acute, repeated-dose and residual effects of amphetamines

80
Acute, Repeated-Dose and Residual Effects of Amphetamines on Psychological Measures in Humans Allison Turza Bajger Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014

Upload: others

Post on 27-Nov-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Acute, Repeated-Dose and Residual Effects of Amphetamines

Acute, Repeated-Dose and Residual Effects of Amphetamines on Psychological Measures in

Humans

Allison Turza Bajger

Submitted in partial fulfillment of the

requirements for the degree of Doctor of Philosophy

in the Graduate School of Arts and Sciences

COLUMBIA UNIVERSITY

2014

Page 2: Acute, Repeated-Dose and Residual Effects of Amphetamines

© 2014 Allison Turza Bajger All rights reserved

Page 3: Acute, Repeated-Dose and Residual Effects of Amphetamines

ABSTRACT

Acute, Repeated-Dose and Residual Effects of Amphetamines on Psychological Measures in

Humans

Allison Turza Bajger

Despite the fact that the database documenting amphetamine-related effects in humans

has increased over the past decade, there remain important gaps in our knowledge of the effects

of these drugs in humans. The current investigations, which examined the acute, repeated-dose

and residual effects of amphetamine derivatives on various psychological measures in humans,

addressed two of these gaps. The first was the lack of empirical evidence directly comparing d-

amphetamine and methamphetamine. Study 1 was the first direct comparison of the regulatory

focus effects of intranasal d-amphetamine and methamphetamine (12, 50 mg/70kg). Results

indicate that the drugs produced overlapping effects on most measures (e.g. increased

“prevention” focus state and task engagement). Under the low dose condition, only

methamphetamine increased “prevention” focus. Study 2 was a within-participant investigation

on the impact of three 3,4-methylenedioxymethamphetamine (MDMA) administrations (12 and

24 hours intervals) on physiological, subjective, and behavioral measures in experienced MDMA

users. Heart rate, blood pressure, oral temperature, subjective effects, psychomotor performance,

and sleep were assessed repeatedly throughout the study. Acute administration of MDMA

produced systematic increases in heart rate, blood pressure, and subjective effects, but oral

temperature was unaltered. Following repeated drug administration, heart rate elevations were no

longer statistically significant; blood pressure and subjective-effect ratings remained

significantly increased, but such increases were diminished relative to acute drug effects.

Measures of sleep were decreased only on the evening following two active MDMA

Page 4: Acute, Repeated-Dose and Residual Effects of Amphetamines

administrations. Performance alterations were not observed nor were MDMA-related toxic

effects. Overall, the data from both studies do not support either: 1) the conventional notion that

d-amphetamine and methamphetamine produce markedly different effects in humans; or 2) the

general perception that MDMA produces dangerous cardiovascular and subjective effects in

humans following repeated administration.

Page 5: Acute, Repeated-Dose and Residual Effects of Amphetamines

i

TABLE OF CONTENTS List of Figures iv List of Tables v Chapter 1: General Introduction 1

1.1 History of Amphetamines in the United States 1 1.1.1 d-amphetamine and methamphetamine 1

1.1.2 3,4-methylenedioxymethamphetamine 15

1.2 Concluding thoughts 17 Chapter 2: The Effects of Intranasal d-Amphetamine and Methamphetamine on 18

Regulatory Focus Measures in Humans

2.1 Introduction 18 2.2 Methods and Materials 22

2.2.1 Participants 22

2.2.2 Pre-study Training 23

2.2.3 Design 23

2.2.4 Procedure 24

2.2.5 Psychomotor Task 24

2.2.6 Regulatory Focus Questionnaire (RFQ) 24

2.2.7 State Regulatory Focus Questionnaire 25

2.2.8 Task Engagement Questionnaire 25

2.2.9 Drug 26

2.2.10 Data Analysis 26

Page 6: Acute, Repeated-Dose and Residual Effects of Amphetamines

ii

2.3 Results 26

2.3.1 Regulatory Focus (RFQ) Effects 26

2.3.2 State Regulatory Focus Effects 27

2.3.3 Task engagement Effects 27

2.4 Discussion 27 Chapter 3: Acute, Repeated-dose and Residual Effects of 3,4- 31

methylenedioxymethamphetamine (MDMA) in Humans

3.1 Introduction 31 3.2 Methods and Materials 34

3.2.1 Participants 34

3.2.2 Pre-study Training 35

3.2.3 Design 35

3.2.4 Procedure 36

3.2.5 Subjective-Effects and Psychomotor Battery 37

3.2.6 Sleep Monitoring 38

3.2.7 Drug 38

3.2.8 Data Analysis 38

3.3 Results 39 3.3.1 Cardiovascular and Oral Temperature Effects 39

3.3.2 Subjective Effects 40

3.3.3 Psychomotor Performance Effects 41

3.3.4 Sleep Effects 42

3.3.5 Effects on Cigarette Smoking 42

Page 7: Acute, Repeated-Dose and Residual Effects of Amphetamines

iii

3.3.6 Effects on Social Interactions 42

3.4 Discussion 43 Chapter 4: General Discussion 47

4.1 d-amphetamine and methamphetamine 47

4.2 3,4-methylenedioxymethamphetamine 48

4.3 Concluding thoughts 49 References 51 Appendices 60 Appendix A 60

Appendix B 65

Appendix C 67

Page 8: Acute, Repeated-Dose and Residual Effects of Amphetamines

iv

LIST OF FIGURES

Figure 1.1 The effects of oral and subcutaneous amphetamine on 61 cardiovascular measures in humans Figure 1.2 Over-the-counter Benzedrine Inhalers 62 Figure 1.3 Smith, Klein and French advertisement for Benzedrine Sulfate 62 (e.g., amphetamine) Figure 1.4 Smith, Klein and French advertisements for use of Benzedrine 63 inhalers in the military Figure 1.5 Burroughs Wellcome & Co. advertisement for Methedrine 63 Figure 1.6 Chemical structure of dextro- and levo-amphetamine 64 Figure 1.7 Burroughs Wellcome & Co. advertisement for Methedrine 64 ampoules Figure 2.1 “Prevention” focus pride ratings as a function of drug condition 65 Figure 2.2 “Prevention” focus state ratings as a function of drug condition 66 Figure 2.3 Engagement ratings on selected items as a function of drug condition 66 Figure 3.1 Acute (Administration 1) cardiovascular and subjective effects as a 69 function of drug dose and time Figure 3.2 Repeated-dose cardiovascular and subjective-effects effects as a 69 function of drug condition and administration Figure 3.3 Sleep effects as a function of drug dose and day 69

