128
Mephedrone in the Rat: Mechanisms of Action and Adverse Consequences _______________________________ Craig Motbey (BSc) A thesis submitted in fulfilment of the requirements for the degree of Doctor of Philosophy School of Psychology University of Sydney November, 2012

Mephedrone in the Rat: Mechanisms of Action and Adverse Consequences

  • Upload
    sydney

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Mephedrone in the Rat: Mechanisms of Action

and Adverse Consequences

_______________________________

Craig Motbey (BSc)

A thesis submitted in fulfilment

of the requirements for the degree of

Doctor of Philosophy

School of Psychology

University of Sydney

November, 2012

i

Table of Contents

List of Figures ............................................................................................................................... vi

List of Tables ................................................................................................................................vii

Peer-reviewed Papers.................................................................................................................. viii

Conference Presentations............................................................................................................ viii

General Readership Publications................................................................................................. viii

Selected Mass Media Coverage and Interviews ........................................................................... ix

Abbreviations................................................................................................................................. xi

Acknowledgements...................................................................................................................... xii

Abstract ....................................................................................................................................... xiii

1. Introduction............................................................................................................................ 1-1

1.1. Origin of Mephedrone (MMC)............................................................................................. 1-2

1.2. Spread of MMC.................................................................................................................... 1-3

1.3. Consequences of MMC ....................................................................................................... 1-6

1.4. Structure of MMC.............................................................................................................. 1-11

1.5. Experimental Approach...................................................................................................... 1-12

1.6. References.......................................................................................................................... 1-16

2. Patterns of Brain Activation .................................................................................................. 2-1

Mephedrone (4-methylmethcathinone,‘meow’): acute behavioural effects and distribution of

Fos expression in adolescent rats.............................................................................................. 2-2

2.1. Abstract ................................................................................................................................ 2-4

2.2. Introduction.......................................................................................................................... 2-4

2.3. Experimental Procedures...................................................................................................... 2-5

Subjects ................................................................................................................................. 2-5

Drugs .................................................................................................................................... 2-5

Locomotor Activity............................................................................................................... 2-6

Social Preference................................................................................................................... 2-6

Immunohistochemistry.......................................................................................................... 2-6

Counting of Labelled Cells.................................................................................................... 2-6

ii

Data Analysis......................................................................................................................... 2-6

2.4. Results.................................................................................................................................. 2-7

Locomotor Activity............................................................................................................... 2-7

Social Preference................................................................................................................... 2-7

c-Fos Immunoreactivity....................................................................................................... 2-10

2.5. Discussion .......................................................................................................................... 2-11

Effects of MMC on Locomotor Activity and Social Behaviour......................................... 2-11

Effects of MMC on c-Fos Expression................................................................................. 2-13

Dose and Developmental Considerations............................................................................ 2-14

Conclusions......................................................................................................................... 2-14

2.6. References.......................................................................................................................... 2-15

3. Influence on Neurotransmitter Systems ............................................................................... 3-1

Mephedrone in Adolescent Rats: Residual Memory Impairment and Acute but Not Lasting 5-

HT Depletion............................................................................................................................... 3-2

3.1. Abstract ................................................................................................................................ 3-4

3.2. Introduction.......................................................................................................................... 3-4

3.3 Experimental Procedures....................................................................................................... 3-5

Subjects ................................................................................................................................. 3-5

Drugs and Doses.................................................................................................................... 3-5

Experiment 1: Effects of Acute and Repeated METH and MMC on Locomotor Activity, DA

and 5-HT........................................................................................................................... 3-6

Procedure.......................................................................................................................... 3-6

Apparatus ......................................................................................................................... 3-6

Neurotransmitter and Metabolite Analysis....................................................................... 3-7

Autoradiography .............................................................................................................. 3-7

Experiment 2: Long-term Residual Effects of MMC............................................................ 3-7

Behavioural Tests.............................................................................................................. 3-7

Elevated Plus Maze........................................................................... .......................... 3-7

Social Preference.......................................................................................................... 3-7

Novel Object Recognition............................................................................................ 3-7

iii

Neurotransmitter and Metabolite Analysis....................................................................... 3-7

Data Analysis.................................................................................................. ...................... 3-7

3.4. Results ................................................................................................................................. 3-7

Drug Analysis........................................................................................................................ 3-7

Experiment 1.......................................................................................................................... 3-7

Weight Gain..................................................................................................................... 3-7

Locomotor Activity......................................................................................................... 3-7

Neurotransmitter Levels.................................................................................................. 3-7

Autoradiography Results................................................................................................. 3-8

Experiment 2.......................................................................................................................... 3-9

Long Term Residual Effects of MMC............................................................................. 3-9

Neurotransmitter Levels.................................................................................................. 3-9

3.5. Discussion ............................................................................................................................ 3-9

3.6. References.......................................................................................................................... 3-12

4. Potential for Addiction ......................................................................................................... 4-1

High Levels of Intravenous Mephedrone (4-Methylmethcathinone) Self-Administration In

Rats: Neural Consequences and Comparison with Methamphetamine................................... 4-2

4.1. Abstract ................................................................................................................................ 4-4

4.2. Introduction.......................................................................................................................... 4-4

4.3. Experimental Procedures...................................................................................................... 4-5

Subjects.................................................................................................................................. 4-5

Drugs..................................................................................................................................... 4-5

Surgery .................................................................................................................................. 4-5

Post-Operative Procedures.................................................................................................... 4-6

Apparatus............................................................................................................................... 4-6

Procedure............................................................................................................................... 4-6

Autoradiography.................................................................................................................... 4-7

Neurotransmitter and Metabolite Analysis............................................................................ 4-7

Data Analysis......................................................................................................................... 4-7

4.4. Results ................................................................................................................................. 4-8

iv

Drug Analysis........................................................................................................................ 4-8

Behavioural Data................................................................................................................... 4-8

Initial Training.................................................................................................................. 4-8

Active Pokes................................................................................................................. 4-8

Inactive Pokes............................................................................................................... 4-8

Infusions....................................................................................................................... 4-8

Activity......................................................................................................................... 4-8

Body Weight ................................................................................................................ 4-8

Dose Response FR1.......................................................................................................... 4-9

Active Pokes................................................................................................................. 4-9

Inactive Pokes............................................................................................................. 4-10

Infusions..................................................................................................................... 4-10

Activity....................................................................................................................... 4-11

Body Weight .............................................................................................................. 4-11

Dose Response Under PR Schedule................................................................................ 4-11

Active Pokes............................................................................................................... 4-11

Inactive Pokes............................................................................................................. 4-11

Infusions..................................................................................................................... 4-11

Activity....................................................................................................................... 4-11

Body Weight .............................................................................................................. 4-13

Maximal Dosing Phase................................................................................................... 4-13

Autoradiography............................................................................................................. 4-13

Neurotransmitter and Metabolite Analysis..................................................................... 4-13

4.5. Discussion .......................................................................................................................... 4-13

4.6. References ......................................................................................................................... 4-15

5. General Discussion ................................................................................................................ 5-1

5.1. Understanding the Mechanisms of MMC............................................................................ 5-2

5.2. Responding to MMC............................................................................................................ 5-8

5.3. Dangerous Possibilities: The Example of MPTP............................................................... 5-11

5.4. Conclusions........................................................................................................................ 5-13

v

5.5. References.................................... ..................................................................................... 5-14

6. Appendix: Statements from Co-Authors ............................................................................... 6-1

vi

List of Figures

Chapter 1

Figure 1. Life-threatening Necrotizing Fasciitis Due to ‘Bath Salts’ Injection.......................... 1-8

Figure 2. Structural Similarities Between Amphetamines and Cathinones.............................. 1-13

Chapter 2

Figure 1. Schematic Diagram of Brain Regions Counted.......................................................... 2-8

Figure 2. Locomotor and Stationary Activity During Locomotor Test...................................... 2-9

Figure 3. Representative Locomotor Paths During Locomotor Test.......................................... 2-9

Figure 4. Social Interaction Time and Locomotor Activity During Social Preference Test...... 2-9

Figure 5. Representative Images of c-Fos Immunoreactivity in the Medial Striatum.............. 2-11

Figure 6. Representative Images of c-Fos Immunoreactivity in the Nucleus Accumbens....... 2-12

Chapter 3

Figure 1. Weight Gain Over the Dosing Period.......................................................................... 3-7

Figure 2. Locomotor Activity on First and Last Days of Dosing............................................... 3-8

Figure 3. Residual Effects on Elevated Plus Maze and Social Preference............................... 3-10

Figure 4. Residual Effects on Novel Object Recognition......................................................... 3-10

Chapter 4

Figure 1. Self-administration Behaviour During the Initial Training Period............................ 4-10

Figure 2. Self-administration Behaviour During Fixed Ratio Dose Response......................... 4-11

Figure 3. Self-administration Behaviour During Progressive Ratio Dose Response................ 4-13

Chapter 5

Figure 1. Survivors of the MPTP epidemic.............................................................................. 5-12

vii

List of Tables

Chapter 2

Table 1. Counts of Fos-positive Cells in Regions Significantly Different From Vehicle......... 2-10

Table 2. Counts of Fos-positive Cells in Regions Not Significantly Different From Vehicle.. 2-12

Chapter 3

Table 1. Test Sequence Employed in Experiment 2.................................................................... 3-7

Table 2. Acute Effects on Neurotransmitter and Metabolite Levels........................................... 3-8

Table 3. Autoradiographic Measures........................................................................................... 3-9

Table 4. Residual Effects on Neurotransmitter and Metabolite Levels..................................... 3-11

Chapter 4

Table 1. Mean (SEM) Autoradiography Results........................................................................4-14

Table 2. Mean (SEM) Striatal Neurotransmitter and Metabolite Levels…….......................... 4-14

viii

Peer-reviewed Papers

Motbey CP, Hunt GE, Bowen MT, Artiss S, Mcgregor IS (2011) Mephedrone (4-

methylmethcathinone, ‘meow’): acute behavioural effects and distribution of Fos

expression in adolescent rats. Addiction Biology 17(2): 409-422.

Motbey CP, Karanges E, Li KM, Wilkinson S, Winstock AR, Ramsay J, Hicks C, Kendig MD,

Wyatt N, Callaghan PD, McGregor IS (2012) Mephedrone in adolescent rats: residual

memory impairment and acute but not lasting 5-HT depletion. PLoS ONE 7(9): e45473.

Motbey CP, Clemens KJ, Apetz N, Winstock AR, Ramsey J, Li KM, Wyatt N, Callaghan PD,

Bowen MT, Cornish J, McGregor IS (2013). High levels of intravenous mephedrone (4-

methylmethcathinone) self-administration in rats: neural consequences and comparison

with methamphetamine. Journal of Psychopharmacology. Epub ahead of print June 5,

2013, doi: 10.1177/0269881113490325

Conference Presentations

Motbey CP, Hunt GE, Karanges E, Apetz N, Kendig M, Callaghan PD, Clemens K, Cornish J,

McGregor IS (2011) Behavioural and neural characteristics of acute and chronic

mephedrone (4- methylmethcathinone, meow) treatment in adolescent rats. Poster

presented at the 2011 International Behavioural Neuroscience Society meeting.

General Readership Publications

Motbey CP (2012) Mephedrone: what doesn’t kill you might still mess you up. The

Conversation. Available at http://theconversation.edu.au/mephedrone-what-doesnt-kill-

you-might-still-mess-you-up-6238

Motbey CP (2012) Party’s over: mephedrone causes memory impairment. The Conversation.

Available at http://theconversation.edu.au/partys-over-mephedrone-causes-memory-

impairment-9672

ix

Selected Mass Media Coverage and Interviews

Mephedrone an “ecstasy, meth” combo. Published online at

http://www.nzdoctor.co.nz/news/2011/october-2011/28/mephedrone-an-%27ecstasy,-

meth%27-combo.aspx on 28 October 2011.

Meow meow has rats firing. Published online at http://www.theage.com.au/national/meow-

meow-has-rats-firing-20111027-1mm64.html on 28 October 2011.

Party drug lights up rodent brains. Published online at

http://www.australasianscience.com.au/article/issue-januaryfebruary-2012/party-drug-

lights-rodent-brains.html in January 2012.

Mephedrone may lead to permanent brain damage. Published online at

http://www.doctortipster.com/11287-mephedrone-may-lead-to-permanent-brain-

damage.html on 19 September 2012.

Party drug users risking memory loss. Published online at

http://www.scoop.co.nz/stories/GE1209/S00078/party-drug-users-risking-memory-

loss.htm on 19 September 2012.

Drug study suggests lasting brain damage. Published online at

http://www.nzdoctor.co.nz/news/2012/september-2012/19/drug-study-suggests-lasting-

brain-damage.aspx on 19 September 2012.

Party drug damaging to memory? Published online at

http://articles.timesofindia.indiatimes.com/2012-09-20/health/33976464_1_party-drug-

mephedrone-high-doses on 20 September 2012.

Party drug causes memory impairment. Published online at

http://www.indiatvnews.com/news/world/party-drug-causes-memory-impairment-

8919.html on 20 September 2012.

x

Dangerous synthetic drugs becoming more widespread. Published online at

http://www.reportageonline.com/2012/10/dangerous-synthetic-drugs-becoming-more-

widespread/ on 22 September 2012.

“Killer” party pill becoming popular. Published online at

http://www.odt.co.nz/news/national/226954/killer-party-pill-becoming-popular on 22

September 2012.

New revellers’ designer drug alarms authorities. Published online at

http://www.bayofplentytimes.co.nz/news/new-revellers-designer-drug-alarms-

authorities/1560749/ on 27 September 2012.

Mephedrone and memory loss: an interview with Craig Motbey. Published online at

http://www.news-medical.net/news/20121016/Mephedrone-and-memory-loss-an-

interview-with-Craig-Motbey.aspx on 16 October 2012.

The hamster wheel: health in the media. Available online at

http://www.youtube.com/watch?v=bXr12OzX62Y. First broadcast on ABC TV Australia

on 31 October 2012.

Popular party drug no good for memory. Available online at

http://www.theleader.com.au/story/748365/popular-party-drug-no-good-for-memory/ on

10 November 2012.

Street drug innovation leaves scientists in wake. Published online at

http://newsciencejournalism.com/01/2013/street-drug-innovation-leaves-scientists-in-

wake/ on 7 January 2013.

xi

Abbreviations

5-HT 5-hydroxytryptamine (serotonin)

DA Dopamine

DAT Dopamine Transporter

FR Fixed Ratio

HPLC High-Performance Liquid Chromatography

IHC Immunohistochemistry

IP Intraperitoneal

IV Intravenous

MDMA 3,4-methylenedioxymethamphetamine (Ecstasy)

MDPV Methylenedioxypyrovalerone

METH Methamphetamine

MMC 4-methylmethcathinone (Mephedrone)

MPPP 1-methyl-4-phenyl-4-propionoxypiperidine

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

NE Norepinephrine

NET Norepinephrine Transporter

PR Progressive Ratio

REU Regular Ecstasy Users

SERT 5-HT Transporter

xii

Acknowledgements

Jim, because I said I would.

xiii

Abstract

Mephedrone (4-methylmethcathinone, MMC) is a novel recreational drug that first made a

significant appearance in Europe in the closing years of the first decade of the 21st century. Users

reported MMC as having the stimulant-like properties of methamphetamine (METH) or cocaine,

combined with prosocial, entactogenic effects suggestive of 3,4-

methylenedioxymethamphetamine (MDMA, “Ecstasy”). Anecdotal reports also suggested that

MMC was a drug with a very high addictive potential, prone to inducing uncontrolled bingeing.

The spread of MMC was accompanied by a growing corpus of emergency room case reports

suggesting toxicity in overdose. However, at the beginning of the MMC boom there was almost

no published research available as to the acute or lasting effects, mechanism of action or

potential for toxicity. Although the research program presented in this thesis and the work of the

various competing laboratories around the world represent an effort to correct this deficiency, it

remains the case that our understanding of MMC is still relatively meagre. As such, the fact that

very large numbers of people have been consuming substantial amounts of MMC over a period

of years is of great concern.

