH) (Conroy et al, 2010).

3.2. The protective role of brain CYP-mediated metabolism in neurotoxicity

A large number of central neurotoxins (e.g. organophosphate pesticides, MPTP, TIQ, paraquat and

harmaline) are metabolized by CYP enzymes (Buratti et al, 2003; Miksys and Tyndale, 2006).

Peripheral metabolism phenotypes have been used as predictors of neurotoxic risk; however,

target-site metabolism by brain CYPs may be a novel source of variation in individual

susceptibility to neurotoxic insult. Parkinson's disease (PD) is a progressive neurodegenerative

disorder with an unknown etiology; both genetic variation and environmental exposure to

neurotoxins contribute to PD (Elbaz et al, 2004). Individuals with a CYP2D6 poor metabolizer

phenotype (no CYP2D6 activity) are at a greater risk for PD, which increases with exposure to

neurotoxins (Elbaz et al, 2004). PD patients have approximately 40% lower brain CYP2D6 levels

than age-matched genotype-matched controls, suggesting that low brain CYP2D6 may be a risk

factor for PD (Mann et al, 2012). The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

(MPTP) and its toxic metabolite 1-methyl-4-phenylpyridinium (MPP+) are metabolically

inactivated by CYP2D6; both MPTP and MPP+ are used in experimental animal models of PD

(Mann and Tyndale, 2010). Overexpression of CYP2D6 in cultured rat adrenal medulla (PC12)

cells is protective against neurotoxicity from MPP+, consistent with increased MPTP and

Page 218: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

196

MPP+ neurotoxicity following inhibition of CYP2D6 in cultured human neuroblastoma (SH-

SY5Y) cells (Mann and Tyndale, 2010; Matoh et al, 2003). This suggests that variation in the

activity or expression of CYP2D6 in the brain may affect neurotoxin metabolic inactivation and

degree of neurotoxicity. Tobacco smokers are at lower risk for PD, and nicotine has been found to

be neuroprotective in several neurotoxin-induced animal models of the disease (Alves et al, 2004;

Quik et al, 2009). Chronic nicotine treatment protects against MPTP-induced nigrostriatal damage

in mice, a brain region predominantly affected in PD (Janson et al, 1992). Nicotine induction of

brain, but not liver, CYP2D, among other CYP enzymes, may contribute to the protection against

PD in smokers by increasing metabolic inactivation of neurotoxins (Miksys and Tyndale, 2006).

Chlorpyrifos (CP) is an organophosphate insecticide that is metabolically activated by CYP2B to

the chlorpyrifos-oxon (CPO), a metabolite that inhibits acetylcholinesterase (AChE) causing

chlorpyrifos neurotoxicity (Sultatos, 1994). CPO is quickly deactivated by esterases in the liver

and circulating blood, suggesting that peripherally formed CPO may not reach the brain (Costa et

al, 1990); however, central CPO production by brain CYP metabolic activation may contribute to

CP-induced neurotoxicity. In a rat-model of CP-induced neurotoxicity, inhibiting rat brain CYP2B

(with ICV C8-xanthate) increased brain CP levels, decreased brain CPO levels, reduced CPO-

mediated AChE inhibition in brain, and reduced the severity of CPO-induced hypothermia and

neurotoxicity (assessed with a battery of behavioural tests) (Khokhar and Tyndale, 2012).

Consistent with the hypothesis that CP neurotoxicity results from brain CYP2B activation to CPO,

there were no differences in plasma CP levels, plasma CPO levels, and in peripheral AChE

inhibition following inhibition of brain CYP2B (Khokhar and Tyndale, 2012). CP is a long-lasting

neurotoxin, remaining in the body for prolonged periods of time. Even days after CP

administration, deficits in behaviour were reduced by subsequent ICV C8-xanthate (brain-specific

CYP2B inhibition) as well as by intra-peritoneal C8-xanthate, given peripherally to model human

consumption of an oral therapeutic inhibitor (shown to inhibit brain and hepatic CYP2B) (Khokhar

and Tyndale, 2014). Inhibiting brain metabolism could be a potential therapeutic approach for

reducing the long-lasting levels of CP in the body, and resulting CP toxicity (Khokhar and Tyndale,

2012, 2014).