Page 9: Acute, Repeated-Dose and Residual Effects of Amphetamines

v

LIST OF TABLES Table 1.1 Early medical uses of amphetamines 60 Table 1.2 Summary of controlled substance schedules 61 Table 2.1 Study design 65 Table 2.2 d-amphetamine- and methamphetamine-related effects 65 on task engagement ratings Table 3.1 Study design 67 Table 3.2 Acute (Administration 1) MDMA-related effects on 67 subjective-effect ratings Table 3.3 MDMA-related effects on physiological and subjective-effect 68 ratings (Administration 3) Table 3.4 Repeated-dose effects on subjective ratings 68

Page 10: Acute, Repeated-Dose and Residual Effects of Amphetamines

vi

ACKNOWLEDGMENTS

I would like to thank all of the remarkable members of my committee (Carl Hart, Tory

Higgins, Walter Mischel, Rae Silver, and Elias Dakwar) for their invaluable insights on my

research. I owe a very special thanks to my longtime mentor and friend, Carl Hart. Thank you

for always pushing me to be a better thinker, writer and teacher. I am indebted to my other

extraordinary advisor, Tory Higgins; thank you for always believing in me and teaching me to

love and respect social psychological principles and theory. I would be remiss if I did not thank

my fellow labmates who have kept me sane over the years: Matt Kirkpatrick, Steen Sehnert,

James Cornwell, Christine Webb, Travis Riddle and Kirsten Frazer. I would also like to thank

my family for their continual support. Finally, as Robert Burns once famously wrote, “the best-

laid plans of mice and men often go awry.” It is an understatement to acknowledge that some

unexpected curveballs have been thrown my way in recent years, and I would like to send a very

inadequate thank you to my incredible husband, Dan Bajger: your patient ears and steady,

insightful eyes have gotten me through some of the most difficult moments of my life. Thank

you for always making me see the silver lining.

Page 11: Acute, Repeated-Dose and Residual Effects of Amphetamines

vii

DEDICATION

In honor of my mother, Dale Turza, whose strength and confidence I strive to achieve everyday.

For my father, Peter Turza, who inspires me to continually ask questions and learn more.

Page 12: Acute, Repeated-Dose and Residual Effects of Amphetamines

1

Chapter 1: General Introduction

1.1 History of Amphetamines in the United States 1.1.1 d-amphetamine and methamphetamine Synthesis of amphetamine

Although certain amphetamines (e.g., d-amphetamine, methamphetamine and 3,4-

methylenedioxymethamphetamine (MDMA)) have attracted popular attention only over the past

two decades, the use of these compounds dates back to the early 1900s. Of these compounds, d-

amphetamine was the first synthesized in 1887 by German chemist, Lazar Edeleano (Anglin et

al., 2000). Before amphetamine could be produced an important scientific development had to

occur: the synthesis of ephedrine (the active ingredient in the herb ma huang, Ephedra sinica

plant) in 1885 by Japanese chemist, G. Yamanashi (Chen & Schmidt, 1926) and the re-synthesis

of the drug by Ku Kuei Chen and Carl Schmidt in 1923. Before amphetamines were used in

medicine, ephedrine was used (e.g., as a nasal decongestant).

This early research laid the groundwork for the use of the drug in medicine. Eli Lilly and

Company launched the first large-scale production of ephedrine in 1926: the drug dominated the

pharmaceutical market as a treatment for 1) anaphylactic shock in patients with cardiac

disorders, 2) chronic hypotension (i.e., low blood pressure), a classic symptom of Addison’s

disease, 3) congestion for the common cold, and 4) asthma.

The success of ephedrine as a therapeutic in relieving symptoms caused by clinical illnesses

increased the public’s confidence in the use of medications. By the late 1920s, ephedrine supply

was low and prices were high due to the reliance of Eli Lilly and Company on overseas

production. This situation helped to stimulate scientific investigations in search of alternative

medications for conditions which ephedrine had been used. In 1929, the American biochemist,

Page 13: Acute, Repeated-Dose and Residual Effects of Amphetamines

2

Gordon Alles, synthesized several compounds that were structurally similar to ephedrine,

including phenylisopropylamine (e.g., later named “amphetamine”): amphetamine is the basic

ephedrine structure without the side chain hydroxyl group. As a graduate student, Alles worked

with John Jay Abel, a well-known pharmacologist who had isolated the pancreatic hormone,

insulin, in 1926 (Parascandola, 2010).

Early scientific research on amphetamine

In 1930, Alles and colleagues published the first study of amphetamine comparing its

cardiovascular effects with related compounds in dogs. The researchers found that a single

intravenous administration of amphetamine (50 mg) produced increases in blood pressure that

persisted longer than other compounds. It is important to note that only one dose level of

amphetamine and related compounds was assessed and as a result, the dose-response curve is

unclear: it is possible that the related compounds could have produced more robust effects than

amphetamine at different doses.