The research presented in this thesis consisted of three laboratory studies. In the first study, we

examined the behavioural and neural effects of two different MMC doses (15 and 30 mg/kg,

intraperitoneal (IP)) relative to the well-known stimulant METH (2 mg/kg IP) in male adolescent

Wistar rats. Rats were injected, assessed for locomotor activity for 60 minutes and then tested in

a 10-minute social preference test (measuring time spent in close proximity to a real rat versus a

dummy rat). Their brains were then processed using Fos immunohistochemistry to determine

patterns of brain activation. Results showed that MMC caused profound locomotor hyperactivity

at both dose levels while tending to reduce social preference. Patterns of Fos expression with

MMC resembled a combination of those observed with METH and MDMA, with particularly

strong Fos expression in the cortex, dorsal and ventral striatum, ventral tegmental area (all

typical of both MDMA and METH) and supraoptic nucleus (typical of MDMA). These results

demonstrated for the first time the powerful stimulant effects of MMC in animal models and its

capacity to activate mesolimbic regions. These results also provided some empirical basis to user

reports that MMC subjectively resembles a MDMA/METH hybrid.

xiv

The second study consisted of two experiments. In Experiment 1 male adolescent Wistar rats

received single or repeated (once a day for 10 days) injections of MMC (30 mg/kg) or the

comparator drug METH (2.5 mg/kg). Both MMC and METH caused robust hyperactivity in the

1 h following injection, and this effect did not tend to sensitize with repeated treatment. Striatal

dopamine (DA) levels were increased 1 h following either METH or MMC while striatal and

hippocampal serotonin (5-HT) levels were decreased 1 h following MMC but not METH. MMC

caused greater increases in 5-HT metabolism and greater reductions in DA metabolism in rats

that had been previously exposed to MMC. Autoradiographic analysis showed no signs of

neuroinflammation ([125

I]CLINDE ligand used as a marker for translocator protein (TSPO)

expression) with repeated exposure to either MMC or METH. In Experiment 2, rats received

repeated MMC (7.5, 15 or 30 mg/kg once a day for 10 days) and were examined for residual

behavioral effects following treatment. Repeated high (30 mg/kg) dose MMC produced impaired

novel object recognition 5 weeks after drug treatment. However, no residual changes in 5-HT or

DA tissue levels were observed at 7 weeks post-treatment. This lasting memory dysfunction was

the first demonstration of MMC-induced cognitive impairment in the rat and provided mutual

support to a contemporaneous finding suggesting memory damage in human MMC users.

The third study explored the characteristics of intravenous MMC self-administration in the rat,

with METH again used as a comparator drug. Male Sprague-Dawley rats were trained to nose

poke for intravenous MMC or METH in daily 2 h sessions over a 10 day acquisition period

before dose-response functions were established under fixed- and progressive-ratio (FR and PR,

3 days at each of 4 doses) schedules, followed by a 3 day period of maximal dosing. Brains were

analysed for striatal 5-HT and DA levels while autoradiography assessed changes in the density

of 5-HT and DA transporters (SERT, DAT) as well as induction of the inflammation marker

Translocator Protein (TSPO) in various brain regions. Under a FR1 schedule, peak responding

for MMC was obtained at 0.1 mg/kg/infusion, versus 0.01 mg/kg/infusion for METH. Break

points under a PR schedule peaked at 1 mg/kg/infusion MMC versus 0.3 mg/kg/infusion for

METH. Final intakes of MMC were 31.3 mg/kg/day compared to 4 mg/kg/day for METH.

METH, but not MMC, self-administration caused elevated TSPO receptor density in the nucleus

accumbens and hippocampus. MMC, but not METH, self-administration caused decreased 5-

xv

HIAA levels. MMC supported high levels of self-administration, matching or exceeding those

previously reported with any other drug of abuse. This finding combined with evidence from a

variety of other sources confirms the anecdotal reports of MMC’s potential for addiction.

Overall, the evidence presented in this thesis suggests that MMC may induce lasting cognitive

impairment and carries a high risk of addiction. MMC use involves a significant risk of

morbidity and a relatively minor risk of mortality. While not amongst the most dangerous of

abused drugs, MMC does appear to be substantially more harmful than the drug it partially

displaced (MDMA). As such, the case of MMC consists of not just another example of the

failure of prohibition to prevent the harms associated with illicit drug use, but also provides a

case study for the accelerating dangers of the novel psychoactive market.

1-1

1. Introduction

_______________________________

1-2

1.1 Origin of Mephedrone (MMC)

Throughout much of the modern era, recreational and addicted drug use has been

focussed upon a few traditionally utilised substances along with a limited set of familiar

pharmaceuticals. While the consequences of this use of alcohol, cannabis, opiates,

amphetamines, barbiturates and psychedelics were sometimes tragic, they were at least

predictable. The situation, while regrettable, was relatively stable. This is no longer the case.

Rising to prominence in the early 1980’s, the phenomenon of “designer drugs” appears to

have begun with the illicit production of established medicinal drugs. The effects of many

existing drugs can be greatly enhanced or altered by slightly modifying their molecular

structures, and the tools and techniques developed by modern chemists to pursue these

possibilities eventually spread into the world of illicit drug development. Although the initial

motivation to develop novel psychoactives was probably largely driven by a desire to evade

narrowly targeted drug laws, it did not take long for the recreational drug industry to embrace the

opportunity to diversify and enhance their products (Langston and Palfreman, 1995).

Most prominent amongst the recently emerging drugs of abuse is mephedrone (MMC1).

As of early 2010, Europol reported MMC detection in twenty European member states

(European Monitoring Centre for Drugs and Drug Addiction (EMCDDA), 2010). MMC use is

also well established in Australia (Bruno et al., 2012) and the U.S.A. (US Department of Justice

Drug Enforcement Agency, 2011), and has been detected in Canada (Maheux et al., 2010) and

New Zealand (Winstock and Wilkins, 2011).

First synthesized in 1929 (de Burnaga Sanchez, 1929; EMCDDA, 2010), MMC remained

little more than a chemical footnote until it developed a following amongst recreational drug

users in the late 2000s. MMC is a substituted cathinone, with a degree of divergence from the

basic cathinone molecule similar to that between 3,4-methylenedioxymethamphetamine

1 AKA 4’-methylmethcathinone or β-keto-(4,N-dimethylamphetamine) or 4,N-dimethylcathinone or p-methyl-methcathinone or 2-

aminomethyl-1-tolyl-propan-1-one or (±)-1-(4-methylphenyl)-2-methylaminopropan-1-one or N-methylephedrone or (RS)-2-methylamino-1-(4-

methylphenyl)propan-1-one; molecular formula C11H15NO, molecular weight 177.242.

1-3

(MDMA) and amphetamine. The human use of cathinones for their psychoactive properties

traces back to prehistory. Known as khat (or some close variation thereof) or miraa in eastern

and southern Africa, the leaves and twigs of the Catha edulis shrub have traditionally been

chewed in order to experience the effects of the natural cathinone contained within (Manghi et

al., 2009). While not entirely devoid of negative consequences (Corkery et al., 2010; Manghi et

al., 2009), traditionally managed cathinone consumption nevertheless lies at the milder end of the

spectrum of stimulants of concern (Klein and Metaal, 2010).

In its most common manifestation, MMC is a white or off-white powder. This

hydrochloride salt form of MMC is both stable and water soluble. Most of the MMC so far seen

has been of extremely high purity, and is apparently the product of industrial-scale production in

Asia (Brunt et al., 2010; Dargan et al., 2011; EMCDDA, 2010; Nutt, 2011; Whalen, 2010; Wood

et al., 2010b). In addition to being sold undisguised as itself, MMC is one of a number of

ingredients that appear in drug mixes sold as “plant food” or “bath salts”. While both of these

terms apparently gained popularity as a means of evading regulations applying to products sold

for human consumption, it appears that the term “plant food” may be due to MMC’s brief use as

an experimental insecticide in Israel (Nutt, 2012).

It appears that the most common non-MMC ingredient of “bath salts” is the related

synthetic stimulant methylenedioxypyrovalerone (MDPV). While the European experience of

novel cathinones has been focussed upon MMC with a relatively minor contribution from

MDPV, the North American situation appears to be the reverse (Schneir et al., 2012). Evidence

suggests that MDPV may carry a higher risk of toxicity and lethality than MMC (Baumann et al.,

2012), and that differences in their mechanisms of action may render them particularly

dangerous in combination (Cameron et al., 2012). Like MMC, a subset of MDPV users consume

the drug by injection, and rates of IV MDPV use appear to be rising rapidly (Csák et al., 2013).

1.2 Spread of MMC

MMC developed a substantial presence on the drug markets of the industrialised world at

the close of the first decade of the 21st century. The first official detection of MMC appears to

1-4

have happened in early 2008, in the analysis of a suspect powder contained in capsules seized by

Finnish customs in late 2007 and early 2008 (EMCDDA, 2010). User reports involving MMC

also began appearing on the online “psychonaut” resource Erowid in early 2008 (Erowid, 2011).

During 2009, there was a steep increase in the frequency with which the U.K. Forensic Science

Service identified MMC and other cathinones in drug seizures, and by March 2010, these

outnumbered MDMA and piperazine seizures combined (Dargan and Wood, 2010).

The popularity of MMC grew rapidly, and was accompanied by an increase in MMC-

related hospital presentations during 2009 (James et al., 2011; Wood and Dargan, 2010). By

2010 the annual Mixmag drug survey (measuring a self-selected sample drawn from a U.K.

based nightclubbing-focused population) stated that 41.7% of their respondents reported prior

use of MMC and 33.6% had consumed the drug in the previous month. A 2010 survey of 1006

college and high school students in Scotland found that 205 (20.3%) reported consuming MMC

at least once previously (Dargan et al., 2010). As well as this intentional consumption of MMC,

there is also evidence that MMC was being substituted for MDMA: 11.5% of the “Ecstasy”

surveyed in the Netherlands in 2009 contained MMC (Brunt et al., 2010).

It has been repeatedly suggested in both the academic and popular press that China is the

root source of MMC (Brunt et al., 2010; Dargan et al., 2011; Nutt, 2011; Whalen, 2010; Wood et

al., 2010b), although the truth of this statement remains to be conclusively demonstrated

(Vardakou et al., 2010). Chinese origin was not uncommon in MMC detections to date (Brunt et

al., 2010; EMCDDA, 2010; Wood et al., 2010b), and several drug interdiction agencies have

identified China as a key source of MMC (EMCDDA, 2010; Vardakou et al., 2010; US

Department of Justice Drug Enforcement Agency, 2011). However, as the location of synthetic

drug production is geographically flexible, the potential rise of indigenous MMC-producing

industries in MMC-using countries cannot be ruled out.

It appears that a substantial driver of the use of MMC was user dissatisfaction with the

purity and availability of MDMA and cocaine, possibly created by an increase in the

effectiveness of prohibition enforcement (Advisory Council on the Misuse of Drugs, 2010;

Measham et al., 2010; Winstock and Wilkins, 2011). A variety of sources confirm that the

worldwide MDMA supply had seen a sharp decline in quality and availability (Brunt et al., 2010;

Dick and Torrance, 2010; Winstock and Wilkins, 2011). Between 2006 and 2010 MDMA

seizures by the U.K. customs service dropped from roughly 1,100 per quarter to less than one

1-5

hundred. Concurrent with this reduction in MDMA confiscation, customs seizures of MMC and

other cathinones rose from a base of essentially zero to a steeply-climbing 600 in early 2010 (just

prior to the introduction of legal controls on the possession of MMC in the U.K. during April of

that year) (Dargan and Wood, 2010).

Although the focus of MMC consumption appears to be concentrated in Europe and the

U.K., MMC has begun developing a presence worldwide. MMC use is reported to be substantial

and increasing in the U.S.A. (US Department of Justice Drug Enforcement Agency, 2011),

although it appears that their novel psychoactive market has a greater focus upon MDPV than is

the case in Europe (Baumann et al., 2012). MMC use is well established in Ireland (Van Hout

and Brennan, 2010; Van Hout and Brennan, 2011b; Van Hout and Brennan, 2011c; Van Hout

and Bingham, 2012) and across central Europe (EMCDDA, 2010; Pharris et al., 2011).

Geographic isolation appears to be little defence, with MMC detected in “ecstasy” tablets seized

in New Zealand (Winstock and Wilkins, 2011). A nationwide survey of Australian regular

ecstasy users (REU) in 2010 found that 17% reported MMC use in the previous six months. The

same survey found that rates of prior six months reported MMC use amongst REU in Tasmania

climbed from zero in 2007 to near 50% in 2010 (Bruno et al., 2012).

Media and political reaction to the rise of MMC has tended to follow the classical

progression from prurient curiosity through hyperbolic panic to knee-jerk response (Davey et al.,

2010; Forsyth, 2012; Lancaster et al., 2011; Measham et al., 2011; Sare, 2011; Schifano et al.,

2011; Silverman, 2010; Van Hout and Bingham, 2012). The U.K. tabloid campaign was notable

for the frequent referencing of fatalities that, on more thorough examination, proved to be

unrelated to MMC (Davey et al., 2010; Lancaster et al., 2011; Sare, 2011; Winterton, 2010). The

subsequent process leading to prohibition was marked by the resignation of several members of

the U.K. Advisory Council on the Misuse of Drugs (still recovering from the recent and

controversial dismissal of David Nutt (Nutt, 2009b; Nutt, 2010)) in protest at political

interference in the activity of the council (Sare, 2011). However, the role of the internet in both

the distribution of and media response to MMC differentiates this case from previous drug panics

(Forsyth, 2012; Measham et al., 2010; Schifano et al., 2011). Two factors in particular stand out.

Firstly, it is commonly observed that initial media reports on an emerging drug of abuse

effectively serve as advertisements for the use of that drug (Forsyth, 2012; Measham et al.,

2011). In the case of MMC, this phenomenon acquired a newly literal meaning, as context-

1-6

sensitive advertising servers accompanied the internet editions of early newspaper reports on

MMC with advertisements for online MMC suppliers (Forsyth, 2012; Schifano et al., 2011).

Secondly, the interactive nature of internet media appears to have somewhat altered the

dynamics of the classical drug scare campaign. The rise of drug-focussed (“psychonaut”) internet

discussion groups and the spread of commenting options on mainstream media reporting provide

a powerful platform for competing narratives to reach large-scale audiences. While it remains too

early to accurately predict what effect this may have on the MMC phenomenon, it appears likely

that this new voice may substantially modify the effect of the traditional media presentation

(Forsyth, 2012).

A significant feature of the early years of the MMC pandemic was the fact that MMC

remained legal in the U.K. and several other European countries until mid to late 2010.

Compared to the subterranean word-of-mouth spread of previous novel drugs, the ready

availability and active marketing of online and main street “research chemical” and “headshop”

suppliers gave MMC a significant boost in establishing its presence. MMC is now controlled

across the European Union (Dargan et al., 2011). In countries with broadly drawn analogue laws

such as Australia, MMC was already arguably illegal when use first surfaced (Duff, 2010).

While MMC regulation in the U.S.A. is complicated by the diversity of state legislation, by mid

2011 a majority of the states had enacted laws controlling MMC and other cathinones (US

Department of Justice Drug Enforcement Agency, 2011) and in July 2012 President Obama

signed the Synthetic Drug Abuse Prevention Act forbidding the sale of MMC, MDPV and a

number of other drugs nationwide (Gershman and Fass, 2012).