Page 219: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

197

Individual risk of experiencing side-effects with acute or chronic antipsychotic treatment varies

substantially. Haloperidol is an antipsychotic drug and CYP2D substrate that is associated with

severe extra pyramidal symptoms (EPS), including tardive dyskinesia (Lockwood and Remington,

2015). Catalepsy is used as a model of acute haloperidol induced EPS; selectively inhibiting rat

brain, not hepatic, CYP2D, reduced catalepsy induced by acute haloperidol, while inducing brain

CYP2D increased catalepsy (Miksys et al, 2017). This suggests that brain CYP2D was responsible

for metabolizing haloperidol to a neurotoxic metabolite and that reducing CYP2D activity in the

brain was neuroprotective for this acute haloperidol induced toxicity. While few reports have

studied the effects, acute or chronic, of smoking on EPS, there is some evidence that antipsychotic-

induced side-effects are more prevalent in smokers (Wagner et al, 1988). Increased brain CYP2D

levels in smokers may contribute to a greater risk for acute EPS from antipsychotic therapy; this

might be transferable to the clinic whereby co-administration of a CYP2D6 inhibitor with initial

haloperidol exposure may reduce acute toxicity. In rats, chronic haloperidol exposure increases

vacuous chewing movements, a model of the chronic side effect tardive dyskinesia (Miksys et al,

2017). In contrast to acute haloperidol side effects, brain CYP2D appears to be neuroprotective for

tardive dyskinesia-like side effects caused by chronic haloperidol; inhibiting brain CYP2D during

chronic haloperidol administration increased vacuous chewing movements (Miksys et al, 2017).

This suggests that haloperidol itself is responsible for this chronic side-effect, or a metabolite made

by an alternative enzyme, and that having more brain CYP2D is protective during long term

haloperidol exposure (Figure 3). The difference in underlying neurochemistry between

haloperidol-induced acute (catalepsy, likely mediated at least in part by a CYP2D-mediated

metabolite) and chronic (VCMs, likely mediated by the parent drug haloperidol, or a metabolite

via a non-CYP2D-mediated pathway) neurotoxicity may be one reason behind the opposing role

of brain CYP2D metabolism in acute (a risk factor) and chronic (a protection factor) side effects.

Page 220: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

198

Figure 3: Inhibiting in vivo rat brain CYP2D activity differentially alters extrapyramidal

side-effects from acute and chronic haloperidol. Rat brain CYP2D was inhibited by a single

ICV pretreatment of propranolol (PL, CYP2D mechanism-based inhibitor given 24 h ahead of

haloperidol; illustrated in a), or chronically inhibited with ICV propranolol 3 times per week (with

chronic haloperidol over 16 weeks; illustrated in e). Inhibiting brain CYP2D with ICV propranolol

(PL) reduced catalepsy from a single haloperidol injection, likely mediated by reduced central

metabolite production. Catalepsy time was shorter (b), indicating decreased neurotoxicity, and

correlated positively with brain (c) but not liver (d) CYP2D activity. Brain CYP2D inhibition

during chronic haloperidol exposure increased vacuous chewing movements (VCMs), likely

mediated by increased brain parent drug, haloperidol. VCMs over the last 4 weeks were higher (f),

indicating increased neurotoxicity, and correlated negatively with brain (g) but not liver (h)

CYP2D activity. The full study is described in Miksys et al. (2017). ⁎p < 0.05 vs control.

3.3. Brain CYP-mediated metabolism of endogenous compounds and neurotransmitters

An initial indication that brain CYP-mediated metabolism of endogenous compounds may impact

physiological function was derived from the association between CYP2D6 genotype and

personality (Penas-Lledo and Llerena, 2014). For example, compared to CYP2D6extensive

metabolizers, CYP2D6 poor metabolizers have lower brain perfusion rates (an index of cerebral

activity) in the thalamus and hippocampus, indicative of lower metabolic activity, which associates

with displayed anxious traits (Kirchheiner et al, 2011). CYP2D6genotype was also associated with

Page 221: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

199

cortical brain activation in multiple cognitive tests (working memory and emotional face

matching), suggesting a role for brain CYP2D6 in brain information processing (Stingl et al, 2012).

The relationship between CYPs and endogenous neurotransmitter systems is complex (Brtko and

Dvorak, 2011; Dvorak and Pavek, 2010; Waxman and O'Connor, 2006; Wojcikowski et al, 2007).