To enhance the translational relevance of the above study, Alles et al. (1930) conducted a

subsequent study during which oral and subcutaneous doses (50 mg, 1 cc of 5% solution) of

amphetamine and related compounds were compared in humans. They observed that both oral

and subcutaneous amphetamine produced marked increases in blood pressure and decreases in

mouth and nose secretions. These effects lasted 8 hours. The comparator compounds failed to

produce effects on any measure. The researchers indicated amphetamine as an effective

alternative medication to ephedrine for a wide range of clinical conditions (i.e., congestion from

the common cold, asthma, hypotension).

Being cognizant of pharmacokinetics, the researchers were also interested in comparing the

effects of amphetamine after different routes of administration. They found that oral and

Page 14: Acute, Repeated-Dose and Residual Effects of Amphetamines

3

subcutaneous administration of the drug produced identical effects. However, the rate of onset

of these effects was different between the two routes. When the drugs were taken orally, peak

effects occurred about two and a half hours after ingestion. In contrast, subcutaneous peak

effects occurred approximately one and a half hours after administration, one hour before the

onset of effects following oral dosing (Figure 1.1). These findings were important because they

led to the manufacturing of the drug in two forms (e.g., pill, injection), each providing patients

with different advantages (i.e. convenience vs. rapid effects, respectively).

In 1932, Alles gained sole patent rights to both amphetamine sulfate (pill) and amphetamine

hydrochloride (injectable solution). Shortly after, in 1933, Smith, Kline and French chemist,

Fred Nabenhauer, developed and patented a slightly different preparation of the volatile free base

form of amphetamine (e.g., a smokable form of the drug). This drug delivery method expanded

the variety of clinical uses of amphetamine (i.e., as a nasal decongestant). That same year, Smith,

Kline and French marketed the drug as an over-the-counter remedy for congestion and asthma

(e.g., “Benzedrine” Inhaler) (Jackson, 1971) (Figure 1.2). After negotiations with Alles, the

pharmaceutical company also patented and manufactured “Benzedrine Sulfate,” the pill form of

the Benzedrine inhaler, in 1934. Amphetamine became one of most versatile medications

available on the market at this time.

In order to best promote their new product directly to doctors, Smith, Kline and French

placed advertisements in top medical journals (e.g., Journal of the American Medical

Association, JAMA). However, before gaining access to commercial space in medical journals,

pharmaceutical companies were first required to receive the “Seal of Approval” of the American

Medical Association’s (AMA) Council on Pharmacy and Chemistry. Under this policy,

manufacturers were required to provide scientific evidence that a drug “present[ed] some real

Page 15: Acute, Repeated-Dose and Residual Effects of Amphetamines

4

advantage” (Kremers & Sonnedecker, 1976). This early requirement spurred a tremendous

amount of research on amphetamine, and the findings and interpretations gave the impression

that amphetamine was a panacea.

In 1935, Smith, Kline and French funded several clinical investigations of amphetamine

(Rasmussen, 2006, 2008). The first of these studies was a clinical trial comparing the

effectiveness of amphetamine and ephedrine to treat narcolepsy (Prinzmetal and Bloomberg,

1935). The researchers found that oral amphetamine (10-90 mg) reduced unexpected, abrupt

onset of sleep (e.g., a hallmark symptom of narcolepsy) in all patients. This therapeutic effect

was shown to last for up to 14 months, suggesting a lack of tolerance to amphetamine-related

effects. Prinzmetal and Bloomberg also found that ephedrine improved alertness in only 30% of

patients and this effect diminished after one month, leading them to conclude that amphetamine

was more effective than ephedrine as a narcoleptic medication. It’s important to note that this

study was not double-blind or placebo-controlled, but at the time it was rigorous enough to be

published in the top medical journal of this era, JAMA.

Over the next few years, findings from several other clinical studies on amphetamine-related

effects were published in scientific literature, increasing the avenues for which amphetamines

could be prescribed. Several investigators assessed the ability of amphetamine to enhance mood

in depressed individuals (Myerson, 1936; Guttman, 1936; Guttman & Sargant, 1937; Bloomberg,

1939). In a seminal, open-label study, Wilbur et al. (1937) administered low oral doses (2.5 - 20

mg) of amphetamine once or twice daily for up to 6 months and found that amphetamine

markedly enhanced mood in patients who suffered from mild to moderate depression, but not in

patients diagnosed with severe depression. Amphetamine effectiveness appeared to diminish

overtime, suggesting a possible development of tolerance to its mood-enhancing effects. These

Page 16: Acute, Repeated-Dose and Residual Effects of Amphetamines

5

findings are intriguing for multiple reasons. First, data from a recent study indicate a different

pattern of effects: the benefit of antidepressant medications (all of which are FDA-approved and

non-amphetamine-based) increases with the severity of depressive symptoms. That is, patients

with severe depression benefit, while those with mild or moderate symptoms do not (Fournier et

al. 2010). Second, this early research contributed to current conventional wisdom that

amphetamine loses its effectiveness as an antidepressant therapeutic largely. It is unclear whether

amphetamine-related effects on depressive symptoms diminished in the majority of patients in

that study. There are some psychiatrists who continue to use low doses of amphetamine (5-20

mg/d) as an adjunctive treatment for depression, including members of the Depression

Evaluation Services at the New York State Psychiatric Institute (communication with Dr. Jon

Stewart).

Another illness for which amphetamine was thought to be a potential clinical intervention

was postencephalitic Parkinsonism. Parkinson’s disease is characterized by sleep and movement

disruptions (i.e., REM sleep disturbances, daytime drowsiness, muscular rigidity). Davis and

Stewart (1938) investigated the effects of oral amphetamine (ranging from 40 – 60 mg, given in

divided doses) using an open-label design. The researchers found that amphetamine reduced

symptoms of fatigue, muscular rigidity, and tremors in 72% of the patients. These findings were

interpreted as extremely promising because the only other treatments were for Parkinsonism

were choline-based drugs such as atropine and scopolamine, which appeared to be considerably

less effective than amphetamine.