1.3 Consequences of MMC use

The reported health consequences of MMC use range from the surprisingly mild to the

alarmingly gruesome. The typical picture of “MMC gone wrong” involves a distressed MMC

consumer presenting at a hospital emergency room with a broad spectrum of sympathomimetic

symptoms (Wood et al., 2010a; Wood et al., 2010b; Wood et al., 2010c; Wood and Dargan,

2010). Clinical reports suggest users occasionally suffer from agitation, combative behaviour,

psychosis, tachycardia, hyperthermia, hypertension and seizures (Bajaj et al., 2010; Baumann et

1-7

al., 2012; McGaw and Kankam, 2010; Wood et al., 2010a; Wood et al., 2010b; Wood et al.,

2010c). However, in the majority of cases, patients were discharged without apparent lasting

harm after a few hours of sedation and symptomatic treatment.

While the possibility of fatal MMC-linked hyperthermia has been raised in one animal

study (McGregor et al., 2011), the lack of any reported hyperthermia or lethality in similar

studies (Miller et al., 2012; Wright et al., 2012) and the scarcity of reports of fatal hyperthermia

in human users (Wood et al., 2010a) argues against over-interpreting this result. Although a

number of deaths have been linked to MMC (Dickson et al., 2010; Gustavsson and Escher, 2009;

Lusthof et al., 2011; Maskell et al., 2011; Schifano et al., 2012; Torrance and Cooper, 2011;

Wood et al., 2010a), there are few in which a causal role for MMC alone has been unequivocal

(Winstock et al., 2011). While it appears certain that MMC can be fatal to the sufficiently

unlucky or foolish, the overall impact of MMC on fatality seems remarkably slight given the

scale of use.

The most serious immediate harms of MMC are focused upon the minority of users who

consume the drug intravenously. MMC is mildly acidic in solution, which may at least partially

explain the common reports of burning sensations on insufflation (Dargan et al., 2010; Erowid,

2011) or injection (Van Hout and Bingham, 2012). However, this acidity does not appear

sufficient to explain the dramatically negative outcomes (Figure 1) seen in studies of MMC

injectors (Dorairaj et al., 2011; Russo et al., 2012; Van Hout and Bingham, 2012). One study

reported “vein blockages, […] skin erosion, localised infections, blisters, spots, cold sores,

abscesses, scabs, lumps, gangrenous tissue, blood clots and large holes at overused injecting

sites”. To quote one of their respondents: “it clots up your veins twice as fast as anything

else…that’s one thing I did learn fast from it, say you used one vein on it and got two hits out of

it, you wouldn’t get the vein again, you’d have to move onto a different vein to get another hit

out of it, that’s why everyone started using their groin” (Van Hout and Bingham, 2012: p 191). A

case series from a single hospital reported multiple cases of MMC induced injuries serious

enough to require major surgical debridement and added that “extravasation of these substances

either intentionally or accidentally appear to result in local cutaneous reactions similar to that

seen with cytotoxic drug use” (Dorairaj et al., 2011: p e39).

1-8

Figure 1. Life-threatening Necrotizing Fasciitis Due to ‘Bath Salts’ Injection (adapted from Russo et al.

(2012)). 1: Patient at time of operative incision. 2: Photograph during forearm debridement.

The exact cause of the dramatic impact of MMC on injecting users remains to be

identified. However, the behavioural characteristics of MMC consumption appear likely to

significantly contribute to the negative outcomes for injectors. Early anecdotal reports of MMC

frequently emphasized its binge-inducing properties (Erowid, 2011). A survey of Dutch MMC

users found that 61 of 70 reported “craving for the drug” as an acute effect (Brunt et al., 2010).

An observational test of MMC intoxication in humans found that ratings on a 1-7 Likert scale of

“want MMC” rose from below 4 to approximately 6 after drug consumption (and were just

above 2 on a drug-free comparison test a week later) (Freeman et al., 2012). A report on injecting

MMC users states that “all participants described intense cravings to re-inject compulsively

within each injecting episode” (Van Hout and Bingham, 2012).

From the point of view of safer injecting practices, the hyper-confidence and risk

insensitivity associated with stimulant intoxication already provides a high-risk setting for

injecting drug use. However, unlike other drugs associated with compulsive re-dosing (e.g.:

cocaine), MMC is not a pure stimulant. The additional sensorimotor distortion induced by the

entactogenic and psychedelic aspects of MMC, when combined with the prospect of multiple

intoxicated injection events, provides an unparalleled opportunity for infection and other

injection-related harms. This is particularly the case when the drug, like MMC, is observed to be

prone to causing direct damage to injection sites (Dorairaj et al., 2011; Van Hout and Bingham,

2012). The first signs of MMC’s impact on the injecting community may already be apparent,

1-9

with a rise in central European rates of I.V. MMC use accompanied by a rise in H.I.V. detections

(Pharris et al., 2011).

Although it is apparently not the typical method, an alternative MMC synthesis route can

potentially result in manganese-contaminated product (Dargan et al., 2011). Manganese toxicity

is known to be related to movement disorders suggestive of Parkinson’s, and such toxicity has

been observed in users of other cathinones (Sikk et al., 2007; Sikk et al., 2010; Stepens et al.,

2008). While the possibility of manganese toxicity in MMC consumers has been considered and

dismissed by other authors (Dargan et al., 2011), this dismissal was based largely upon the lack

of reports of Parkinsonian symptoms amongst MMC users. Given the recent reports of

Parkinsonian symptoms in I.V. MMC users (Brennan and Van Hout, 2012; Van Hout and

Bingham, 2012), the possibility of manganese contamination in the MMC supply may need to be

reconsidered.

Behavioural evidence from human studies suggests that MMC use may induce cognitive

impairment, as human MMC users displayed substantially impaired performance on a prose

recall task even while not acutely intoxicated (Freeman et al., 2012). In this study, regular recent

MMC users (reporting more than two instances of use per month) were compared to a control

group of participants who had consumed MMC on a single occasion more than six months ago.

Psychometric testing was conducted on two consecutive weekends, with participants in the

MMC group consuming participant-provided drug during the first testing session. MMC group

participants displayed elevated depression and schizotypy scores relative to controls when drug-

free. Measures of impulsivity correlated positively with reported length of MMC-using sessions.

User reports subjective effects showed substantial increases in “self-confidence”, “buzzing”,

“dizziness” and “impaired concentration and memory” after MMC consumption.

In addition to direct neurological or other physical damage, there is also the social and

psychological toll of addiction and uncontrolled use to consider. MMC is subjectively perceived

by human users to be intensely prone to inducing compulsive use (Brunt et al., 2010; Erowid,

2011; Freeman et al., 2012; Van Hout and Bingham, 2012). A highly common feature of early

reports of MMC use was that drug consumption continued until the supply was exhausted, even

when this supply was intended to last for an extended period (Erowid, 2011). Clinical and

anecdotal reports of sustained addiction to MMC are not uncommon (Bajaj et al., 2010; Dargan

et al., 2010; Erowid, 2010; Simu et al., 2010; Van Hout and Bingham, 2012). MMC has been

1-10

shown to induce a conditioned place preference in rats, mice and flatworms (Lisek et al., 2012;

Ramoz et al., 2012) and promotes self-administration behaviour in rats (Hadlock et al., 2011,

Aarde et al., 2013; Motbey et al., 2013).

The behavioural consequences of MMC use extend beyond addiction alone. MMC use

has been associated with both violent and accidental death (Prosser and Nelson, 2011) as well as

a wide assortment of high-risk sexual behaviours (Van Hout and Brennan, 2011a). Significantly

enhanced confidence and euphoria, when combined with dizziness, disturbed concentration,

general confusion and a distorted time sense (Freeman et al., 2012), are features that can be

expected to seriously impair the risk-assessment abilities of MMC users (Borlik, 2012; Van Hout

and Bingham, 2012; Van Hout and Brennan, 2011a). Also of concern is the apparent connection

between MMC and suicide, with self-inflicted deaths making up 29% of a recent sample of

MMC-linked fatalities (Schifano et al., 2012) and elevated levels of depression reported amongst

MMC users (Freeman, 2012).

The speculation as to links between MMC and the "Miami Cannibal" case, in which a

man brutally attacked a randomly chosen victim, were based upon nothing more than the

unsupported comment of one police officer (Helberg, 2012). Media coverage of the event far

outpaced toxicological analysis, which ultimately confirmed that MMC had no role in the attack

(Hiaasen et al., 2012). However, events such as these are not altogether without precedent in the

MMC literature. One of the early MMC fatality reports was the case of a man in the Netherlands

who, under the influence of MMC and a variety of other drugs, died after smashing a number of

windows while in a “rage of fury” (Lusthof et al., 2011). As with the Miami case, the subject of

this report was also found to have stripped himself naked. Indeed, public nakedness appears to be

a fairly common theme in media reports of “bath salts” intoxication (Borlik, 2012; Shepard,

2012; Warren, 2012). MMC-induced hyperthermia likely plays a role in these events, along with

the entactogenic, disinhibiting and aphrodisiac effects of the drug. More alarmingly, it appears

that face-biting is also a recurring feature (Campbell, 2012; McCorquodale, 2012). However, the

unfounded speculation around the Miami case has likely introduced a strong selection bias into

the media coverage of MMC and the causative role of “bath salts” (MMC or not) in some of

these events is rather questionable.

1-11

1.4 Structure of MMC

The cathinone family of molecules consists of a nearly exact replication of the

amphetamines (Figure 2), with the sole exception of the addition of a ketone group at the β

position of the carbon chain (which is why the cathinones are sometimes referred to as “beta

keto-” or “BK-amphetamines”). In all of the examples studied so far (Dal Cason et al., 1997;

Patel, 2009), the psychoactive properties of the cathinones have closely resembled those of their

amphetamine “twins”, albeit with slightly reduced potency by mass (most likely due to an

impaired ability to cross the blood-brain barrier, as a result of the increased hydrophilicity

induced by the extra ketone group (Gibbons and Zloh, 2010)).

Cathinone functions similarly to amphetamine (Griffiths et al., 2010), affecting the

dopamine (DA), serotonin (5-HT) and norepinephrine (NE) systems through the stimulation of

neurotransmitter release and the modulation of reuptake. Cathinone also slows neurotransmitter

metabolism by inhibiting the action of monoamine oxidase (MAO), with a preferential action

biased more towards MAO-B than MAO-A (Osorio-Olivares et al., 2004). As MAO-B is the

primary enzyme responsible for DA breakdown, inhibition results in an increase in the synaptic

availability of DA. Chronic administration of cathinone to rats creates DA depletions similar to

those seen after chronic amphetamine or cocaine (Ellison, 2002). The cathinones have also been

shown to affect NA systems through the influence of both release (Cleary et al., 2003) and

reuptake (Cozzi et al., 1999).

The existence of well-characterized amphetamine analogues allows one to predict the

likely behavioural and neural effects of many of the cathinones. However, while its production is

not entirely unknown (Davis et al., 2012), the lack of research on the effects of 4’-

methylmethamphetamine (the amphetamine equivalent of MMC) leaves a broad scope of

investigation for researchers of MMC. While the picture has now become somewhat clearer,

there is still considerable work to be done.

1-12

1.5 Experimental Approach

Much of the information now available regarding the neural mechanisms and impacts of

MMC was provided over the last few years by a variety of research groups utilising animal

models of MMC consumption. At the initiation of the research underlying this thesis, no

previous findings had been published exploring the effects of MMC in animal models. Virtually

nothing was known about the mechanisms or dangers of this drug. For the sake of coherence and

clarity, all of the information derived from research with animal models is discussed in Chapter

5, where it is integrated with the findings of the research presented in the body of this thesis. The

literature reviews in the previously published research papers (Chapters 2-4) demonstrate the

development of MMC-related knowledge over the past few years.

The use of animal models provides a significant methodology for the study of illicit

drugs. Given the invasive nature of many neuroscientific research methods and the high risk of

neurotoxic effects with these drugs, both ethical and practical considerations limit the ability of

researchers to investigate these topics in human subjects. Animal models have long been utilised

by many researchers to explore the behavioural impact (e.g.: Clemens et al., 2006; Cornish et al.,

2008; Cornish et al., 2012; Frohmader et al., 2010; Hadlock et al., 2011; Krasnova et al., 2010;

Lisek et al., 2012; Ramoz et al., 2012) as well as the mechanisms of action (e.g.: Kehr et al.,

2012; Martinez-Clemente et al., 2011; Patel, 2009) and potential toxicity (e.g.: McGregor et al.,

2011; Miller et al., 2012; Wright et al., 2012) of illicit drug use. Although the differences

between human and non-human physiology create the need for caution when interpreting the

results of such investigations, these studies provide an essential starting point for further human-

based research.

The experimental program forming the core of this thesis consisted of three rat-based

studies, which were intended to address three basic questions. Firstly, how does MMC influence

the brain? Secondly, does MMC use damage the brain? Thirdly, is MMC addictive, and if so,

how much so?

1-13

Figure 2. Structural similarities between amphetamines and cathinones (adapted from Baumann et al.

(2012)).

The first of these studies (Chapter 2) used c-Fos immunohistochemistry to display the

pattern of brain activation induced by MMC intoxication. As Fos protein is transiently expressed

by neurons in response to strong activation, visualising the distribution of this protein allows the

equivalent of a cellular-resolution timelapse brainscan. Researchers from the University of

Sydney Psychopharmacology group have previously reported the brain-wide distribution of Fos

expression with a diverse range of party drugs including MDMA (Stephenson et al. 1999;

Hargreaves et al. 2007), cannabis (Arnold et al. 2001), methamphetamine (Carson et al. 2010)

and g-hydroxybutyric acid (GHB) (Van Nieuwenhuijzen, McGregor & Hunt 2009a).

A notable benefit of using c-Fos immunohistochemistry is that the protein requires

approximately half an hour to express. Because of this feature, any neural activity in the last half

hour of life will not be recorded by c-Fos expression. As well as removing the influence of the

perfusion process, this also allows for the possibility of excluding portions of the experimental

program from the c-Fos signal. This was done in the experiment presented in Chapter 2; although

the behavioral data from a social preference test is presented alongside the Fos results, the study

1-14

was arranged so that the experience of this test would be excluded from the c-Fos data. This

allowed for the presentation of the clearest possible picture of the basic neural effects of MMC

dosage.

The second study (Chapter 3) aimed to further explore the acute mechanisms of MMC as

well as investigating the potential for neurotoxicity. Autoradiographic tracking of the

inflammation marker TSPO (Translocator Protein) was used to investigate the possibility of an

acute inflammatory response, while High Performance Liquid Chromatography (HPLC) allowed

measurement of the influence of MMC upon DA and 5-HT systems. Acute inflammatory

responses (Pascual et al., 2007) and lasting alterations in neurotransmitter metabolism (Wagner

et al., 1980) are both known consequences of some psychoactive drug use. This analysis was

conducted on brain tissue collected shortly after dosing as well as tissue harvested from animals

given an extended (47 day) washout period. This allowed the examination of both the acute

impact of the drug as well as the possiblity of lasting neuroadaptation or neurotoxicity.

A variety of behavioural measures were also employed during the washout period,

investigating the potential lasting influence of MMC treatment upon anxiety, social behaviour

and memory. Evidence from human studies (Freeman, 2012) had raised concern regarding the

potentially harmful influence of MMC use on memory and other cognitive functions. A broad

range of pilot testing of behavioural measures was undertaken, only some of which is reported in

Chapter 3. These tests were intended to function as the preliminary screen for neurotoxicity and

other health concerns. This screen was performed in conjunction with related work by other

researchers from the University of Sydney Psychopharmacology lab and used a wide variety of

testing paradigms well established in behavioural neuroscience research.