Brain CYP2D6 can biosynthesize the monoaminergic neurotransmitters dopamine and serotonin

in vitro; this may represent an additional pathway contributing to total synthesis of these brain

neurotransmitters (Hiroi et al, 1998b; Yu et al, 2003). Formation of dopamine

via tyramine hydroxylation in rat brain microsomes was inhibited by the CYP2D inhibitor quinine

and anti-CYP2D4 antibodies (rat CYP2D4 is a potential homologue of human CYP2D6) (Bromek

et al, 2010). Using cDNA-expressed rat CYPs and human CYP2D6, as well as rat brain

microsomes, CYP2Ds were found to be the most efficient CYPs in forming serotonin from 5-

methyltryptamine; this formation could be inhibited by the CYP2D inhibitors quinine

and fluoxetine (Haduch et al, 2013). Compared to cDNA-expressed wild-type CYP2D6 (in

recombinant E. coli), allelic variants of CYP2D6(CYP2D6*10 and *12, associated with deficient

activity) exhibited decreased 6β-hydroxylation of progesterone and decreased formation of

dopamine from tyramine (Niwa et al, 2004).

Rat brain CYP2D formation of dopamine and serotonin in vivo has been assessed using

microdialysis (Bromek et al, 2011; Haduch et al, 2015). Following enhancement of the potential

role of brain CYP2D via ablation of the classic pathways of dopamine and serotonin production

(e.g. inhibition of dopamine formation from tyrosine with α-methyl-p-tyrosine, and inhibition of

serotonin formation from tryptophan with the serotonergic neurotoxin 5,7-dihydroxytryptamine)

there was some evidence that brain CYP2D could create dopamine and serotonin from

exogenously administered dopamine and serotonin precursors (e.g. tyramine or 5-

methyltryptamine and melatonin) (Figure 4) (Bromek et al, 2011; Haduch et al, 2015; Haduch et

al, 2016). Humanized Tg-2D6 mice (as mentioned previously, with human CYP2D6 protein

expressed in the liver and brain) have higher brain basal serotonin levels relative to wild-type mice

(Cheng et al, 2013). So, while there is currently no direct evidence for a role of brain CYP2D

metabolism in endogenous neurotransmitter and neurosteroid production or removal, studies of

human genetic associations, exogenous precursor metabolism, and humanized mice provide

Page 222: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

200

motivation to continue to examine this potential role for brain CYP-mediated metabolism in

neurochemical homeostasis.

Figure 4: Brain CYP2D may contribute to in vivo central metabolism of

exogenous tyramine to dopamine. When the primary dopamine formation pathway from

tyrosine, and dopamine catabolic pathways, were inhibited (a), the alternate pathway of brain

CYP2D-mediated dopamine formation from tyramine was isolated (b). Exogenous tyramine was

given via microdialysis probe directly into the striatum with or without administration of a striatal

injection of the CYP2D inhibitor quinine (Q; illustrated in b). Compared to controls (grey), brain

CYP2D inhibition had no effect on dopamine levels (grey with red border; c: % of basal level, d:

AUC0–240 min). However, inhibiting brain CYP2D in the presence of exogenous tyramine, which

results in increased brain dopamine levels (green; c, d), attenuated the increase in dopamine

formation (green with red border; c, d). The full experimental results can be found in Bromek et

al. (2011). ⁎p < 0.05, ⁎⁎p < 0.001 vs control; #p < 0.001 vs exogenous tyramine. (For interpretation

of the references to colour in this figure legend, the reader is referred to the web version of this

article.)

In vitro, CYP2C19 metabolizes endogenous (e.g. estradiol, progesterone, and testosterone) and

synthetic (e.g. selective serotonin reuptake inhibitors sertraline and citalopram) psychoactive

compounds (Ingelman-Sundberg et al, 2007) and is expressed in fetal, but not adult, human brain

(Persson et al, 2014). Humans homozygous for the loss of function CYP2C19*2 allele, who do not

express CYP2C19 enzyme, have a lower incidence of depressive symptoms, relative

to CYP2C19 extensive metabolizers (CYP2C19*1/*1) (Sim et al, 2010). The hippocampus is

Page 223: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

201

critical in inhibiting stress responses and reduced hippocampal volume has been found in mood

disorders like major depressive disorder (Femenia et al, 2012). Transgenic male mice