Together, above data provided the necessary evidence for Smith, Kline and French to receive

approval by the AMA Council to advertise “Benzedrine Sulfate” tablets for the treatment of

narcolepsy, minor depression, and postencephalitic Parkinsonism (AMA Council on Pharmacy

Page 17: Acute, Repeated-Dose and Residual Effects of Amphetamines

6

and Chemistry, 1937) (Figure 1.3). Amphetamine gained an increasing reputation as a panacea.

This is consistent with the notion that began in the early 1900s that drugs were viewed as

pharmacological tools to solve several health problems. Like insulin and penicillin, amphetamine

was a newly minted member of a generation of breakthrough medications.

Amphetamines and the military (World War II [1939-1945])

During World War II, the German military introduced a novel type of military warfare

consisting of speed and surprise tactics (i.e., “Blitzkrieg,” “lightning war”), which often resulted

in overworked, sleep-deprived soldiers. German forces utilized drugs to solve such military

problems. In 1939, a new amphetamine-based drug, “Pervitin” (later named methamphetamine;

in 3 mg tablets), was distributed to soldiers to offset battle fatigue, and enhance performance

during demanding ground and air missions. The drug closely resembles amphetamine in

chemical structure and profile of effects: methamphetamine is the amphetamine structure with

the addition of a methyl group and produces nearly identical effects as amphetamine (i.e.,

increases heart rate, blood pressure, alertness, euphoria) (Martin et al., 1971; Sevak et al., 2009;

Kirkpatrick et al., 2011). It appears that the German military primarily used methamphetamine

because of manufacturing limitations: amphetamine (Benzedrine) was predominantly

manufactured in the U.S.

Although methamphetamine was originally synthesized by Japanese scientist, Nagayoshi

Nagai, in 1893 (and later synthesized by Akira Ogata, in 1919), it was not until 1938 that the

Berlin-based Temmler pharmaceutical company marketed the drug to German doctors, namely

military physicians (Grobler et al., 2011; Weisheit & White, 2009). Estimates indicate that

approximately 35 million methamphetamine tablets were used by German forces between April

and June of 1940 at the peak of the Blitzkrieg (Garber, 2013).

Page 18: Acute, Repeated-Dose and Residual Effects of Amphetamines

7

Following their major defeat in 1940, British allied forces recognized the beneficial effects of

amphetamines in war. British media coverage was filled with accounts of “heavily drugged,

fearless and berserk” German paratroopers who were largely credited at the time for the success

of the 1940 mission (Rasmussen, 2008). Shortly after their defeat, the Royal Air Force (RAF)

commissioned experiments investigating the effects of various stimulants (i.e., amphetamine,

methamphetamine and caffeine) on performance and mood in soldiers. In a series of field studies

on long-range Coastal Command missions between 1941 and 1942, Winfield, a British medical

officer, administered low, oral doses (up to 10 mg) of methamphetamine (e.g., “Methedrine”)

and amphetamine (e.g., “Benzedrine”) to troops. The researcher concluded that amphetamine

and methamphetamine produced overlapping effects: both drugs increased alertness. Because

amphetamine was reportedly more effective at enhancing mood than methamphetamine,

Winfield ultimately only recommended the use of amphetamine (5 mg) for long bomber flights.

It is important to note that this study lacked double-blind conditions, allowing for the possibility

of expectancy effects. Nevertheless, the Royal Air Force heeded Winfield’s suggestion in 1942

(Rasmussen, 2008).

Aware of British research, the U.S. National Research Council (NRC) funded scientific

amphetamine-related investigations in an effort to advise the U.S. Air Force on the possible

benefits of pharmacological tools in war. In the first of a series of decompression-chamber

studies (e.g., under conditions of high altitude), Ivy and colleagues (1942) administered caffeine,

amphetamine and methamphetamine alone and the combinations, caffeine and amphetamine, and

amphetamine and methamphetamine, in normal and low pressure (e.g., altitudes up to 18,000

feet) conditions. Again, the researchers found that amphetamine and methamphetamine produced

similar effects: the drugs both alone and in combination significantly improved psychomotor

Page 19: Acute, Repeated-Dose and Residual Effects of Amphetamines

8

performance compared to placebo (as cited in Rasmussen, 2011).

Alongside this military research, the Illinois-based pharmaceutical company, Abbott

Laboratories, funded studies investigating performance effects of amphetamines between 1941

and 1942. In one of these experiments, Ivy and colleagues administered intravenous caffeine

(unknown dose), amphetamine (10-15 mg), methamphetamine (5 mg) and placebo to humans

and found that caffeine and methamphetamine improved performance on strenuous physical

fitness tests compared to amphetamine (Rasmussen, 2008). These amphetamine-related effects

are consistent with the fatigue-reducing effects of amphetamine in narcoleptic patients. It is

important to note, however, that the study included a design that varied both the dose and drug.

It is unclear whether the drugs would produce similar effects at the same dose. Regardless, in

1942, Abbott expanded their product line and began marketing methamphetamine (“Desoxyn”)

to physicians and the U.S. military (Weisheit & White, 2009).

Despite empirical evidence indicating similarities between the two drugs, Ivy and colleagues

ultimately only recommended amphetamine (Benzedrine in 5 mg tablets) for use by the U.S. Air

Force in 1943 (Figure 1.4). A major motivation for this decision was that amphetamine was

viewed as a panacea for a number of ailments (i.e., fatigue, depression), some of which soldiers

suffered. Military psychiatrists and psychologists were increasingly concerned with U.S. troop

“morale”: “shell shock” and “psychoneurotic breakdown” were thought to be on the rise in U.S.

servicemen (Rasmussen, 2011). Amphetamine was the only approved antidepressant at the time

and researchers argued that, unlike methamphetamine, amphetamine increased performance

primarily by enhancing mood. In retrospect, this statement seems arbitrary in light of the fact

that no empirical evidence existed to support this claim.