The third study (Chapter 4) focussed upon the addictive potential of MMC use via an

extended self-administration paradigm which compared the properties of MMC to the well-

characterised stimulant METH. A substantial number of previous studies have examined the

impact of self-administration of stimulants on the brain using a variety of indices (Krasnova et

al., 2010; Letchworth et al., 2001; Norwood et al., 2003; Sim-Selley et al., 2000; Turchan et al.,

1998; Wilson and Kish, 1996; Wise et al., 1995). It has long been known that variations in

reinforcement schedule play a key role in such studies (Ferster and Skinner, 1957). Due to this

fact, both fixed and progressive ratio schedules were utilized here in the testing of MMC. Fixed

1-15

ratio schedules allow the measurement of willingness to consume: do the rats like it? In contrast,

progressive ratio schedules measure commitment to gaining reward: how much do the rats want

it? Although subtle, the exploration of differences such as these helps form a thorough

understanding of the addictive potential of a drug.

In this study, dose-response curves were generated for METH and MMC under both

fixed- and progressive-ratio reward schedules. Autoradiographic techniques were also employed

to investigate potential changes in levels of DA and 5-HT transporters as well as the

inflammation marker TSPO. HPLC analysis of striatal tissue was conducted once again in order

to explore the possible differences between the relatively brief experimenter-administered drug

treatment employed in Chapter 3 and the more sustained subject-administered drug treatment of

Chapter 4.

The conclusion of this thesis aims to place these findings into the broader context of

emerging psychoactives and explores some potential responses to this rapidly changing situation.

It will be argued that the example of MMC provides a case study for the flaws of conventional

approaches to drug control in the context of the modern psychoactive drug market.

1-16

1.6 References

Aarde SM, Angrish D, Barlow DJ, et al. (2013) Mephedrone (4‐methylmethcathinone) supports

intravenous self‐administration in Sprague‐Dawley and Wistar rats. Addiction Biology.

Epub ahead of print 30 Jan 2013, doi: 10.1111/adb.12038

Advisory Council on the Misuse of Drugs. (2010) Consideration of the cathinones. London:

Home Office.

Angoa-Pérez M, Kane MJ, Francescutti DM, et al. (2012) Mephedrone, an abused psychoactive

component of ‘bath salts’ and methamphetamine congener, does not cause neurotoxicity

to dopamine nerve endings of the striatum. Journal of Neurochemistry 120: 1097-1107.

Arnold JC, Topple AN, Mallet PE, Hunt GE, McGregor IS (2001) The distribution of

cannabinoid-induced Fos expression in rat brain: differences between the Lewis and

Wistar strain. Brain Research 921:240–255.

Australia21. (2012) The prohibition of illicit drugs is killing and criminalising our children and

we are all letting it happen. University of Sydney: Australia21.

Bajaj N, Mullen D and Wylie S. (2010) Dependence and psychosis with 4-methylmethcathinone

(mephedrone) use. BMJ Case Reports. Epub ahead of print 3 November 2010,

doi:10.1136/bcr.02.2010.2780

Ballard PA, Tetrud JW and Langston JW. (1985) Permanent human parkinsonism due to

1‐methy 1–4‐phenyl‐1, 2, 3, 6‐tetrahydropyridine (MPTP). Neurology 35: 949-949.

Baumann MH, Ayestas MA, Partilla JS, et al. (2011) The designer methcathinone analogs,

mephedrone and methylone, are substrates for monoamine transporters in brain tissue.

Neuropsychopharmacology 37(5): 1192-1203.

Baumann MH, Partilla JS and Lehner KR. (2012) Psychoactive “bath salts”: Not so soothing.

European Journal of Pharmacology. Epub ahead of print 22 November 2012, doi:

10.1016/j.ejphar.2012.11.020

Borlik J. (2012) Police arrest naked car surfer on 'bath salts'. Fox 8 WGHP, 16 June 2012.

Brennan R and Van Hout MC. (2012) Miaow miaow: a review of the new psychoactive drug

mephedrone. Drugs and Alcohol Today 12: 241-253.

1-17

Bruno R, Matthews AJ, Dunn M, et al. (2012) Emerging psychoactive substance use among

regular ecstasy users in Australia. Drug and Alcohol Dependence 124: 19-25.

Brunt TM, Poortman A, Niesink RJ, et al. (2010) Instability of the ecstasy market and a new kid

on the block: mephedrone. Journal of Psychopharmacology 25(11): 1543-1547.

Cameron KN, Kolanos R, Solis E, et al. (2012) Bath salts components mephedrone and

methylenedioxypyrovalerone (MDPV) act synergistically at the human dopamine

transporter. British Journal of Pharmacology 168: 1750-1757.

Campbell A. (2012) Carl Jacquneaux bit a chunk of face off victim todd credeur in louisiana,

cops say. Huffington Post, 6 June 2012.

Carson DS, Hunt GE, Guastella AJ, Barber L, Cornish JL, Arnold JC, Boucher AA, McGregor

IS (2010) Systemically administered oxytocin decreases methamphetamine activation of

the subthalamic nucleus and accumbens core and stimulates oxytocinergic neurons in the

hypothalamus. Addiction Biology 15:448–463.

Cleary L, Docherty JR. (2003) Actions of amphetamine derivatives and cathinone at the

noradrenaline transporter. European Journal of Pharmacology 476(1-2): 31-34.

Clemens KJ, Cornish JL, Hunt GE, et al. (2006) Intravenous methamphetamine self-

administration in rats: effects of intravenous or intraperitoneal MDMA co-administration.

Pharmacology Biochemistry and Behavior 85: 454-463.

Corkery JM, Schifano F, Oyefeso A, et al. (2010) 'Bundle of fun'or'bunch of problems'? Case

series of khat-related deaths in the UK. Drugs: Education, Prevention and Policy: 1-18.

Cornish JL, Clemens KJ, Thompson MR, et al. (2008) High ambient temperature increases

intravenous methamphetamine self-administration on fixed and progressive ratio

schedules in rats. Journal of Psychopharmacology 22: 100-110.

Cornish J, Hunt G, Robins L, et al. (2012) Regional c-Fos and FosB/ΔFosB expression

associated with chronic methamphetamine self-administration and methamphetamine-

seeking behavior in rats. Neuroscience 206: 100-114.

Csák R, Demetrovics Z and Rácz J. (2013) Transition to injecting 3, 4-methylene-dioxy-

pyrovalerone (MDPV) among needle exchange program participants in Hungary. Journal

of Psychopharmacology. Epub ahead of print March 4 2013, doi:

10.1177/0269881113480987

1-18

Cozzi NV, Sievert MK, Shulgin A, Jacob P, Ruoho AE (1999) Inhibition of plasma membrane

monoamine transporters by β–ketoamphetamines. European Journal of Pharmacology

381(1): 63-69.

Dal Cason TA, Young R and Glennon RA. (1997) Cathinone: An investigation of several N-

alkyl and methylenedioxy-substituted analogs. Pharmacology Biochemistry and Behavior

58: 1109-1116.

Dargan PI, Albert S and Wood DM. (2010) Mephedrone use and associated adverse effects in

school and college/university students before the UK legislation change. Quarterly

Journal of Medicine 103: 875-879.

Dargan PI, Sedefov R, Gallegos A, et al. (2011) The pharmacology and toxicology of the

synthetic cathinone mephedrone (4‐methylmethcathinone). Drug Testing and Analysis

3(7-8): 454-463.

Dargan PI and Wood DM. (2010) Annex 1 to the risk assessment report: technical report on

mephedrone. Available at:

http://www.ofdt.fr/BDD/publications/docs/rarOEDTmephAnn1.pdf (accessed 15

September 2012).

Davey Z, Corazza O, Schifano F, et al. (2010) Mass-information: mephedrone, myths, and the

new generation of legal highs. Drugs and Alcohol Today 10: 24-28.

Davis S, Blakey K and Rands-Trevor K. (2012) GC–MS and GC–IRD analysis of 2-, 3-and 4-

methylmethamphetamine and 2-, 3-and 4-methylamphetamine. Forensic Science

International 220(1-3): 67-73.

de Burnaga Sanchez JS. (1929) Sur un homologue de l'ephedrine. Bulletin de la Société

Chimique de France 45: 284.

Dick D and Torrance C. (2010) MixMag drugs survey. Mix Mag 225: 44-53.

Dickson AJ, Vorce SP, Levine B, et al. (2010) Case report: multiple-drug toxicity caused by the

coadministration of 4-methylmethcathinone (mephedrone) and heroin. Journal of

Analytical Toxicology 34: 162-168.

Dorairaj J, Healy C, McMenamin M, et al. (2011) The untold truth about “bath salt” highs: A

case series demonstrating local tissue injury. Journal of Plastic, Reconstructive &

Aesthetic Surgery 65(2): e37-41.

Duff E. (2010) Miaow: ecstasy-like drug hits streets. Sydney Morning Herald, 7 March 2010.

1-19

Ellison G. (2002) Neural degeneration following chronic stimulant abuse reveals a weak link in

brain, fasciculus retroflexus, implying the loss of forebrain control circuitry. European

Neuropsychopharmacology 12(4): 287-297.

Erowid. (2010) Serious addiction potential. Available at:

http://www.erowid.org/experiences/exp.php?ID=85851 (accessed 17 August 2012).

Erowid. (2011) Erowid experience vaults: 4-methylmethcathinone (also mephedrone, 4-MMC, 4-

MMC, meow, m-cat, 2-methylamino-1-p-tolylpropan-1-one) main index. Available at:

http://www.erowid.org/experiences/subs/exp_4Methylmethcathinone.shtml (accessed 17

August 2012).

European Monitoring Centre for Drugs and Drug Addiction (EMCDDA). (2010) Europol–

EMCDDA joint report on a new psychoactive substance: 4-methylmethcathinone

(mephedrone). Report, EMCDDA, Lisbon, March 2010.

European Monitoring Centre for Drugs and Drug Addiction (EMCDDA). (2011) 2010 annual

report on the implementation of council decision 2005/387/JHA. Report, EMCDDA,

Lisbon, May 2011.

Skinner BF and Ferster CB. (1957) Schedules of reinforcement, New York: Appleton-Century-

Crofts.

Forsyth AJM. (2012) Virtually a drug scare: mephedrone and the impact of the internet on drug

news transmission. International Journal of Drug Policy 23(3): 198-209.

Freeman TP, Morgan CJA, Vaughn-Jones J, et al. (2012) Cognitive and subjective effects of

mephedrone and factors influencing use of a ‘new legal high’. Addiction 107: 792-800.

Frohmader KS, Pitchers KK, Balfour ME, et al. (2010) Mixing pleasures: review of the effects of

drugs on sex behavior in humans and animal models. Hormones and Behavior 58: 149-

162.

Gershman JA and Fass AD. (2012) Synthetic cathinones (‘bath salts’): legal and health care

challenges. Pharmacy and Therapeutics 37: 571.

Gibbons S and Zloh M. (2010) An analysis of the 'legal high' mephedrone. Bioorganic &

Medicinal Chemistry Letters 20(14): 4135-4139.

Griffiths P, Lopez D, Sedefov R, et al. (2010) Khat use and monitoring drug use in Europe: the

current situation and issues for the future. Journal of Ethnopharmacology 132: 578-583.

1-20

Gustavsson D and Escher C. (2009) [Mephedrone--internet drug which seems to have come and

stay. Fatal cases in Sweden have drawn attention to previously unknown substance].

Lakartidningen 106: 2769-2771.

Hadlock GC, Webb KM, McFadden LM, et al. (2011) 4-methylmethcathinone (mephedrone):

neuropharmacological effects of a designer stimulant of abuse. Journal of Pharmacology

and Experimental Therapeutics 339: 530-536.

Hargreaves GA, Hunt GE, Cornish JL, McGregor IS (2007) High ambient temperature increases

3,4-methylenedioxymethamphetamine (MDMA, ‘ecstasy’)-induced Fos expression in a

region-specific manner. Neuroscience 145:764–774.

Helberg T. (2012) Causeway cannibal identified; fears grow over drug possibly involved. CBS

Miami, 28 May 2012.

Hiaasen S, Green N and Ovalle D. (2012) No bath salts detected: Causeway attacker Rudy

Eugene had only pot in his system, medical examiner reports. Miami Herald, 27 June

2012.

Hughes B and Winstock AR. (2012) Controlling new drugs under marketing regulations.

Addiction. Epub ahead of print 31 January 2012. doi: 10.1111/j.1360-0443.2011.03620.x.

Izco M, Orio L, O’Shea E, et al. (2007) Binge ethanol administration enhances the MDMA-

induced long-term 5-HT neurotoxicity in rat brain. Psychopharmacology 189: 459-470.

Jackson-Lewis V, Blesa J and Przedborski S. (2012) Animal models of Parkinson's disease.

Parkinsonism & Related Disorders 18: S183-S185.

James D, Adams RD, Spears R, et al. (2011) Clinical characteristics of mephedrone toxicity

reported to the UK national poisons information service. Emergency Medicine Journal

28(8): 686-689.

Kehr J, Ichinose F, Yoshitake S, et al. (2011) Mephedrone, compared to MDMA (ecstasy) and

amphetamine, rapidly increases both dopamine and serotonin levels in nucleus

accumbens of awake rats. British Journal of Pharmacology 164: 1949-1958.

Kelly JP. (2011) Cathinone derivatives: a review of their chemistry, pharmacology and

toxicology. Drug Testing and Analysis 3: 439-453.

Klein A and Metaal P. (2010) A good chew or good riddance--how to move forward in the

regulation of khat consumption. Journal of Ethnopharmacology 132: 584-589.

1-21

Krasnova IN, Justinova Z, Ladenheim B, et al. (2010) Methamphetamine self-administration is

associated with persistent biochemical alterations in striatal and cortical dopaminergic

terminals in the rat. PLoS ONE 5: e8790.

Lancaster K, Hughes C, Spicer B, et al. (2011) Curiosity killed the M-Cat: an examination of

illicit drugs and media. Sydney: Sydney Institute of Criminology.

Langston JW and Palfreman J. (1995) The case of the frozen addicts, New York: Pantheon

Books.

Letchworth SR, Nader MA, Smith HR, et al. (2001) Progression of changes in dopamine

transporter binding site density as a result of cocaine self-administration in rhesus

monkeys. Journal of Neuroscience 21: 2799–2807.

Lisek R, Xu W, Yuvasheva E, et al. (2012) Mephedrone (‘bath salt’) elicits conditioned place

preference and dopamine-sensitive motor activation. Drug and Alcohol Dependence.

Epub ahead of print 29 May 2012. doi 10.1016/j.drugalcdep.2012.04.021.

Lusthof KJ, Oosting R, Maes A, et al. (2011) A case of extreme agitation and death after the use

of mephedrone in The Netherlands. Forensic Science International 206(1-3), e93-95.

Maheux CR, Copeland CR and Pollard MM. (2010) Characterization of three methcathinone

analogs: 4-methylmethcathinone, methylone, and bk-MBDB. Microgram Journal 7(2):

42-49.

Malberg JE and Seiden LS (1998) Small changes in ambient temperature cause large changes in

3, 4-methylenedioxymethamphetamine (MDMA)-induced serotonin neurotoxicity and

core body temperature in the rat. Journal of Neuroscience 18: 5086-5094.

Manghi RA, Broers B, Khan R, et al. (2009) Khat use: lifestyle or addiction? Journal of

Psychoactive Drugs 41(1): 1-10.

Martinez-Clemente J, Escubedo E, Pubill D, et al. (2011) Interaction of mephedrone with

dopamine and serotonin targets in rats. European Neuropsychopharmacology 22: 231-

236.

Maskell PD, De Paoli G, Seneviratne C, et al. (2011) mephedrone (4-methylmethcathinone)-

related deaths. Journal of Analytical Toxicology 35: 188-191.

McCorquodale A. (2012) Brandon DeLeon, high on cloud 9 bath salts and four loko, tries to bite

police: report (VIDEO). Huffington Post, 5 June 2012.

1-22

McGaw C and Kankam O. (2010) The co-ingestion of alcohol and mephedrone-an emerging

cause of acute medical admissions in young adults and a potential cause of

tachyarrhythmias. The West London Medical Journal 2: 9-13.