(CYP2C19Tg-Hem) that are hemizygous for human CYP2C19 display greater anxiety and

impairment of hippocampal serotonin turnover (indicated by 5-HIAA (metabolite)/serotonin)

and brain-derived neurotrophic factor homeostasis as well as reduced hippocampal volume and

adult neurogenesis (Jukic et al, 2016; Persson et al, 2014). Although CYP2C19 is expressed in

adult human brain at negligible levels, genetically variable CYP2C19 enzyme function may alter

normal hippocampal activity during neurodevelopment (Toselli et al, 2015). While the mechanism

for the impact of CYP2C19 on hippocampal development is unknown, metabolism of endogenous

compounds involved in overall brain development (e.g. fatty acids, sex hormones) may contribute

(Persson et al, 2014).

CYP3A4 can also metabolize neuroactive steroids (e.g. testosterone and estradiol) thereby having

the potential to influence mood, behaviour, memory, and cognition (Lee et al, 2003; Wang et al,

1997). The incidence of mood disorders and cognitive deficits is increased in epilepsy patients

compared to healthy controls, as well as epilepsy patients treated with anti-epileptics (Beyenburg

et al, 2005; Isojarvi et al, 2005; Verrotti et al, 2011). Human CYP3A4 is higher in post mortem

brains of individuals exposed to anti-epileptic drugs (e.g. carbamazepine and phenytoin); likewise,

mouse CYP3A11 is induced by anti-epileptic drugs in a hippocampal cell line (HN25.1) (Killer et

al, 2009). Administration of anti-epileptic drug phenytoin to mice

induced CYP3A and androgen receptor expression in the hippocampus (Meyer et al, 2006).

Compared to controls, phenytoin treated mice had lower brain testosterone levels, suggesting that

induced brain CYP3A increased testosterone metabolism, thereby yielding a compensatory

increase in androgen receptor expression (Meyer et al, 2006). Similarly, rat brain CYP2E1 was

induced by phenytoin, and following acutely-induced status epilepticus by kainic acid (Boussadia

et al, 2014).

Other CYPs expressed in the brain may also play a role in modulating CNS active

compounds. CYP1A1 and CYP1A2 metabolize numerous endogenous compounds (e.g.

arachidonic acid, other eicosanoids, estrogens, bilirubin, and melatonin) (Rifkind, 2006). In

addition to exogenous opioid analgesia previously discussed, neuronal CYP (e.g. CYP2C, CYP2J

Page 224: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

202

and CYP4X1) epoxygenation of AA to EET plays a role in endogenous opioid-mediated stress-

induced analgesia (Hough et al, 2014b). Compared to control mice, neuronal CYP-deficient (null)

mice exhibited less stress-induced analgesia (as measured by the restraint model of stress-induced

analgesia), similar to the decrease in stress-induced analgesia after pretreatment with the opioid

antagonist naltrexone (Hough et al, 2014b). EETs exhibit anti-inflammatory properties and are

involved in protection against stroke, cerebral ischaemia and pain (Iliff et al, 2010). Exogenous

glutamate increased rat brain CYP2J2 mRNA levels and the sum of EET and

dihydroxyeicosatrienoic acids, indicative of increased CYP2J2 EET production (Liu et al, 2017);

altered CYP-metabolic production of EETs in vivo may be an important drug-target for treating

various disease states. Alternatively, CYP4A and CYP4F can produce hydroxyeicosatetraenoic

acids (HETEs, e.g. 20-HETE) by hydroxylating AA; 20-HETE elicits a wide range of organ-

specific effects on the human vascular system (e.g. maintaining preglomerular vascular tone in the

kidney) (Elshenawy et al, 2017). Brain metabolism of AA to 20-HETE occurs mainly through

CYP2U1, which is abundant in human cerebellum (Chuang et al, 2004). The human brain does

not store glycogen as a reservoir of energy, but instead requires a continuous supply of glucose

and oxygen; regulation of brain vascular tone by 20-HETE (e.g. through brain CYP2U1 production

from AA) may play a role in sustaining normal brain function (Elshenawy et al, 2017; Pratt et al,

2004).