Page 20: Acute, Repeated-Dose and Residual Effects of Amphetamines

9

Civilian use after World War II (1940-1950s)

After the War in 1945, the medical use of amphetamine was expanded. One new use was the

treatment of “hyperkinetic” behavior in children (later known as Hyper-Attention Deficit

Disorder (ADHD)). In one of the first studies to show that the drug produced beneficial effects

in this population, Bradley and Bowen (1940) examined the effects of single and repeated oral

doses of amphetamine (10, 20, 30, and 40 mg) on the behavior of hyperkinetic children aged 5-

12 years. They found that amphetamine enhanced attention and academic performance in more

than 50 percent of the sample studied. Around the same time, other researchers showed that the

drug was an effective appetite suppressant (Rosenthal, 1940; Kunstadter, 1940). By 1949, SKA

had received approval from the AMA Council to market amphetamine as a treatment for

depression, hyperkinetic behavior, and weight loss. As a result, SKA sales increased from $5.7

million in 1947 to $7.3 million in 1949 (AMA, 1947; Jackson, 1976).

Benzedrine advertisements increasingly included statements indicating the multiple

therapeutic uses of amphetamine (i.e., for depression, unwanted weight gain): housewives were

the main target of such ads because they were thought to suffer from a number of clinical

conditions due to the “bor[dem] with the monotony of their unexciting lives” (Figure 1.5)

(Rasmussen, 2006; Bett, 1946). As can be seen in Table 1.1, by 1946, amphetamine was

indicated for over 30 medical conditions (Bett, 1946) and was so versatile that it was even used

as an analgesic (pain-reliever) in the White House by John F. Kennedy for his hurt back

(Rasmussen, 2008).

Amphetamines were also widely used for purposes other than medical. More people became

aware of the multiple uses of amphetamines from scientific reports (Heal et al., 2013) and,

unhindered by strict regulations, easily accessed the drug (inhaler) over-the-counter at

Page 21: Acute, Repeated-Dose and Residual Effects of Amphetamines

10

pharmacies until 1959 (Anglin et al., 2000; Guttman & Sargent, 1937). Truckers and students

used amphetamines to offset sleep and extend hours beyond their normal sleep period (Tidy,

1938; Heal et al., 2013). Professional athletes, especially cyclists and baseball players, used the

drugs to enhance alertness and counteract psychological decrements caused by fatigue.

Amphetamines remain the drugs of choice in baseball, however, in 2006, they were finally

banned and a prescription is now needed. Furthermore, housewives and “Bohemian” groups

(e.g., jazz, bebop, and beat subcultures) used the drugs to enhance their mood and sociability

(Weisheit and White, 2009).

Media Reports

Over the course of only a few years, amphetamines shifted from panaceas to dangerous

substances. The many personal uses of amphetamines mirrored the variety of labels popular

reports ascribed to the drugs in the 1940s and 1950s (e.g., “Pep-Pills,” “Confidence Drug[s],”

and “New York’s Benzedrine Set”) (Jackson, 1971; Bett, 1946). However, overtime, media

coverage became increasingly filled with accounts of “supercharged” students cramming for

exams and collapsing from substance use (Time, 1942, Science Newsletter, 1937). Several

articles focused on the “harmful aftereffects” of the drugs on students’ “poisonous brain” (Time,

1937, 1942). Media outlets initiated rumors over amphetamine-induced neurosis, even though

the drugs were actually indicated to treat psychotic episodes (W.L.L, New York Times, 1947). In

an article published in Time magazine, Benzedrine and Pervitin were stigmatized as “Nazi Pep-

Pills.”

Scientific Reports: Long-term Effects of Amphetamines

In addition to negative media reports, there were statements in the scientific literature

attesting to the multiple deleterious medical consequences of amphetamine abuse. Commonly

Page 22: Acute, Repeated-Dose and Residual Effects of Amphetamines

11

described unfavorable effects of long-term use of large amphetamine doses were hallucinations

and paranoia (i.e., termed “amphetamine psychosis”) (Norman & Shea, 1945; Goodman &

Gillman, 1941; Young & Scoville, 1938). Such statements, however, were based on data from

case studies conducted on an unrepresentative sample of the population (e.g., some participants

had a history of depression, alcoholism etc.). Other frequently reported deleterious effects

associated with amphetamine abuse were malnutrition from appetite loss, xerostomia (dry

mouth) and “burning of the gums” (Monroe and Drell, 1947). But, it is important to consider the

fact that these claims were based on behavioral observations from a field study and it is unclear

if these phenomena had more to do with non-pharmacological factors, such as poor dental

hygiene and nutrition, than pharmacological effects of amphetamines.

At the time, data from scientific studies indicating no deleterious effects of long-term

amphetamine use were largely ignored. For example, Bakst (1944) conducted a case study on a

user who had administered low to moderate doses (15-30 mg) of amphetamine daily for about 9

years. No untoward dental or psychological effects of the drug were observed. Although this

study shared similar limitations as aforementioned studies (e.g., lacked controlled, double-blind

conditions), it provided important information to the growing database documenting the long-

term effects of amphetamine use in humans.

Despite these mixed findings, members of the medical community became increasingly

concerned about the unfavorable effects associated with amphetamine use. JAMA headlined one

article, “Benzedrine Sulfate—A Warning,” describing the toxic and addictive effects of long-

term use of the drug, urging doctors to limit prescriptions for medical conditions. By 1947, new

regulations made amphetamine tablets (Benzedrine Sulfate) available by prescription only in

several states and required manufacturers to include a label cautioning that this “dangerous drug”

Page 23: Acute, Repeated-Dose and Residual Effects of Amphetamines

12

should only be used under the close supervision of a physician (Jackson, 1971; Anglin, 2000;

Monroe & Drell, 1947).