McGregor IS, Apetz N, Cornish JL, et al. (2011) Mephedrone ('meow'): recent studies using

animal models. Drug and Alcohol Review 30: 64.

Measham F, Moore K and Newcombe R. (2010) Tweaking, bombing, dabbing and stockpiling:

the emergence of mephedrone and the perversity of prohibition. Drugs and Alcohol

Today 10: 14-21.

Measham F, Moore K and Østergaard J. (2011) Mephedrone,“Bubble” and unidentified white

powders: the contested identities of synthetic “legal highs”. Drugs and Alcohol Today 11:

137-146.

Meng H, Cao J, Kang J, et al. (2011) Mephedrone, a new designer drug of abuse, produces acute

hemodynamic effects in the rat. Toxicology Letters 208: 62-68.

Miller M, Creehan K, Angrish D, et al. (2012) Changes in ambient temperature differentially

alter the thermoregulatory, cardiac and locomotor stimulant effects of 4-

methylmethcathinone (mephedrone). Drug and Alcohol Dependence. Epub ahead of print

23 July 2012, doi: 10.1016/j.drugalcdep.2012.07.003.

Motbey CP, Clemens KJ, Apetz N, Winstock AR, Ramsey J, Li KM, Wyatt N, Callaghan PD,

Bowen MT, Cornish J, McGregor IS (2013). High levels of intravenous mephedrone (4-

methylmethcathinone) self-administration in rats: neural consequences and comparison

with methamphetamine. Journal of Psychopharmacology. Epub ahead of print June 5,

2013, doi: 10.1177/0269881113490325.

Nicholson PJ, Quinn MJ and Dodd JD. (2010) Headshop heartache: acute mephedrone 'meow'

myocarditis. Heart 96: 2051-2052.

Norwood CS, Cornish JL, Mallet PE, et al. (2003) Pre-exposure to the cannabinoid receptor

agonist CP 55,940 enhances morphine behavioral sensitization and alters morphine self-

administration in Lewis rats. European Journal of Pharmacology 465: 105–114.

Nutt D. (2009a) Estimating drug harms: a risky business? The Eve Saville Lecture, The Centre

for Crime and Justice Studies (CCJS), King’s College, London.

Nutt D. (2009b) Government vs science over drug and alcohol policy. The Lancet 374: 1731-

1733.

1-23

Nutt D. (2010) Nutt damage? Author's reply. The Lancet 375: 724.

Nutt D. (2011) Perverse effects of the precautionary principle: how banning mephedrone has

unexpected implications for pharmaceutical discovery. Therapeutic Advances in

Psychopharmacology 1: 35-36.

Nutt D. (2012) Drugs - Without the Hot Air. Cambridge: UIT Cambridge.

Nutt D, King LA, Saulsbury W, et al. (2007) Development of a rational scale to assess the harm

of drugs of potential misuse. The Lancet 369: 1047-1053.

Nutt DJ, King LA and Phillips LD. (2010) Drug harms in the UK: a multicriteria decision

analysis. The Lancet 376: 1558-1565.

Osorio-Olivares M, Rezende MC, Sepulveda-Boza S, et al. (2004) MAO inhibition by

arylisopropylamines: the effect of oxygen substituents at the beta-position. Bioorganic

and Medicinal Chemistry 12: 4055-4066.

Pascual M, Blanco AM, Cauli O, et al. (2007) Intermittent ethanol exposure induces

inflammatory brain damage and causes long-term behavioural alterations in adolescent

rats. European Journal of Neuroscience 25: 541.

Patel N. (2009) Mechanism of action of cathinone: The active ingredient of Khat (Catha Edulis).

East African Medical Journal 77(6): 329-332.

Pharris A, Wiessing L, Sfetcu O, et al. (2011) Human immunodeficiency virus in injecting drug

users in Europe following a reported increase of cases in Greece and Romania, 2011.

Eurosurveillance 16: 48.

Prosser JM and Nelson LS. (2011) The toxicology of bath salts: a review of synthetic cathinones.

Journal of Medical Toxicology 8(1): 33-42.

Ramoz L, Lodi S, Bhatt P, et al. (2012) Mephedrone ('bath salt') pharmacology: insights from

invertebrates. Neuroscience 208: 79-84.

Reuter P. (2011) Options for regulating new psychoactive drugs: a review of recent experiences.

Report, U.K. Drug Policy Commission, London, 15 May 2011.

Russo R, Marks N, Morris K, et al. (2012) Life-threatening necrotizing fasciitis due to ‘bath

salts’ injection. Orthopedics 35: e124-e127.

Sare J. (2011) How the media helped ban mephedrone. BMJ. Epub ahead of print, 24 Feb 2011.

doi: 10.1136/bmj.d1138.

1-24

Schifano F, Albanese A, Fergus S, et al. (2011) Mephedrone (4-methylmethcathinone;‘meow

meow’): chemical, pharmacological and clinical issues. Psychopharmacology 214(3):

593-602.

Schifano F, Corkery J and Ghodse AH. (2012) suspected and confirmed fatalities associated with

mephedrone (4-methylmethcathinone," meow meow") in the United Kingdom. Journal of

Clinical Psychopharmacology 32(5): 710-714.

Schneir A, Ly BT, Smollin C, et al. (2012) Comprehensive Drug Analysis of "Bath Salts"

Purchased in the United States. North American Congress of Clinical Toxicology.

Available at:

http://www.clintox.org/NACCT/2012/NACCT_Platform_Abstracts_2012.pdf (accessed

23 Aug 2013).

Shepard W. (2012) Completely naked man ran at 3-year-old girl at playground: mother. NBC

Miami, 8 June 2012.

Shortall SE, Green AR, Swift KM, et al. (2012) Differential effects of cathinone compounds and

MDMA on body temperature in the rat, and pharmacological characterisation of

mephedrone-induced hypothermia. British Journal of Pharmacology. Epub ahead of

print, 8 Oct 2012. doi: 10.1111/j.1476-5381.2012.02236.x

Shulgin A. (1991) PIHKAL: a chemical love story. Berkely, California: Transform Press.

Sikk K, Taba P, Haldre S, et al. (2010) Clinical, neuroimaging and neurophysiological features

in addicts with manganese‐ephedrone exposure. Acta Neurologica Scandinavica 121:

237-243.

Sikk K, Taba P, Haldre S, et al. (2007) Irreversible motor impairment in young addicts–

ephedrone, manganism or both? Acta Neurologica Scandinavica 115: 385-389.

Silverman J. (2010) Addicted to getting drugs wrong. British Journalism Review 21: 31.

Sim-Selley LJ, Selley DE, Vogt LJ, et al. (2000) Chronic heroin self-administration desensitizes

μ opioid receptor-activated G-proteins in specific regions of rat brain. Journal of

Neuroscience 20: 4555–4562.

Simu MI, Hudita C, Rosca MC, et al. (2010) The mephedrone addiction phenomenon (abstract).

International Journal of Neuropsychopharmacology 13: 56.

Stepens A, Logina I, Liguts V, et al. (2008) A Parkinsonian syndrome in methcathinone users

and the role of manganese. New England Journal of Medicine 358: 1009-1017.

1-25

Stephenson CP, Hunt GE, Topple AN, McGregor IS (1999) The distribution of 3,4-

methylenedioxymethamphetamine ‘Ecstasy’-induced c-fos expression in rat brain.

Neuroscience 92:1011–1023.

Torrance H and Cooper G. (2011) The detection of mephedrone (4-methylmethcathinone) in 4

fatalities in Scotland. Forensic Science International 202: e62-63.

Turchan J, Przewlocka B, Lason W, et al. (1998) Effects of repeated psychostimulant

administration on the prodynorphin system activity and kappa opioid receptor density in

the rat brain. Neuroscience 85: 1051–1059.

United Nations Office on Drugs and Crime. (2009) World drug report, 2009. United Nations:

Vienna.

US Department of Justice Drug Enforcement Agency. (2011) Background, data and analysis of

synthetic cathinones: mephedrone (4-MMC), methylone (MDMC), and 3,4-

methylenedioxypyrovalerone (MDPV). Washington DC.: Office of Diversion Control,

Drug and Chemical Evaluation Section.

Upreti VV, Eddington ND, Moon KH, et al. (2009) Drug interaction between ethanol and 3, 4-

methylenedioxymethamphetamine. Toxicology Letters 188: 167-172.

Van Hout MC and Bingham T. (2012) “A costly turn on”: patterns of use and perceived

consequences of mephedrone based head shop products amongst Irish injectors.

International Journal of Drug Policy 23(3): 188-197.

Van Hout MC and Brennan R. (2010) Plant food for thought: A qualitative study of mephedrone

use in Ireland. Drugs: Education, Prevention and Policy 18(5): 371-381.

Van Hout MC and Brennan R. (2011a) “Bump and grind”: an exploratory study of mephedrone

users' perceptions of sexuality and sexual risk. Drugs and Alcohol Today 11: 93-103.

Van Hout MC and Brennan R. (2011b) Curiosity killed M-Cat: a post-legislative study on

mephedrone use in Ireland. Drugs: Education, Prevention and Policy 19(2): 156-162.

Van Hout MC and Brennan R. (2011c) ‘Heads held high’: an exploratory study of legal highs in

pre-legislation Ireland. Journal of Ethnicity in Substance Abuse 10: 256-272.

van Nieuwenhuijzen PS, McGregor IS, Hunt GE (2009) The distribution of [gamma]-

hydroxybutyrate-induced Fos expression in rat brain: comparison with baclofen.

Neuroscience 158:441–455.

1-26

Vardakou I, Pistos C and Spiliopoulou C. (2010) Drugs for youth via internet and the example of

mephedrone. Toxicology Letters 201(3): 191-195.

Wagner GC, Ricaurte GA, Seiden LS, et al. (1980) Long-lasting depletions of striatal dopamine

and loss of dopamine uptake sites following repeated administration of

methamphetamine. Brain Research 181: 151-160.

Warren L. (2012) Mom who choked and punched son, 3, while 'high on bath salts' pictured

running naked through street - moments before she was Tasered to death. Daily Mail, 14

June 2012.

Whalen J. (2010) In quest for “legal high,” chemists outfox law. Wall Street Journal, 29 October

2010.

Wilson JM and Kish SJ (1996) The vesicular monoamine transporter, in contrast to the dopamine

transporter, is not altered by chronic cocaine self-administration in the rat. Journal of

Neuroscience 16: 3507–3510.

Winstock A, Mitcheson L and Marsden J. (2010a) Mephedrone: still available and twice the

price. The Lancet 376: 1537.

Winstock A, Mitcheson LR, Deluca P, et al. (2011) Mephedrone, new kid for the chop?

Addiction 106(1): 154-161.

Winstock A and Wilkins C. (2011) ‘Legal highs’ The challenge of new psychoactive substances.

Series on Legislative Reform of Drug Policies, Number 16: Transnational Institute;

London: International Drug Policy Consortium.

Winstock AR, Marsden J and Mitcheson L. (2010b) What should be done about mephedrone?

British Medical Journal 340: c1605.

Winstock AR and Ramsey JD. (2010) Legal highs and the challenges for policy makers.

Addiction 105: 1685-1687.

Winterton N. (2010) Relevance, impact and academic spin. Clean Technologies and

Environmental Policy 12: 325-329.

Wise RA, Leone P, Rivest R, et al. (1995) Elevations of nucleus accumbens dopamine and

DOPAC levels during intravenous heroin self-administration. Synapse 21: 140–148.

Wood DM and Dargan PI. (2010) Mephedrone: A novel synthetic cathinone - a case series of

sympathomimetic toxicity associated with its use. Clinical Toxicology 48: 307.

1-27

Wood DM, Davies S, Greene SL, et al. (2010a) Case series of individuals with analytically

confirmed acute mephedrone toxicity. Clinical Toxicology 48: 924-927.

Wood DM, Davies S, Puchnarewicz M, et al. (2010b) Recreational use of mephedrone (4-

methylmethcathinone, 4-MMC) with associated sympathomimetic toxicity. Journal of

Medical Toxicology 6: 327-330.

Wood DM, Greene SL and Dargan PI. (2010c) Clinical pattern of toxicity associated with the

novel synthetic cathinone mephedrone. Emergency Medicine Journal 28(4): 280-282.

Wood DM, Measham F and Dargan PI. (2012) ‘Our favourite drug’: prevalence of use and

preference for mephedrone in the London night-time economy 1 year after control.

Journal of Substance Use 17(2): 91-97.

Wright MJ, Jr., Angrish D, Aarde SM, et al. (2012) Effect of ambient temperature on the

thermoregulatory and locomotor stimulant effects of 4-methylmethcathinone in wistar

and sprague-dawley rats. PLoS ONE 7: e44652.

2-1

2. Patterns of Brain Activation

_______________________________

2-2

Mephedrone (4-methylmethcathinone,‘meow’): acute

behavioural effects and distribution of Fos expression

in adolescent rats

Craig P. Motbey, Glenn E. Hunt, Michael T. Bowen, Suzanne Artiss and Iain S. McGregor.

Addiction Biology, 17: 409-422

2-3

Co-Author Contribution

McGregor, I.S.

Provided technical assistance, contributed to the research design and manuscript editing.... 15%

Hunt, G.E.

Provided technical assistance .....................................................................................................8%

Bowen, M.T.

Provided technical assistance .....................................................................................................8%

Artiss, S.

Provided technical assistance .....................................................................................................3%

TOTAL........................................................................................................................34%

2-4

2-5

2-6

2-7

2-8

2-9

2-10

2-11

2-12

2-13

2-14

2-15

2-16

2-17

3-1

3. Influence on Neurotransmitter Systems

_______________________________

3-2

Mephedrone in Adolescent Rats: Residual Memory

Impairment and Acute but Not Lasting 5-HT Depletion

Craig P. Motbey, Emily Karanges, Kong M. Li, Shane Wilkinson, Adam R. Winstock, John

Ramsay, Callum Hicks, Michael D. Kendig, Naomi Wyatt, Paul D. Callaghan and Iain S.

McGregor.

PLoS ONE, 7(9): e45473. doi:10.1371/journal.pone.0045473

3-3

Co-Author Contribution

McGregor, I.S.

Provided technical assistance, contributed to the research design and manuscript editing.... 10%

Callaghan, P.D.

Provided technical assistance..................................................................................................... 4%

Wyatt, N.

Provided technical assistance..................................................................................................... 1%

Kendig, M.D.

Provided technical assistance..................................................................................................... 4%

Hicks, C.

Provided technical assistance..................................................................................................... 4%

Ramsay, J.

Provided technical assistance..................................................................................................... 1%

Winstock, A.R.

Provided technical assistance..................................................................................................... 1%

Wilkinson, S.

Provided technical assistance..................................................................................................... 4%

Li, K.M.

Provided technical assistance..................................................................................................... 4%

Karanges, E.

Provided technical assistance..................................................................................................... 4%

TOTAL................................................................................................................37%

3-4

3-5

3-6

3-7

3-8

3-9

3-10

3-11

3-12

3-13

4-1

4. Potential for Addiction

_______________________________

4-2

High Levels of Intravenous Mephedrone (4-

Methylmethcathinone) Self-Administration In Rats: Neural

Consequences and Comparison with Methamphetamine

Craig P. Motbey1, Kelly Clemens

2, Nadine Apetz

1, Adam R. Winstock

3, John Ramsey

4, Kong M. Li

5, Naomi

Wyatt6, Paul D. Callaghan

6, Michael T. Bowen

1, Jennifer Cornish

2, Iain S. McGregor

1

1School of Psychology, University of Sydney, NSW 2006, Australia

2School of Psychology, Macquarie University, NSW 2109, Australia

3Institute of Psychiatry, Kings College, University of London, UK

4TICTAC Communications Ltd., St George’s College, University of London, UK

5Department of Pharmacology, University of Sydney, NSW 2006, Australia

6Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia

Manuscript submitted to “Journal of Psychopharmacology”

4-3

Co-Author Contribution

Cornish, J.