4. Expression and activity of brain CYPs

CYPs are expressed, and active, in the brains of a wide-range of species (Meyer et al, 2007). The

cellular distribution and expression levels of brain CYPs can vary across brain regions and species;

we have outlined some of these differences where important, however more detailed species

comparisons can be found in prior reviews (Toselli et al, 2016). While hepatic CYPs are primarily

expressed in the endoplasmic reticulum, brain CYPs are found on mitochondrial membranes, the

plasma membrane, and other cell compartment membranes (Walther et al, 1986). CYP1B1 is

expressed at the human BBB, where it may act in conjunction with transport proteins by regulating

xenobiotic permeation (Dauchy et al, 2008; Decleves et al, 2011; Dutheil et al, 2009). CYP2B

expression in human brain is highest in the dentate gyrus of the hippocampus and in astrocytes

adjacent to cerebral blood vessels; here it may play a similar role to CYP1B1 in neuroprotection at

the BBB by acting with transporters (Miksys et al, 2003). CYP2B expression in monkey brain is

Page 225: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

203

highest in pyramidal cells of the cortex, Purkinje cells in the cerebellum, and neurons in the

substantia nigra (Lee et al, 2006b), while expression in rats and mice is highest in neurons of the

cerebellum, hippocampus, and thalamus (Volk et al, 1991), suggesting some species differences

in CYP distribution.

CYP2D expression in human and monkey brain is primarily located in pyramidal neurons, with

high expression in the cortex, cerebellum, and hippocampus (Mann et al, 2008; Miksys et al,

2002). In rat and mouse brain, CYP2D is widely expressed in neuronal and glial populations

heterogeneously across brain regions, including regions unprotected by the BBB, such as the

choroid plexus (Miksys et al, 2000a; Miksys et al, 2005; Norris et al, 1996). CYP2E1 shares a

relatively similar expression profile in rat, monkey, and human brain; for example, CYP2E1

protein is expressed in pyramidal neurons of the frontal cortex, cerebellum, and hippocampus

(Garcia-Suastegui et al, 2017; Hansson et al, 1990; Joshi and Tyndale, 2006b; Tindberg and

Ingelman-Sundberg, 1996; Upadhya et al, 2000).

Transcripts for CYP1A2 and the orphan CYP2S1 and CYP4X1 have been detected in rat

hippocampus, cortex, and cerebellum (Stamou et al, 2014). Mouse neurons express CYP4A10 and

CYP4A12a, which metabolize AA and may function to protect the brain from ischemic injury (e.g.

oxygen-glucose deprivation) (Zhang et al, 2017).

5. Regulation of brain CYPs

A unique feature of brain CYPs is their sensitivity to xenobiotic inducers, which may differ from

induction of liver CYPs. Regulation of brain CYPs is region-specific, depending on CYP isoform

and inducer (Miksys and Tyndale, 2004). For example, smokers and alcoholics have higher levels

of brain CYP2B6 in the hippocampus, caudate nucleus, putamen, and cerebellum, with greater

increased expression in individuals classified as both smokers and alcoholics (Miksys et al, 2003).

Repeated nicotine administration (7 days) increases rat brain CYP2B expression and mRNA, with

the largest increases occurring in brain stem, frontal cortex, striatum, and olfactory tubercle

(Miksys et al, 2000a). Nicotine pretreatment did not alter rat liver CYP2B expression or mRNA

levels (Khokhar et al, 2010). The increase in rat brain CYP2B protein and mRNA levels after

nicotine pretreatment was not affected by inhibition of nicotinic-acetylcholine receptor (nAChR)

Page 226: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

204

with chlorisondamine (nAChR irreversible inhibitor), indicating that the mechanism of nicotine

induction of brain CYP2B may be through increased transcription, independent of nAChR

activation (Khokhar et al, 2010). Monkey brain CYP2B6 is induced by nicotine (alone and in

combination with ethanol) with no apparent increase in brain CYP2B6 mRNA levels, suggesting

a species-specific difference in regulation (Ferguson et al, 2013).

While liver CYP2D activity in humans is primarily determined by genetics, brain CYP2D levels

can be altered by both genetics and xenobiotic inducers. Smokers have higher CYP2D6 levels, as

assessed in post-mortem brain, while liver CYP2D levels do not differ between smokers and non-

smokers (Mann et al, 2008; Miksys et al, 2002; Miksys and Tyndale, 2004). Nicotine pretreatment

(7 days) increases rat brain CYP2D expression, with the largest increases in the cerebellum,

hippocampus, and striatum (Yue et al, 2008). Nicotine and ethanol dose-dependently increase

monkey brain CYP2D expression (Mann et al, 2008; Miller et al, 2014). Neither rat nor monkey

brain CYP2D increases were associated with changes in brain CYP2D mRNA levels, and neither

hepatic enzyme activity nor mRNA levels were changed, suggesting a brain-specific effect on

CYP2D by non-transcriptional regulation (Mann et al, 2008; Miller et al, 2014; Yue et al, 2008).