Pharmaceutical Companies’ Response to Negative Reports

Following warnings of the unpredictable and addictive effects of their product,

Benzedrine Sulfate, Smith, Kline and French commissioned several studies investigating the

safety and efficacy of a potential alternative amphetamine-based drug, dextro-amphetamine (e.g.,

“Dexedrine”). The amphetamine molecule exists in two, asymmetric forms known as optical

isomers (e.g., left-handed [levo-amphetamine] and right-handed [dextro-amphetamine] versions)

(Figure 1.6) and in 1938, company chemists discovered a method of separating the two isomers,

allowing for comparison studies of the effects of all three types of amphetamine.

Alles, the chemist who previously synthesized and patented Benzedrine (e.g., a racemic

mixture of equal parts dextro- and levo-amphetamine), and Prinzmetal, the first scientist to study

the effects of amphetamine in narcoleptic patients, conducted a series of comparison studies on

the effects of all three compounds. In the one of these studies, Alles and Prinzmetal (1940)

administered oral doses of dextro-amphetamine (10-28 mg), levo-amphetamine (20-50 mg), and

racemic amphetamine (Benzedrine; 20-50 mg) to patients diagnosed with narcolepsy or

postencephalitic Parkinson’s disease. They found that, in comparison to levo-amphetamine and

racemic amphetamine, dextro-amphetamine produced greater decreases in 1) unexpected, abrupt

onset of sleep in all narcoleptic patients, and 2) fatigue, muscular rigidity and tremors in 50% of

patients diagnosed with postencephalitic Parkinson’s disease. Furthermore, no negative effects

of dextro-amphetamine were observed. It is important to note, however, that researchers

compared different doses of each drug, making it difficult to decipher the real differences

between the two isomers. Despite this study limitation, in 1939, Smith, Kline and French

Page 24: Acute, Repeated-Dose and Residual Effects of Amphetamines

13

patented and marketed dextro-amphetamine (“Dexedrine”) as a safe and effective substitute

medication for the treatment of narcolepsy and postencephalitic Parkinson’s disease.

Amphetamine “Epidemic” in 1950s-1970s

By the mid 1950s, national concerns about recreational amphetamine abuse were on the

rise. The New York Times headlined one story, “Ours is the Addicted Society” (Farber, 1966).

One factor that contributed to this increased worry about “habit-forming” drugs was that

intravenous (IV) administration, which is associated with more intense, potentially more

deleterious drug-related effects, was becoming popular in drug subcultures (e.g., heroin users).

When a drug is given by injection, it directly enters the bloodstream without going through the

stomach, intestines, and liver. In comparison to oral administration, IV administration produces

a more rapid onset of effects because drugs access the brain faster. This route is associated with

a higher abuse potential in humans primarily because some researchers have argued that the

more immediate drug-effects allow the drug to act as a more robust reinforcer. This route of

administration is also stigmatized because it is the preferred route of heroin users, who are

commonly viewed as being “more committed” drug abusers.

In the early 1960s, heroin and cocaine were often administered together using the

intravenous route of administration (e.g., speedball) and IV users soon realized that similar drug

effects could be produced by combining heroin and amphetamine. Amphetamine gained the

nickname, “speed” (most likely due to the rapid “high” produced by IV administration).

Recreational users would extract the medicated wick inside the Benzedrine Inhaler, soak it in

water or coffee, and inject the remaining solution (Jackson, 1975). An injectable form of

methamphetamine was also available by prescription for the treatment of heroin addiction

(Anglin et al., 2000; Weisheit & White, 2009) (Figure 1.7).

Page 25: Acute, Repeated-Dose and Residual Effects of Amphetamines

14

Stories on this new, potentially more deleterious method of administering amphetamines

started to appear in the national press. For example, New York Times articles commented that

this form of amphetamine “induces a strong, fast high” and an “intense feeling of power”

(Kifner, 1967). Popular press articles describing street corners of “speed freaks” were common

(Golden, 1967), and by 1967, public service announcements from an anti-amphetamine

campaign, whose slogan was “Speed Kills,” were familiar messages on radio stations

(Rasmussen, 2008). Warnings, such as “Methedrine Use is Growing,” also headlined articles

from prominent newspapers (Kifner, 1967, Severo, 1971). These statements in the popular press

increased public concern about a supposed amphetamine “epidemic” (Robinson, 1967; Black,

1970).

In 1970, growing public concern, in turn, motivated policy-makers to pass new legislation:

Congress enacted the Comprehensive Drug Abuse Prevention and Control Act which created

five “schedules” of controlled substances as a system to balance the potential medical uses of a

substance with its abuse potential (Miller and Hughes, 1994). Drugs are classified into one of

five of these categories depending on the accepted medical use and abuse potential of the drug

(Table 1.2). For example, Schedule I drugs (e.g., Heroin, Marijuana, LSD) are considered to

have no acceptable medical use and a high potential for abuse. Drugs in this category are

considered to have no acceptable medical use in treatment in the U.S., lack safety for use, have a

high potential for abuse, and are not available by prescription. Drugs classified in Schedules II-V

(e.g., morphine, cocaine, oxycodone) are considered to have an accepted medical use and a low

to moderate potential for abuse, and are available by prescription.

Under this policy, amphetamine and methamphetamine were placed under Schedule II. Strict

record-keeping regulations and prescription refill rules limited manufacturing of the two drugs

Page 26: Acute, Repeated-Dose and Residual Effects of Amphetamines

15

and recreational use of these drugs was made illegal in the U.S. (Miller and Hughes, 1994).

Pharmaceutical amphetamines companies (e.g., Abbott and Burroughs Wellcome) withdrew their

intravenous amphetamine products from the prescription marketplace, giving rise to synthesis of

illicit recreational methamphetamine in clandestine underground laboratories (“meth labs”)

(Miller, 1996) (Anglin et al., 2000).