Provided technical assistance ...................................................................................................10%

McGregor, I.S.

Provided technical assistance, contributed to the research design and

manuscript editing .....................................................................................................................10%

Clemens, K.

Provided technical assistance .....................................................................................................8%

Bowen, M.T.

Assisted with statistical analyses................................................................................................7%

Callaghan, P.D.

Provided technical assistance .....................................................................................................2%

Li, K.M.

Provided technical assistance .....................................................................................................2%

Apetz, N.

Provided technical assistance .....................................................................................................1%

Wyatt, N.

Provided technical assistance .....................................................................................................1%

Ramsey, J.

Provided technical assistance .....................................................................................................1%

Winstock, A.R.

Provided technical assistance .....................................................................................................1%

TOTAL...................................................................................................................................... 43%

4-4

4-5

4-6

4-7

4-8

4-9

4-10

4-11

4-12

4-13

4-14

4-15

4-16

4-17

5-1

5. General Discussion

_______________________________

5-2

5.1 Understanding the Mechanisms of MMC

Overall, the findings presented here and elsewhere have been largely in accord with what

would be predicted based upon the anecdotal reports of MMC consumers regarding the euphoric,

entactogenic and stimulant properties of the drug. Users frequently described MMC using

analogies to MDMA and METH (Erowid, 2012). Both MDMA and METH induce significant c-

Fos expression in the prelimbic cortex, caudate-putamen, Islands of Calleja, nucleus accumbens

core, ventral tegmental area and dorsal raphe. METH additionally activates the ventral orbital

cortex, lateral orbital cortex, barrel fields 6a/6b, and primary motor cortex, while MDMA

activates the nucleus accumbens shell, supraoptic nucleus, anterior paraventricular thalamic

nucleus, central and basolateral amygdala, Edinger-Westphal nucleus and lateral parabrachial

nucleus. MMC induces significant levels of c-Fos expression in all of these areas.

The pattern of brain activation demonstrated by c-Fos expression in MMC-dosed rats

resembles an MDMA/METH hybrid, in accord with early descriptions of the drug (Erowid,

2012). Unsurprisingly therefore, MMC intoxication has a strong acute impact upon NE, DA and

5-HT systems (Hadlock et al., 2011; Kehr et al., 2011; Motbey et al., 2012; Shortall et al., 2013).

Acute MMC induces a rapid and substantial efflux of all three of these neurotransmitters

(Baumann et al., 2012; Baumann et al., 2012; Cameron et al., 2012; Kehr et al., 2011; Wright et

al., 2012), and in all three cases this release is mediated at least partially by the reversal of

transporter function (Baumann et al., 2012). However, it appears that both the speed of release

and the pace of metabolism may substantially differ between these neurotransmitter systems

(Kehr et al., 2011; Motbey et al., 2011). MMC effects upon DAT function appear to be similar to

those of METH (Cameron et al., 2012), in that both drugs induce a depolarizing current in DAT,

thereby acting as an excitatory ligand and driving DA release. In contrast, the related synthetic

cathinone MDPV (like MMC, a common component of “bath salts” and “plant food” drugs)

produces a hyperpolarizing current in DAT (an effect similar to that seen with cocaine),

inhibiting DA reuptake (Cameron et al, 2012; Baumann et al., 2013). However, the effects of

MMC and MDPV on DA levels are synergistically enhanced when these two drugs are given in

combination, due to the differing temporal pattern of each drug’s effects (Cameron et al., 2013).

5-3

MMC appears to carry a substantial potential for addiction and uncontrolled bingeing

(Hadlock et al., 2011; Lisek et al., 2012; Motbey et al., 2013; Ramoz et al., 2012). User reports

frequently refer to the addictive potential of the drug (eg: Erowid, 2009; Erowid, 2010) and a

diverse range of animal models of addictive behaviour have displayed positive results with

MMC (Hadlock et al, 2011; Lisek et al., 2012; Motbey et al., 2013; Ramoz et al., 2012;

Robinson et al., 2012). Accumulating evidence from multiple sources strongly suggests that

MMC consumption may lead to lasting cognitive impairment (den Hollander et al., 2012;

Freeman et al., 2012; Motbey et al., 2012). However, no researcher has yet provided an

explanation as to the mechanism of this cognitive impairment or any direct evidence of

significant MMC-induced neurotoxicity. We also do not yet possess any evidence to explain the

differing temporal patterns of MMC’s expression or metabolism.

Behavioural observations in the first experiment of this thesis (Chapter 2; Motbey et al.,

2011) suggested thigmotaxic effects, which may relate to the entactogenic reports from human

consumers. This suggestion was further supported by the strong c-Fos expression found in the

somatosensory barrel fields of MMC-dosed rats, as primary sensory areas such as these could be

expected to be stimulated by entactogenic or thigmotaxic intoxication. MMC in humans appears

to combine entactogenic with pro-social effects (Erowid, 2012). As with MDMA, it may be the

case than any prosocial effects of MMC are mediated by the oxytocin system (Green et al.,

2003). However, no study has yet directly addressed the question of MMC-related oxytocin and

vasopressin expression or their influence on MMC-driven behaviour. Pursuing this question

provides an obvious future target for MMC research.

The c-Fos expression demonstrated in the supraoptic nucleus (SON) of rats, an area rich

in both oxytocin and vasopressin (Kohno et al., 2008), also suggests a potential interaction of

MMC with these systems. Activation in areas associated with oxytocin receptors (such as the

central amygdala) also provides support. However, the levels of SON activation found in MMC-

dosed rats were relatively mild compared to those typically found with MDMA or GHB

(Hargreaves et al., 2007; van Nieuwenhuijzen et al., 2010), suggesting that the interaction of

MMC with these systems may be of a lower intensity than that seen with these other drugs.

The extremely strong c-Fos expression demonstrated in the striatum of MMC-dosed rats

suggested a 5-HT/dopamine mode of action. This suggestion was later confirmed by the research

5-4

described in Chapter 3 of this thesis (Motbey et al., 2012). Neurochemical analysis of brains

extracted from MMC-dosed rats 1 h post injection revealed contrasting effects on DA and 5-HT.

In both the striatum and hippocampus, 5-HT levels were substantially lowered (by approximately

30%) while 5-HIAA/5-HT ratios (a measure of 5-HT metabolic turnover) were increased. In

contrast, striatal DA levels were significantly increased while measures of DA metabolism

((DOPAC+HVA)/DA ratios) were decreased.

These findings suggested that MMC may induce a very rapid efflux of 5-HT (largely

metabolized by the 1 h post dosing point) and a more gradual and sustained increase in DA,

possibly by inhibition of reuptake or metabolism. Such a hypothesis is consistent with other

evidence that MMC acts preferentially as a 5-HT releaser, but also induces a vigorous efflux of

DA (Baumann et al., 2011, Baumann et al., 2012). Evidence from a number of studies has

demonstrated that MMC blocks the uptake of NE, DA and 5-HT (Hadlock et al., 2011; Lopez-

Arnau et al., 2012; Martinez-Clemente et al., 2012; Simmler et al., 2012), and a recent release-

assay study (Baumann et al., 2012) showed that MMC functions as a non-selective transporter

substrate, actively stimulating the release of NE, DA and 5-HT. Given these facts, it follows that

there must be some alteration in metabolic rates in order to explain the contrasting pictures of

DA and 5-HT levels from the brains of rats examined 1 hr after MMC dosing (Motbey et al.,

2012; Chapter 3). However, the close investigation of the mechanisms required to clarify exactly

what mechanism is driving these differences remains to be done.

One plausible mechanism for the reduction in DA metabolism would be a selective

inhibition of monoamine oxidase B (MAO-B). Indeed, research suggests that the basic cathinone

molecule does act in this manner (Kelly, 2011). However, an investigation of the interaction of

monoamine oxidases with a wide variety of cathinones revealed that MMC does not substantially

impact MAO-A or B (Osorio-Olivares et al., 2004). Therefore, any MMC-induced inhibition of

DA metabolism must utilise an alternative mechanism, possibly via the next step in the

metabolic process, aldehyde dehydrogenase (ALDH). This potential for MMC-induced

inhibition of ALDH raises the possibility that MMC and alcohol may produce a synergistic

neurotoxicity similar to that seen with MDMA (Izco et al., 2007; Upreti et al., 2009). There is

also some suggestion that the combination of MMC and alcohol may induce cardiac impairment

(McGaw and Kankam, 2010), although there is no current evidence to suggest that this is more

5-5

than a reversible acute effect of the drugs. Due to the influence of MMC on cardiac function

(Nicholson et al., 2010; Vardakou et al., 2010; Wood et al., 2010a), an investigation has been

made into the potential for MMC-induced cardiac injury in the rat (Meng et al., 2011). While this

study confirmed the strong acute effects of MMC on cardiac function, it did not find any

evidence of lasting harm.

Several authors have investigated the possibility of lasting neurotoxicity or

neuroadaptation in response to both acute and chronic MMC consumption (Hadlock et al., 2011;

Angoa-Pérez et al., 2012). In agreement with the results presented here in Chapter 3 (Motbey et

al., 2012), a group of mice given a binge-style treatment of 4 x 40mg/kg I.P. MMC (doses at 2

hour intervals) failed to display any signs of toxicity, inflammation or any lasting alteration in

neurotransmitter systems 7 days afterwards (Angoa-Pérez et al., 2012). However, a study in

which rats were dosed with 4 x 25 mg/kg S.C. MMC (also at 2 hour intervals) while maintained

at substantially elevated ambient temperatures (≥ 27° C) did find lasting decreases in

hippocampal 5-HT uptake when tested 7 days after treatment (Hadlock et al., 2011).

While this research may present an important lead towards the discovery of MMC-

induced neurotoxicity, the complicating variable of temperature and contrasting results of other

studies (Angoa-Pérez et al., 2012; Motbey et al., 2012; Simmler et al., 2012) constrains the

breadth of conclusions to be drawn from it. The interaction of MMC with temperature is

particularly problematic, as studies have shown that the effect of MMC on thermoregulation can

be substantially altered by variations in dosage and environmental conditions. While singly-

housed rats given one relatively low dose of MMC have demonstrated a hypothermic response

(Shortall et al., 2013), studies utilising binge-dosing protocols with group-housed animals have

shown hyperthermic responses at both normal and elevated ambient temperatures (Baumann et

al., 2011; Hadlock et al., 2011). In addition, elevated ambient temperatures and group housing

have both been shown to abolish the hypothermic response even in singly-dosed animals (Miller

et al., 2012; Shortall et al., 2013). Recent work has also revealed strain-based differences in

thermal response (Aarde et al., 2013, Wright et al., 2012). As variations in ambient temperature

and thermoregulatory impairments have been shown to have a significant influence upon

neurotoxicity with other stimulants (Malberg and Seiden, 1998), careful consideration of this

variable is necessary when investigating the toxicity of such drugs.

5-6

Of the research in this thesis, the results presented in chapters 2 and 3 derive from Wistar

rats while those of chapter 4 involved Sprague-Dawley animals. Although later work has shown

that MMC-based self-administration characteristics do not sharply differ between Wistar and

Sprague-Dawley rats (Aarde et al., 2013), the strain-based diversity of MMC responses shown in

thermoregulatory and other measures (Aarde et al., 2013, Wright et al., 2012) suggests that this is

a factor that deserves consideration during further investigation.

In the rat, MMC binds to the 5-HT, NE and DA transporters (SERT, NET and DAT) with

a selectivity and potency similar to MDMA, reversing the action of these transporters and

driving neurotransmitter release (Baumann et al., 2011). Similar results have been found with

transporter-transfected human embryonic kidney cells (Simmler et al., 2012) and human DAT-

expressing Xenopus laevis oocytes (Cameron et al., 2012). The influence of MMC on 5-HT, NE

and DA has been demonstrated by an increasing variety of studies (Baumann ete al., 2011;

Hadlock et al., 2011; Kehr et al., 2011; Motbey et al., 2012, 2013; Shortall et al., 2013). Acute

MMC treatment evokes dose-related increases in 5-HT efflux in the nucleus accumbens, again

similar to MDMA on a per-mg basis. DA efflux in the nucleus accumbens after MMC treatment

appears to exceed that found with MDMA, albeit not to the extent seen with methamphetamine

(METH) (Baumann et al., 2011; Kehr et al., 2011; Wright et al., 2012).

Although MMC has a profound acute influence on 5-HT and DA systems, no lasting

alteration could be detected in the levels of these neurotransmitters or their metabolites 47 days

post-treatment (Chapter 3, Motbey et al., 2012). This finding was somewhat surprising given the

evidence of persistent effects upon neurotransmitter systems with similar drugs such as MDMA

(Beaumann et al., 2007). While this result is in agreement with the findings of Angoa-Pérez et al.

(2012) and Simmler et al. (2012), it does present a contrast with the decreases in rat hippocampal

5-HT levels 7 days post-treatment discovered by Hadlock et al. (2011). The recent work of den

Hollander et al. (2012) further complicates the pictures, with their finding of an MMC-induced

reduction in HVA two weeks after a four-day binge treatment in mice. However, a group of rats

given similar treatment in the same study showed no significant neurochemical changes, in

agreement with the findings presented in Chapter 3 of this thesis (Motbey et al., 2012). Although

it provides useful early evidence of possible species differences in the MMC response, the

reduction of HVA in den Hollander’s (2012) mice does little to clarify the picture regarding

MMC neurotoxicity.

5-7

However, the possibility of at least short-term MMC-induced alteration in

neurotransmitter levels was supported by the results presented in Chapter 4 (Motbey et al.,

2013). Striatal tissue collected 3 days post-treatment from rats given 44 days of access to self-

administered MMC was found to have significantly reduced levels of 5-HIAA. However, the

finding of a lack of lasting alteration in 5-HIAA levels amongst rats given brief chronic MMC

(Motbey et al., 2012) suggests that this 5-HIAA reduction may have been destined to fade if

given more than 3 days of recovery. Although it appears that any lasting influence of MMC

consumption on neurotransmitter systems is likely to be subtle, the possibility of such an effect

should not be ruled out. Further research is obviously required to clarify this point.

Regardless of whether or not the mechanism is as straightforward as a simple alteration in

neurotransmitter levels, there is significant behavioural evidence that MMC use can induce

cognitive impairment. Rats given 10 days of 30 mg/kg I.P. MMC show a clear deficit in

recognition memory when tested 35 days post-dosing (Motbey et al., 2012; Chapter 3). This

finding is in agreement with the prose recall impairment displayed by human MMC users

(Freeman et al., 2012), and has since been further supported by the finding of an MMC-induced

impairment of working memory in mice when tested by a T-maze spontaneous alternation task

two weeks after a four day binge treatment (den Hollander et al., 2012). These findings in

combination present the clearest evidence to date regarding MMC neurotoxicity or adverse

neuroadaptation. Although the direct physical cause has yet to be located, it appears clear that

MMC consumption can induce cognitive impairment, and that this impairment may be persistent.

Identifying the mechanisms behind this cognitive impairment is a clear priority for further

research.

The evidence from animal studies also supports the anecdotal reports of MMC’s

addictiveness (eg: Erowid, 2011; 2012). MMC has been shown to induce a conditioned place

preference in rats, mice and flatworms (Lisek et al., 2012; Ramoz et al., 2012; Motbey et al.,

2013) and maintain self-administration behaviour in rats (Hadlock et al., 2011; Aarde et al.,

2013). Levels of MMC-seeking responses in rats (Motbey et al., 2013; Chapter 4) given

extended access to 2 h day MMC nosepoke-triggered self administration dramatically exceeded

those seen in methamphetamine-dosed comparison animals. This response was achieved with no

pretraining and may represent the most vigorous self-administration response ever found with

any drug. Under progressive ratio testing conditions, rats continued to work for MMC infusions

5-8

even when response/reward ratios rose into the hundreds (Motbey et al., 2012). MMC induces

high levels of c-Fos activation across the mesolimbic dopamine pathway (Motbey et al., 2011)

and stimulates a rapid and substantial release of DA and 5-HT into the nucleus accumbens in rats

(Kehr et al., 2011). Overall, the evidence suggests that MMC is a drug with an extremely high

risk of uncontrolled consumption.