The mechanism of brain CYP2D induction is currently unknown, but may involve

increased protein stability or decreased protein degradation; chronic (14 day) nicotine

administration downregulates ubiquitination enzymes in rat hypothalamus which suggests one

possible mechanism (Kane et al, 2004). Brain CYP2E1 may be differently regulated by nicotine

and ethanol; ethanol treatment induces rat brain CYP2E1 levels and mRNA, suggesting a

transcriptional mechanism of regulation, while induction of CYP2E1 by nicotine is not

accompanied by an increase in mRNA and therefore may involve translational efficiency or protein

stability (Howard et al, 2003; Joshi and Tyndale, 2006a; Zhong et al, 2012). Mitogen-activated

protein kinase signaling may play a role in ethanol induction of brain CYP2E1; inhibitors of the

p38 and ERK1/2 signaling pathways attenuated the induction of rat brain CYP2E1 mRNA and

protein levels by ethanol (Na et al, 2017). Nicotine and ethanol alone, and in combination,

increased monkey brain CYP2E1 levels with no change in mRNA, again suggesting a non-

transcriptional mechanism of regulation (Ferguson et al, 2013). In the case of ethanol, there is a

potential species or dosing paradigm difference between rat and monkey brain CYP2E mechanism

of regulation by xenobiotic inducers (Ferguson et al, 2013). These findings are consistent with the

Page 227: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

205

higher brain CYP2E1 levels observed in alcoholics and smokers compared to non-alcoholics and

non-smokers (Ferguson et al, 2013; Howard et al, 2003; Joshi and Tyndale, 2006b; Warner and

Gustafsson, 1994).

Many known xenobiotic inhibitors and inducers of liver CYP expression have effects on brain

CYP expression as well. Prolonged exposure to antidepressants (imipramine and mirtazapine) and

neuroleptics (thioridazine and risperidone) decreases CYP2D activity and expression in rat brain

and liver (Haduch et al, 2011; Haduch et al, 2004). Similarly, the neuroleptic clozapine and solvent

toluene induce rat CYP2D expression in brain and liver (Hedlund et al, 1996).

Phenobarbital increases the expression and activity of rat brain CYP2B1, CYP2B2, and CYP3A1,

and has been shown to increase monkey brain CYP2B6; phenobarbital also increases liver CYP2B

expression in both species (Lee et al, 2006c; Schilter et al, 2000). Phenobarbital increases monkey

brain CYP2E1 expression in the cerebellum and putamen, with no alteration of liver CYP2E1

protein levels or in vivo chlorzoxazone (CYP2E1 probe drug) disposition (Lee et al, 2006a). In

contrast, rat liver CYP2E1 mRNA is increased with phenobarbital exposure, again suggesting

species-dependent (or dose-dependent) effect of xenobiotics on CYP expression (Caron et al,

2005b).

Brain CYP expression is further regulated by endogenous factors (e.g. developmental or

hormonal). Liver CYP2D6 levels in humans increase rapidly after birth to adult levels and remain

constant with age (Treluyer et al, 1991), however CYP2D6 enzyme levels assessed in frontal

cortex increase with age from fetal stages to 80 years of age, which may occur in other brain

regions as well (Mann et al, 2012). Human sex-based differences in liver CYP expression vary

between CYP isoforms; sex hormones may play a role in regulating brain CYP expression

(Aichhorn et al, 2005; Hildebrandt et al, 2003; Waxman and Holloway, 2009). Exogenous

estrogen and testosterone increases, while progesterone decreases, brain CYP2D expression and

mRNA levels in female rats (Baum and Strobel, 1997; Bergh and Strobel, 1996). In contrast to

this, testosterone decreases CYP2D6 activity in human cell lines (SH-SY5Y and U251) and

orchiectomized growth hormone receptor knockout (GHR-KO) male mice and rats (Li et al,

2015a). In liver, the expression of some CYPs is regulated by xenobiotic receptors (e.g. aryl

hydrocarbon receptor (AhR), peroxisome proliferator-activated receptor (PPAR), and retinoid X