1.1.2 3,4-methylenedioxymethamphetamine

3,4-methylenedioxymethamphetamine (MDMA) is similar in structure to amphetamine and

methamphetamine: the drug is the basic methamphetamine with an additional methyldioxy ring.

Recent research findings indicate that it produces a similar profile of effects as the two other

stimulants (i.e., increased heart rate, stimulation, and euphoria) (Kirkpatrick et al., 2011).

Although MDMA was initially discovered and patented in 1912 by Merck Scientists as an

intermediate compound in the chemical synthesis of methylhydrastinine (e.g., a medication used

to reduce bleeding), the drug received little further attention until 1953 (Bernschneider-Reif and

Freudenmann, 2006). From 1953 to 1964, the Scientific Intelligence Division of the Central

Intelligence Agency (CIA) in collaboration with the Special Operations Division of the Army’s

Chemical Corps launched the classified MK-ULTRA experimental program which consisted of a

series of studies investigating different methods of “manipulating human behavior” (Michale-

Robinet, 1994). MDMA was included as one of several psychoactive drugs (i.e., LSD,

psilocybin, heroin) tested as potential tools for espionage (e.g., “brainwashing,” polygraph

testing) in these confidential studies. The CIA primarily focused on MDMA most likely because

of two effects that were thought to be divergent from amphetamine and methamphetamine:

MDMA was considered to have various hallucinogenic (i.e., changes in perception of one’s own

Page 27: Acute, Repeated-Dose and Residual Effects of Amphetamines

16

body, alterations of auditory and visual input) and empathogenic properties (i.e., increased

openness and communication, “truth serum”).

In general, Army researchers recruited individuals from U.S. universities, prisons and

hospitals and surreptitiously administered these drugs to subjects. Although some reports from

the project were declassified in 1994, details regarding the results of these experiments are

limited. The program was eventually curtailed in the early 1960s following an accidental death

of a subject due to overdose and was officially terminated in 1973 (Karch, 2011). It is important

to note, however, that previous research found no evidence of MDMA-related cardiovascular

toxicity in animals (e.g., rats, mice, monkeys) (Hardman et al., 1973).

Around the time of the program conclusion, Alexander Shulgin, a U.S. chemist, was growing

increasingly interested in the effects of mescaline analogs (e.g., MDA, MMDA) and in 1965, re-

synthesized MDMA (Karch, 2011). In the first published article on the effects of MDMA in

humans, Shulgin and Nichols (1978) reported that moderate to high, oral doses (75-150 mg) of

MDMA produced perceptual alterations (e.g., “an altered state of consciousness with emotional

and sensual overtones”) in humans. The researchers also noted that the drug produced limited

physiological residual effects.

Despite the fact that there was minimal evidence on the psychological effects of the drug in

humans, Shulgin reportedly suggested the use of MDMA as a therapeutic aide to the psychiatric

community. Due to its purported ability to increase openness and empathy in humans, the drug

became popular among medical professionals (e.g., psychiatrists, psychotherapists) as well as the

general public (Benzenhofer & Passie, 2010). A distribution group in Texas (e.g., “The Texas

Group”) made the drug readily accessible: it has been reported that users could buy it with a

Page 28: Acute, Repeated-Dose and Residual Effects of Amphetamines

17

credit card over the phone by calling a toll-free number and at nightclubs in Dallas and Fort

Worth, Texas (Holland, 2001).

Texas Senator and Member of the Senate Judiciary Committee, Lloyd Bentsen, grew

concerned over this new recreational use pattern of MDMA in his state and in 1984, submitted a

request to the Drug Enforcement Administration (DEA) to “emergency” schedule MDMA.

Under this policy, the Justice Department was permitted to immediately place a drug on a

Schedule without a congressional hearing and in 1985, MDMA was placed on Schedule I, where

it remains today.

2.1 Concluding Thoughts

Despite the tremendous amount of early research indicating that amphetamines can be

used safely within a therapeutic dose range, sensationalized media portrayals of the drugs

trumped scientific findings. Accordingly, public concerns and policies related to amphetamines

were ultimately shaped by anecdotal reports. There was, however, no real evidence supporting

popular claims about amphetamine-associated toxicity. Despite the growing scientific database

documenting the effects of amphetamines in humans, there are still considerable gaps in our

knowledge. The following studies were conducted to address these gaps and dispel some of the

myths about amphetamines that still exist today.

Page 29: Acute, Repeated-Dose and Residual Effects of Amphetamines

18

Chapter 2: The Effects of Intranasal d-Amphetamine and Methamphetamine on Regulatory Focus Measures in Humans 2.1 Introduction

Following decades of dormancy, the abuse of amphetamines has again become an

important public health issue globally. Methamphetamine, in particular, has generated the

greatest amount of concern because it is considered to have a higher abuse potential relative to

other amphetamines (e.g., d-amphetamine, the active ingredient in “Adderall”). The U.S.

Treatment Episode Data Set (TEDS) system enumerates substance abuse treatment episodes

across the Nation. According to this Data Set, in 2011, approximately 102,00 treatment-seeking

individuals aged 12 years or older reported methamphetamine as their primary substance of use,

while about only 8,000 users seeking treatment reported d-amphetamine as a principal drug of

abuse (TEDS, 2013). Furthermore, the Drug Abuse Warning Network (DAWN) provides data on

drug-related emergency room visits from major metropolitan areas (e.g., number of visits

associated with a particular drug or drug combination). In 2011, d-amphetamine-related

emergency department episodes were also low in comparison to methamphetamine-related

episodes (e.g., ~17,000 vs. ~102,000 episodes, respectively) (DAWN, 2011). An important

consideration, however, is that these abuse rates may be related to the fact that methamphetamine

is apparently easier to synthesize than d-amphetamine, making it a more readily accessible drug

on the illicit market.