Intravenous MMC carries a high danger of extremely serious physical harm (Dorairaj et

al., 2011; Pharris et al., 2011; Russo et al., 2012; Van Hout and Bingham, 2012). The exact cause

of these harms are as yet unclear, but they appear to be due to a combination of the behavioural

characteristics of MMC use (i.e.: repeated dosing episodes while intoxicated, leading to poor

injection hygeine) and an innate toxicity of the drug (Dorairaj et al., 2011).

5.2 Responding to MMC

The evidence as to the impact of legal restrictions on MMC use is mixed. Although

prohibition in the U.K. has driven an increase in the price of MMC (Winstock et al., 2010a), a

survey of London club-goers in mid 2010 (3 months after prohibition) found that 27% reported

use or intention to use on the night of the survey (Measham et al., 2011). A follow-up survey on

the same site one year later reported that this figure had risen to 41% (Wood et al., 2012). In both

cases, MMC was the most consumed drug reported on the night. Participants in the second

survey were also questioned as to their favourite recreational drug; MMC was the leading

contender, taking 20.4% of the vote (ahead of cocaine at 14.9%) (Wood et al., 2012). It appears

likely that MMC and other synthetic cathinones will remain part of the contemporary drug

culture for some time to come (Brennan and Van Hout, 2012).

While it may have ended its phase of rapid growth, MMC still maintains a substantial

presence amongst UK drug users (Archer et al., 2013). Australian rates of MMC consumption

appear to have remained relatively slight (Sindicich and Burns, 2012) except amongst some

small samples drawn from a population of regular drug users (Bruno et al., 2012). However,

reports from around the world show that MMC use continues to be popular in many areas

regardless of legality (Archer et al., 2012; Brennan and Van Hout, 2012; Bruno et al., 2012;

5-9

Brunt et al., 2010; US Department of Justice Drug Enforcement Agency, 2011; Wood et al.,

2012).

Some recent data suggests that MMC use in the UK has ceased increasing and may have

begun to decline (Measham et al., 2012). However, this study was deliberately targeted upon a

sample of “mainstream” nightclubs rather than the dance-culture venues typical of much earlier

research. It is unclear how much of the apparent decline is due to this factor, as lower rates of

novel psychoactive use are not unexpected amongst this subject group relative to the drug early-

adopters of the dance music community. If there is a genuine decline, it is also unclear what

proportion relates to increasing legal restriction versus an increased awareness within the drug

using community of the undesirable effects of MMC consumption or competitive displacement

by newly-arriving novel psychoactives.

The reports of injection-related harms occuring in human populations (Dorairaj et al.,

2011; Pharris et al., 2011; Russo et al., 2012; Van Hout and Bingham, 2012) suggest that

responses to MMC should include a strong emphasis on discouraging intravenous use. While

non-injecting use of MMC is not without danger, the dramatic impact of MMC on injecting users

justifies a focus upon harm minimization and diversion to less damaging methods of

consumption.

There may also be worthwhile harm minimization strategies to be pursued with non-

injecting users. For example, the tendency of MMC to induce uncontrolled bingeing suggests

that minimizing on-hand supplies of the drug may be a useful strategy. However, while MMC

bingeing may be reduced if users acquire their drugs via relatively smaller (although possibly

more frequent) purchases, such an approach increases the exposure of users to the legal and

physical risks of illicit drug purchasing transactions. The effect of prohibition is to encourage

fewer, larger purchasing transactions and promote stockpiling, which in turn provides the setting

for increased binge consumption.

Although it has been suggested that the rise of novel psychoactive drugs provides an

opportunity to “consider the trial of alternative policy and legislative approaches to drug control”

(Winstock and Wilkins, 2011; Hughes and Winstock, 2012), it appears more likely that the

response to MMC will follow the traditional patterns of simplistic prohibition that are so widely

seen to be counterproductive (United Nations Office on Drugs and Crime, 2009; Winstock et al.,

2010b; Winstock and Wilkins, 2011; Australia21, 2012; Hughes and Winstock, 2012). Legal

5-10

prohibition and the interdiction of precursor chemicals may eventually prove effective in limiting

supply, as appears to have been the case with MDMA (Winstock and Wilkins, 2011). However,

even if the prohibition of MMC does prove to be successful in reducing use, such an approach

provides a temporary respite at best.

A large number of easily manufactured cathinone variants exist, most of which are likely

to be psychoactive to some degree. The flexibility of modern chemistry allows for an almost

inexhaustible supply of alternative molecules. In 2010 alone, European authorities identified 41

new psychoactive drugs (EMCDDA, 2011). Just the contents of the phenethylamine family

(Shulgin, 1991) are enough to occupy the global research community for decades, to say nothing

of the cathinones, the tryptamines and so on.

To date, the main reaction has been to reflexively ban each new molecule as it appears on

the market, frequently before any adequate evidence has been presented as to whether the

potential harms of the substance justify such a response (Hughes and Winstock, 2012; Measham

et al., 2010). This approach creates the perverse risk of driving users towards an endless stream

of novel drugs, many of which may be more harmful than their already-established alternatives.

Although evidence as to relative risk is still being collected, it appears that this is exactly what

has happened with the substitution of MMC for MDMA consumption. While there is still no

conclusive evidence of MMC-driven neurotoxicity, it appears clear that MMC induces persistent

cognitive impairment and carries an extremely high risk of addiction and excessive consumption.

While MDMA use is not without dangers of its own (Burgess et al., 2000), it appears clear that it

is a lower-risk option than MMC.

There is no reason to believe that prohibition is likely to be any more successful with the

continuously expanding list of new drugs than it has been with the drugs of the 20th

century. An

effective response to the fast-moving realities of the modern drug market is likely to require a

radical departure from business as usual. The development of systems to facilitate the rapid

assessment and provisional classification of emerging drugs appears to be a clear priority

(Winstock and Ramsey, 2010; Hughes and Winstock, 2012). However, any system of provisional

classification followed by later refinement necessarily implies the occasional lessening of

restrictions on drugs found to be relatively less harmful. The reality of the current drug debate

renders such an action politically unpalatable to governments in many countries. Therefore, it is

essential that such classification bodies be rigorously independent of political influence if they

5-11

are to function as intended (Nutt, 2009a; Nutt, 2009b; Reuter, 2011; Winstock and Wilkins,

2011). The recent series of controversies involving the U.K. Advisory Council on the Misuse of

Drugs (Nutt, 2009b; Nutt, 2010; Sare, 2011) provide a stark example of the failure of such

systems if this requirement is not met.

5.3 Dangerous Possibilities: The Example of MPTP

The history of MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) suggests a chilling

possibility that argues for urgent action in response to this situation. In 1982, the accidental

production of MPTP by illicit chemists in the U.S.A. led to a micro-epidemic of drug-induced

Parkinsonism (Ballard et al., 1985; Langston and Palfreman, 1995). Closely related to the

synthetic heroin substitute MPPP (1-methyl-4-phenyl-4-propionoxypiperidine, an “enhanced”

variant of pethidine which was the intended product of most of the illicit chemists responsible for

this incident), MPTP is a highly potent and selective dopaminergic neurotoxin that is commonly

used today to create animal models of Parkinson’s disease (Jackson-Lewis et al., 2012).

However, the dangers of MPTP were almost entirely unknown to science in 1982 (Langston and

Palfreman, 1995), much as is the case with many emerging drugs of abuse today. The first

warning of the MPTP incident came when a wave of young heroin addicts appeared in a variety

of hospitals with mysteriously rapid and premature symptoms of Parkinson’s disease (Figure 1).

5-12

Figure 1. Survivors of the MPTP epidemic. This photo was taken in 1991, 9 years after the poisoning

incident. George Carillo (standing left) and Juanita Lopez (standing right) had achieved a substantial

degree of recovery following fetal-tissue transplants. Bill Silvey (seated left), Connie Sainz (seated

centre) and David Silvey (standing right) were still profoundly impaired by their illness. Image from

Langston and Palfreman (1995, p150).

5-13

While the history of MPTP was tragic for those directly affected, from the point of view

of the wider community the incident could be seen as a fortunate escape from calamity. The

small scale of MPTP production and the rapid action of MPTP toxicity allowed the situation to

be detected and contained before it affected more than a relatively small number of people

(Langston and Palfreman, 1995). If we continue with the revolving door of reflexively banning

each new psychoactive drug as it appears, thereby encouraging further exploration with novel

pharmaceuticals, it is only a matter of time before the recurrence of a situation analogous to that

of MPTP. However, next time we may not be so lucky.

Two factors combine to exacerbate the danger. The rapid and worldwide spread of novel

psychoactive drugs (as seen with MMC) in advance of any substantial research into toxicity is

the first factor. The second issue is the subtle and progressive nature of many neurological

disorders, with substantial and lasting neural damage often preceding overt behavioural

symptoms and “incubation” periods varying from instantaneous to life-long. The combination of

these factors provides for the alarming possibility of a global pandemic of drug-induced

neurological injury. A hypothetical novel psychoactive drug which induced a slowly-appearing

neurotoxic impairment, when combined with market penetration of speed and depth similar to

MMC, has the potential to cause an immense amount of harm before the danger of such a drug is

even detected by science.

5.4 Conclusion

Overall, it would appear that the acute risks of MMC consumption, while real, are

nonetheless relatively mild, perhaps somewhere towards the midpoint between MDMA and

methamphetamine. However, although there is no conclusive evidence of direct MMC

neurotoxicity, several studies strongly suggests that MMC consumption may induce lasting

cognitive impairment (den Hollander et al., 2012; Freeman et al., 2012; Motbey et al., 2012). In

addition, the behavioural risks of MMC intoxication and addiction present a substantial harm in

themselves (Motbey, 2012; Van Hout and Bingham, 2012; Van Hout and Brennan, 2010; Van

Hout and Brennan, 2011a; Van Hout and Brennan, 2011b), and the physical damage to the

minority of MMC users consuming the drug by injection is extreme (Dorairaj et al., 2011; Van

Hout and Bingham, 2012).

5-14

Looking beyond the direct impact of MMC use, the example of MMC is also useful as a

case study for the wider problem of novel psychoactive drug consumption. The increasing pace

and diversity of recreational drug markets worldwide has altered the situation to such an extent

that the already-flawed conventional approach to drug control has become actively dangerous. At

the very least, the situation calls for a considerable increase in the resources allocated to the

investigation of the toxicity of emerging drugs of abuse. However, although essential, such a

response represents at best a band-aid reaction to the new reality.

The rise of MMC was arguably a consequence of the prohibition of MDMA. An

increased focus upon suppression of MMC is most likely to shift the market towards a greater

reliance on the apparently more dangerous MDPV. Eliminating MDPV will create a market for

the next new drug, and the next, and the next. Each iteration of this process carries the risk of

another MPTP, and the iterations are accelerated by our prohibitionist responses. The novel

psychoactive market is a hydra that grows more dangerous with each head we remove.

An increased focus on toxicity research is a necessary but not sufficient step towards

addressing the underlying problem, which is rooted in the lack of concordance between the

relative risks of drug harms and varying degrees of legal restriction (Hughes and Winstock,

2012; Nutt et al., 2007; Nutt et al., 2010; Winstock and Ramsey, 2010). Even with greatly

enhanced resources, the psychopharmacology community cannot possibly keep ahead of the

ever-expanding modern drug market. Effects that take years to manifest require years to

research, and the current situation is such that hundreds of thousands of people could potentially

be severely affected before any evidence of danger came to the attention of science. Eventually,

society may have to face the difficult choice of deciding to openly permit a limited set of

tolerably-dangerous drugs that are sufficient to satisfy at least most of the apparently inflexible

demand. If we wish to avert the substantial risks associated with the emergence of novel

psychoactive drugs, scientific research must be accompanied by a substantial reform of the way

in which governments worldwide seek to control the use of these drugs.

5-15

5.5 References

Aarde SM, Angrish D, Barlow DJ, et al. (2013) Mephedrone (4‐methylmethcathinone) supports

intravenous self‐administration in Sprague‐Dawley and Wistar rats. Addiction Biology.

Epub ahead of print 30 Jan 2013, doi: 10.1111/adb.12038

Angoa-Pérez M, Kane MJ, Francescutti DM, et al. (2012) Mephedrone, an abused psychoactive

component of ‘bath salts’ and methamphetamine congener, does not cause neurotoxicity

to dopamine nerve endings of the striatum. Journal of Neurochemistry 120: 1097-1107.

Archer J, Dargan P, Hudson S, et al. (2013) Analysis of anonymous pooled urine from portable

urinals in central London confirms the significant use of novel psychoactive substances.

Quarterly Journal of Medicine. Epub ahead of print 22 November 2012. doi:

10.1093/qjmed/hcs219

Australia21. (2012) The prohibition of illicit drugs is killing and criminalising our children and

we are all letting it happen. University of Sydney: Australia21.

Ballard PA, Tetrud JW and Langston JW. (1985) Permanent human Parkinsonism due to

1‐methy 1–4‐phenyl‐1, 2, 3, 6‐tetrahydropyridine (MPTP). Neurology 35: 949-949.

Baumann MH, Wang X and Rothman RB. (2007) 3, 4-methylenedioxymethamphetamine

(MDMA) neurotoxicity in rats: a reappraisal of past and present findings.

Psychopharmacology 189: 407-424.

Baumann MH, Ayestas MA, Partilla JS, et al. (2011) The designer methcathinone analogs,

mephedrone and methylone, are substrates for monoamine transporters in brain tissue.

Neuropsychopharmacology 37(5): 1192-1203.

Baumann MH, Partilla JS and Lehner KR. (2012) Psychoactive “bath salts”: not so soothing.

European Journal of Pharmacology. doi: 10.1016/j.ejphar.2012.11.020.

5-16

Baumann MH, Partilla JS, Lehner KR, Thorndike EB, Hoffman AF, et al. (2013) Powerful

cocaine-like actions of 3,4-methylenedioxypyrovalerone (MDPV), a principal constituent

of psychoactive 'bath salts' products. Neuropsychopharmacology 38: 552-562.

Brennan R and Van Hout MC. (2012) Miaow miaow: a review of the new psychoactive drug

mephedrone. Drugs and Alcohol Today 12: 241-253.

Bruno R, Matthews AJ, Dunn M, et al. (2012) Emerging psychoactive substance use among

regular ecstasy users in Australia. Drug and Alcohol Dependence 124: 19-25.

Brunt TM, Poortman A, Niesink RJ, et al. (2010) Instability of the ecstasy market and a new kid

on the block: mephedrone. Journal of Psychopharmacology 25(11): 1543-1547.

Burgess C, O’Donohoe A and Gill M. (2000) Agony and ecstasy: a review of MDMA effects

and toxicity. European Psychiatry 15: 287-294.

Cameron KN, Kolanos R, Solis E, et al. (2012) Bath salts components mephedrone and

methylenedioxypyrovalerone (MDPV) act synergistically at the human dopamine

transporter. British Journal of Pharmacology 168: 1750-1757.

Cameron K, Kolanos R, Verkariya R, et al. (2013) Mephedrone and

methylenedioxypyrovalerone (MDPV), major constituents of “bath salts,” produce

opposite effects at the human dopamine transporter. Psychopharmacology: 1-7.

den Hollander B, Rozov S, Linden AM, et al. (2012) Long-term cognitive and neurochemical

effects of “bath salt” designer drugs methylone and mephedrone. Pharmacology,

Biochemistry and Behavior 103: 501-509.