Page 228: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

206

receptor (RXR)) (Honkakoski and Negishi, 2000). In brain, the mRNA of numerous receptor

subtypes has been detected (e.g. PPAR subtypes β and δ, RXRβ and AhR) (Nishimura et al, 2004;

Petersen et al, 2000). Their expression varies among brain regions, is cell-specific, and is

genetically variable (Jeninga et al, 2009; Koyano et al, 2005; Lamba et al, 2005; Lamba et al,

2004a; Lamba et al, 2004b; Moreno et al, 2004; Petersen et al, 2000). Expression of nuclear

receptors in brain can be modulated by other receptors such as the estrogen receptor, and there can

be cross-talk among receptors (Tetel, 2009; Xiao et al, 2010). The role of nuclear receptors in

regulating brain CYP expression, independent of liver CYPs, is currently poorly understood.

6. Conclusions

The functional role of CYP enzymes in the brain is a new, exciting and quickly evolving area of

research. CYPs are present and active in the brain, with heterogeneous patterns of expression,

activity, and sensitivity to modulation across cell types, regions, and species. Despite total brain

CYP expression being a fraction of hepatic, the expanding literature on brain CYP-mediated

metabolism has shown that brain CYPs can impact acute (e.g. codeine analgesia (McMillan and

Tyndale, 2015; Zhou et al, 2013)) and chronic (e.g. nicotine dependence Garcia, et al., 2015))

response to drugs, susceptibility to damage by neurotoxins, and are associated with altered

personality and behaviour, and risk of neurological disease; they may also play a role in

neurotransmitter levels. The multifaceted impact of CYP-mediated brain metabolism is delineated

in the review figures; for any single CYP enzyme active in the brain it may be involved in drug

activation (e.g. Figure 2), protection against neurotoxicity (e.g. Figure 3), and/or neurotransmitter

metabolism (e.g. Figure 4). Understanding the functional role for brain CYP-mediated metabolism

in vivo has been aided by the development of animal models that allow modulation of brain-

specific CYP activity without affecting CYP activity in the liver. This includes (but is not limited

to) direct ICV injection of mechanism-based inhibitors (e.g. C8-xanthate against rat brain CYP2B

(Miksys and Tyndale, 2009) and propranolol against rat brain CYP2D (Zhou et al, 2013)),

peripherally administered inducers of brain CYP activity (e.g. chronic sub-cutaneous nicotine

induction of rat brain CYP2D (Yue et al, 2008)), and brain-specific CYP knockdown models (e.g.

brain-Cpr-null mice (Conroy et al, 2010)). The mechanisms behind the sensitivity of brain CYPs

to xenobiotic inducers are still poorly understood. The use of in vivo brain microdialysis as a

technique for assessing brain metabolism in real-time should prove to be a useful additional tool,

Page 229: Brain CYP2D Metabolism of Opioids Impacts Brain Levels ... · Graduate Department of Pharmacology and Toxicology University of Toronto 2018 Abstract Cytochrome P450 2D (CYP2D) is

207

for example to assess rat brain nicotine levels after brain CYP2B inhibition (Garcia et al, 2015)

and induction (Garcia et al, 2016). Animal imaging studies, as well as human studies which

incorporate human CYP genetic variants, may further elucidate a role for brain CYPs. For

example, neuroimaging studies have begun to explore the link between CYP2D6 polymorphisms

and basic brain function, via tests of working memory and facial expression recognition (Stingl et

al, 2013). Understanding brain CYP function may eventually produce novel strategies for the

development of therapeutics that incorporate not only the consequences of metabolism by brain

CYPs, but also the therapeutic targeting of brain CYP-mediated metabolism.

Acknowledgments

We would like to thank Dr. Sharon Miksys, Marlaina Stocco, and Julie-Anne Tanner for their

assistance in revising this manuscript. We acknowledge our sources of funding: Canada Research

Chair in Pharmacogenomics (R. F. Tyndale); Canadian Institutes of Health Research (CIHR)

grants FDN 154294, TMD 132557, and MOP 136937; the Campbell Family Mental Health

Research Institute of the Centre for Addiction and Mental Health (CAMH);

the CAMH Foundation; the Canada Foundation for Innovation (#20289 and #16014); and

the Ontario Ministry of Research and Innovation. RFT has consulted for Apotex on topics

unrelated to this work. DMM has no conflicts of interest to declare.