Dorairaj J, Healy C, McMenamin M, et al. (2011) The untold truth about “bath salt” highs: A

case series demonstrating local tissue injury. Journal of Plastic, Reconstructive &

Aesthetic Surgery 65(2): e37-41.

European Monitoring Centre for Drugs and Drug Addiction (EMCDDA). (2010) Europol–

EMCDDA joint report on a new psychoactive substance: 4-methylmethcathinone

(mephedrone). Report, EMCDDA, Lisbon, March 2010.

5-17

Erowid. (2009) Dangerously addictive, but awesome. Available at:

http://www.erowid.org/experiences/exp.php?ID=76345 (accessed 17 August 2012).

Erowid. (2010) Serious addiction potential. Available at:

http://www.erowid.org/experiences/exp.php?ID=85851 (accessed 17 August 2012).

Erowid. (2012) Erowid experience vaults: 4-methylmethcathinone (also mephedrone, 4-MMC,

meow, m-cat, 2-methylamino-1-p-tolylpropan-1-one) main index. Available at:

http://www.erowid.org/experiences/subs/exp_4Methylmethcathinone.shtml (accessed 17

August 2012).

Freeman TP, Morgan CJA, Vaughn-Jones J, et al. (2012) Cognitive and subjective effects of

mephedrone and factors influencing use of a ‘new legal high’. Addiction 107: 792-800.

Green AR, Mechan AO, Elliott JM, et al. (2003) The pharmacology and clinical pharmacology

of 3,4-methylenedioxymethamphetamine (MDMA, "ecstasy"). Pharmacological Reviews

55: 463-508.

Hadlock GC, Webb KM, McFadden LM, et al. (2011) 4-methylmethcathinone(mephedrone):

neuropharmacological effects of a designer stimulant of abuse. Journal of Pharmacology

and Experimental Therapeutics 339: 530-536.

Hargreaves GA, Hunt GE, Cornish JL, et al. (2007) High ambient temperature increases 3,4-

methylenedioxymethamphetamine (MDMA, "ecstasy")-induced fos expression in a

region-specific manner. Neuroscience 145: 764-774.

Hughes B and Winstock AR. (2012) Controlling new drugs under marketing regulations.

Addiction. Epub ahead of print 31 January 2012. doi: 10.1111/j.1360-0443.2011.03620.x.

Izco M, Orio L, O’Shea E, et al. (2007) Binge ethanol administration enhances the MDMA-

induced long-term 5-HT neurotoxicity in rat brain. Psychopharmacology 189: 459-470.

Kehr J, Ichinose F, Yoshitake S, et al. (2011) Mephedrone, compared to MDMA (ecstasy) and

amphetamine, rapidly increases both dopamine and serotonin levels in nucleus

accumbens of awake rats. British Journal of Pharmacology 164: 1949-1958.

5-18

Kelly JP. (2011) Cathinone derivatives: a review of their chemistry, pharmacology and

toxicology. Drug Testing and Analysis 3: 439-453.

Kohno D, Nakata M, Maejima Y, et al. (2008) Nesfatin-1 neurons in paraventricular and

supraoptic nuclei of the rat hypothalamus coexpress oxytocin and vasopressin and are

activated by refeeding. Endocrinology 149: 1295-1301.

Lisek R, Xu W, Yuvasheva E, et al. (2012) Mephedrone (‘bath salt’) elicits conditioned place

preference and dopamine-sensitive motor activation. Drug and Alcohol Dependence.

Epub ahead of print 29 May 2012. doi 10.1016/j.drugalcdep.2012.04.021.

López‐Arnau R, Martínez‐Clemente J, Pubill D, et al. (2012) Comparative neuropharmacology

of three psychostimulant cathinone derivatives: butylone, mephedrone and methylone.

British Journal of Pharmacology 167: 407-420.

Martinez-Clemente J, Escubedo E, Pubill D, et al. (2011) Interaction of mephedrone with

dopamine and serotonin targets in rats. European Neuropsychopharmacology 22: 231-

236.

McGaw C and Kankam O. (2010) The co-ingestion of alcohol and mephedrone-an emerging

cause of acute medical admissions in young adults and a potential cause of

tachyarrhythmias. The West London Medical Journal 2: 9-13.

Measham F, Moore K and Newcombe R. (2010) Tweaking, bombing, dabbing and stockpiling:

the emergence of mephedrone and the perversity of prohibition. Drugs and Alcohol

Today 10: 14-21.

Measham F, Moore K and Østergaard J. (2011) Mephedrone,“bubble” and unidentified white

powders: the contested identities of synthetic “legal highs”. Drugs and Alcohol Today 11:

137-146.

Measham F, Moore K and Welch Z. (2012) Emerging drug trends in lancashire: nightclub

surveys. Lancaster University, available from

http://ldaat.org/files/emerging_trends_third_report.pdf (last accessed 23 Nov 2012).

5-19

Motbey CP, Clemens KJ, Apetz N, Winstock AR, Ramsey J, Li KM, Wyatt N, Callaghan PD,

Bowen MT, Cornish J, McGregor IS (2013). High levels of intravenous mephedrone (4-

methylmethcathinone) self-administration in rats: neural consequences and comparison

with methamphetamine. Journal of Psychopharmacology. Epub ahead of print June 5,

2013, doi: 10.1177/0269881113490325.

Motbey CP, Hunt GE, Bowen MT, et al. (2011) Mephedrone (4-methylmethcathinone, ‘meow’):

acute behavioural effects and distribution of fos expression in adolescent rats. Addiction

Biology 17(2): 409-422.

Motbey CP, Karanges E, Li KM, et al. (2012) Mephedrone in adolescent rats: residual memory

impairment and acute but not lasting 5-HT depletion. PLoS ONE 7(9): e45473.

Nutt D. (2009a) Estimating drug harms: a risky business? The Eve Saville Lecture, The Centre

for Crime and Justice Studies (CCJS), King’s College, London.

Nutt D. (2009b) Government vs science over drug and alcohol policy. The Lancet 374: 1731-

1733.

Nutt D. (2010) Nutt damage? Author's reply. The Lancet 375: 724.

Osorio-Olivares M, Rezende MC, Sepulveda-Boza S, et al. (2004) MAO inhibition by

arylisopropylamines: the effect of oxygen substituents at the beta-position. Bioorganic

and Medicinal Chemistry 12: 4055-4066.

Pharris A, Wiessing L, Sfetcu O, et al. (2011) Human immunodeficiency virus in injecting drug

users in Europe following a reported increase of cases in Greece and Romania, 2011.

Eurosurveillance 16: 48.

Ramoz L, Lodi S, Bhatt P, et al. (2012) Mephedrone ('bath salt') pharmacology: insights from

invertebrates. Neuroscience 208: 79-84.

Reuter P. (2011) Options for regulating new psychoactive drugs: a review of recent experiences.

Report, U.K. Drug Policy Commission, London, 15 May 2011.

5-20

Robinson JE, Agoglia AE, Fish EW, et al. (2012) Mephedrone (4-methylmethcathinone) and

intracranial self-stimulation in C57BL/6J mice: comparison to cocaine. Behavioural

Brain Research 234: 76-81.

Russo R, Marks N, Morris K, et al. (2012) Life-threatening necrotizing fasciitis due to ‘bath

salts’ injection. Orthopedics 35: e124-e127.

Sare J. (2011) How the media helped ban mephedrone. BMJ. Epub ahead of print, 24 Feb 2011.

doi: 10.1136/bmj.d1138.

Shortall SE, Green AR, Swift KM, et al. (2013) Differential effects of cathinone compounds and

MDMA on body temperature in the rat, and pharmacological characterisation of

mephedrone-induced hypothermia. British Journal of Pharmacology 168: 966-977.

Shulgin A. (1991) PIHKAL: A chemical love story. Berkely, California: Transform Press.

Simmler L, Buser T, Donzelli M, et al. (2012) Pharmacological characterization of designer

cathinones in vitro. British Journal of Pharmacology. Epub ahead of print. doi:

10.1111/j.1476-5381.2012.02145.x

Sindicich N & Burns L (2012). Ecstasy returns and the emerging class of drugs. EDRS Drug

Trends Bulletin, October 2012. Sydney: National Drug and Alcohol Research Centre,

University of New South Wales.

United Nations Office on Drugs and Crime. (2009) World drug report, 2009. United Nations:

Vienna.

Upreti VV, Eddington ND, Moon KH, et al. (2009) Drug interaction between ethanol and 3, 4-

methylenedioxymethamphetamine. Toxicology Letters 188: 167-172.

US Department of Justice Drug Enforcement Agency. (2011) Background, data and analysis of

synthetic cathinones: mephedrone (4-MMC), methylone (MDMC), and 3,4-

methylenedioxypyrovalerone (MDPV). Washington DC.: Office of Diversion Control,

Drug and Chemical Evaluation Section.

5-21

Van Hout MC and Bingham T. (2012) “A costly turn on”: patterns of use and perceived

consequences of mephedrone based head shop products amongst Irish injectors.

International Journal of Drug Policy 23(3): 188-197.

Van Nieuwenhuijzen P, McGregor I and Hunt G. (2009) The distribution of [gamma]-

hydroxybutyrate-induced Fos expression in rat brain: comparison with baclofen.

Neuroscience 158: 441-455.

Winstock AR and Ramsey JD. (2010) Legal highs and the challenges for policy makers.

Addiction 105: 1685-1687.

Winstock A and Wilkins C. (2011) ‘Legal highs’ The challenge of new psychoactive substances.

Series on Legislative Reform of Drug Policies, Number 16: Transnational Institute;

London: International Drug Policy Consortium.

Winstock A, Mitcheson L and Marsden J. (2010a) Mephedrone: still available and twice the

price. The Lancet 376: 1537.

Winstock AR, Marsden J and Mitcheson L. (2010b) What should be done about mephedrone?

British Medical Journal 340: c1605.

Wood DM, Measham F and Dargan PI. (2012) ‘Our favourite drug’: prevalence of use and

preference for mephedrone in the London night-time economy 1 year after control.

Journal of Substance Use 17(2): 91-97.

Wright MJ, Jr., Angrish D, Aarde SM, et al. (2012) Effect of ambient temperature on the

thermoregulatory and locomotor stimulant effects of 4-methylmethcathinone in Wistar

and Sprague-Dawley rats. PLoS ONE 7: e44652.

6-1

6. Appendix: Statements from Co-Authors

_______________________________

6-2

Callum Hicks B.Sc PhD Candidate, Psychopharmacology Laboratory

School Of Psychology, A18 , Australia, NSW 2006 Email:[email protected]

18 March 2013

To whom it may concern,

Craig was the primary author on the original manuscript entitled “Mephedrone in Adolescent Rats:

Residual Memory Impairment and Acute but Not Lasting 5-HT Depletion” published in the journal PLoS

ONE, 7(9): e45473. doi:10.1371/journal.pone.0045473.

I was a co-author on this manuscript and helped with the behavioural tests.

Yours Sincerely,

Callum Hicks (Co-author)

PhD Candidate, Psychopharmacology Laboratory

School of Psychology

The University of Sydney

6-3

6-4

I was co-author on one manuscript appearing in Addiction Biology (vol 17, 409-422). Craig was the

primary author, and I contributed to the Fos staining, counting and data analysis. .

Signed,

March 18, 2013.

Glenn Hunt, Discipline of psychiatry, University of Sydney

6-5

6-6

To Whom It May Concern,

This written statement is to attest to the fact that Craig P. Motbey was the primary author on the papers

listed below on which I am a co-author. My contribution to Paper 1 was performing the transcardial

perfusions of the rats, the extraction of their brains, and the preparation of the brains for slicing. My

contribution to Paper 2 was statistical analysis of the results for the self-administration data and writing

the related sections of the methods and results with the primary author.

Paper 1

Mephedrone (4-methylmethcathinone,‘meow’): acute behavioural effects and distribution of Fos

expression in adolescent rats

Craig P. Motbey, Glenn E. Hunt, Michael T. Bowen, Suzanne Artiss and Iain S. McGregor.

Addiction Biology, 17: 409-422

Paper 2

High Levels of Intravenous Mephedrone (4-Methylmethcathinone) Self-Administration In Rats: Neural

Consequences and Comparison with Methamphetamine

Craig P. Motbey, Kelly Clemens, Nadine Apetz, Adam R. Winstock, John Ramsey, Kong M. Li, Naomi

Wyatt, Paul D. Callaghan, Michael Bowen, Jennifer Cornish, Iain S. McGregor

Submitted to the Journal of Psychopharmacology

Kind regards,

Michael T. Bowen

6-7

6-8

To whom it may concern,

Craig Motbey was the primary author of: Mephedrone (4-methylmethcathinone,‘meow’): acutebehavioural effects and distribution of Fos expressionin adolescent rats. Craig P. Motbey, Glenn E. Hunt, Michael T. Bowen, Suzanne Artiss and Iain S. McGregor. I contributed to the acquisition of animal data and, along with the other co-authors, reviewed the content of the paper and approved final version for publication. Suzanne Artiss [email protected] 0433 615 178

6-9

Statement of Contribution

I, Nadine Apetz, herewith assure that Craig C. Motbey was the primary author of the article "High Levels

of Intravenous Mephedrone (4-Methylmethcathinone) Self-Administration In Rats: Neural

Consequences and Comparison with Methamphetamine".

My contribution to this project consisted of assistance in the conduction of the self-administration

experiments.

6-10

6-11

6-12

6-13

I, Dr Kong M. Li, of the Sydney University is to confirm that Craig Motbey was the primary

author and I was co-author for the publication of following manuscripts:

Mephedrone in Adolescent Rats: Residual Memory Impairment and Acute but Not Lasting 5-HT

Depletion

Craig P. Motbey, Emily Karanges, Kong M. Li, Shane Wilkinson, Adam R. Winstock, John

Ramsay, Callum Hicks, Michael D. Kendig, Naomi Wyatt, Paul D. Callaghan and Iain S.

McGregor.

PLoS ONE, 7(9): e45473. doi:10.1371/journal.pone.0045473

High Levels of Intravenous Mephedrone (4-Methylmethcathinone) Self-Administration In Rats:

Neural Consequences and Comparison with Methamphetamine

Craig P. Motbey, Kelly Clemens, Nadine Apetz, Adam R. Winstock, John Ramsey, Kong M. Li,

Naomi Wyatt, Paul D. Callaghan, Michael Bowen, Jennifer Cornish, Iain S. McGregor

Submitted to the Journal of Psychopharmacology

6-14

I, Emily Karanges, certify that Craig Motbey was the primary author on the publication entitled

“Mephedrone in Adolescent Rats: Residual Memory

Impairment and Acute but Not Lasting 5-HT Depletion” (PLoS ONE, 7(9): e45473.

doi:10.1371/journal.pone.0045473). I provided practical assistance with the running and analysis of the

HPLC data (neurotransmitter and metabolite analysis).

Emily Karanges

6-15

I was co-author on the manuscripts featured in Chapters 3 and 4. Craig Motbey was the primary author,

and I contributed to the provision and characterization of the test compound (mephedrone).

Signed March 27. 2013. Adam Winstock Kings College London

6-16

Iain S. McGregor MA (Oxon) PhD (Sydney)

Professor of Psychopharmacology

Tel +61 2 9351 3571 Fax +61 2 9351 8023

School Of Psychology, A18 , Australia, NSW 2006 Email: [email protected]

28 December 2013

I was the primary supervisor of Craig Motbey’s PhD thesis and the senior (last) author on the two

published papers and one submitted paper that appear in his thesis.

Craig was the first and primary author and performed most of the hands-on research and the writing of

the manuscripts.

I contributed towards the conception of the studies, experimental design, the forming of collaborative

networks, the writing of the manuscripts and the funding of the research via ARC, NHMRC and AINSE

grants.

Signed

1st April 2013

Professor Iain McGregor, School of Psychology, University of Sydney

6-17