Another explanation for the greater incidence of methamphetamine abuse has to do with the

slight structural difference between the two drugs: methamphetamine is the N-methylated analog

of d-amphetamine. Some researchers argue that the additional N-methyl group (CH3) in the

methamphetamine structure increases the lipid solubility of the compound, perhaps allowing it to

cross the blood-brain barrier more easily and have a faster onset of effects than d-amphetamine.

Page 30: Acute, Repeated-Dose and Residual Effects of Amphetamines

19

(e.g., Gulaboski et al., 2007; Nichols, 1994, de Wit et al., 1993). Consequently,

methamphetamine is considered to be a more robust reinforcer, which is expected to lead to a

higher abuse liability.

This notion, however, is inconsistent with empirical data from research collected in animals

(Hall et al., 2008; Yokel and Pickens, 1973) and humans (Lamb and Henningfield, 1994; Sevak

et al., 2009, Kirkpatrick et al., 2011). For example, data from behavioral studies conducted in

rats indicate that d-amphetamine and methamphetamine similarly increase locomotor activity

and self-administration at equipotent doses (Hall et al., 2008; Balster and Schuster, 1973; Yokel

and Pickens, 1973). Furthermore, in one double-blind study, Martin et al. (1971) administered

subcutaneous doses of d-amphetamine (7.5-30 mg/kg) and methamphetamine (15-30 mg/kg) in

humans and found that both drugs similarly increased cardiovascular activity and ratings of

euphoria in comparison to placebo. In general, these data are consistent with other reports

indicating that oral doses of d-amphetamine (2.5-45 mg) and methamphetamine (2.5-30 mg)

produce overlapping mood and reinforcing effects (Lamb and Henningfield, 1994; Sevak et al.,

2009).

It is important to note, however, that these investigations compared oral and subcutaneous

amphetamines at doses lower than those typically used by abusers of the drugs. Abusers also

tend to use drugs via routes that are associated with rapid onset of effects such as intranasal,

intravenous injection, and smoked. For example, illicit methamphetamine is reportedly used in

larger doses (e.g., greater than 25 mg) via the intranasal, intravenous, and smoked routes of

administration, presumably due to their faster onset of effects (Simon et al., 2002). As a result,

the above findings might be limited when attempting to extrapolate them to illicit

methamphetamine abuse.

Page 31: Acute, Repeated-Dose and Residual Effects of Amphetamines

20

Another reason for concerns surrounding methamphetamine abuse might be related to the

database collected in laboratory animals on methamphetamine-related neurotoxic effects. In

general, data from these studies indicate that methamphetamine produces deleterious

neurochemical consequences. For example, Ricaurte and colleagues (1980) administered large,

subcutaneous doses of methamphetamine (25 and 100 mg/kg) for 4 consecutive days to rats.

They found that methamphetamine produced significant decreases in dopamine and serotonin

levels in several brain regions (i.e., striatum, amygdala) 3-6 weeks following drug-

administration. The researchers concluded that methamphetamine produced long-term

neurotoxicity to dopamine and serotonin neurons. It is important to note, however, that the doses

administered in this study far exceeded those typically used by recreational humans users. For

example, recreational methamphetamine users usually take approximately 0.25-3.5 mg/kg (Cho

et al., 2001). In addition, Hart et al. (2012) recently published a critical review of findings from

neuroimaging research investigating the acute and long-term cognitive and neurotoxic effects of

methamphetamine. The researchers reported that the majority of theses studies included

inappropriate controls and limited cognitive testing and concluded that the findings were largely

overstated. As a result, the above results provide limited information about the consequences of

methamphetamine use in humans.

In an effort to address the limitations of previous research, the current experiment was

conducted as part of a larger investigation comparing the effects of amphetamines (Kirkpatrick et

al., 2011). Kirkpatrick and colleagues administered relatively large, intranasal doses of d-

amphetamine (12 and 50 mg/70kg), methamphetamine (12 and 50 mg/70kg), and placebo to

current methamphetamine users over the course of five 2-day blocks of sessions. On the first day

of each block (the “sample” session), participants were administered a single drug dose and a

Page 32: Acute, Repeated-Dose and Residual Effects of Amphetamines

21

monetary reinforcer (U.S. $5 or $20). On the following day (the “choice” session), participants

were given the opportunity to choose between the sampled drug and/or money. They found that

the drugs produced similar reinforcing effects: when the competing monetary reinforcer was

relatively low (i.e., $5), d-amphetamine and methamphetamine similarly increased drug self-

administration. It is important to note, however, that this larger investigation primarily examined

drug-related effects on self-administration using a standard choice procedure. Although these

measures provide potentially useful information about the similar abuse liability of

amphetamines, it is possible that only more sensitive subjective measures are able to detect

potential differences between d-amphetamine and methamphetamine.

Current research in the field of social psychology may provide useful tools for identifying

possible discrepant effects of amphetamines. For example, investigations on regulatory focus

theory (Higgins 1997, 1998) indicate that individuals can temporarily experience motivational

states of “promotion” focus (e.g., focus on hopes and accomplishments [gains]) and/or

“prevention” focus (e.g., focus on safety and responsibility [nonlosses]) (Higgins et al., 2001).

Other investigations indicate that when these motivational orientations (e.g., “promotion” focus

and “prevention” focus) are sustained by preferred strategies of goal pursuit (“eager” for

promotion focus and “vigilant” for prevention focus), individuals experience a regulatory “fit”

that strengthens their engagement in a task (Higgins, 2000, 2005). It is important to note that

human behavioral pharmacology labs often measure performance on psychomotor tasks, some of

which (i.e., Divided Attention Task [DAT]) require “vigilance” (prevention), while others (e.g.,

Digit Substitution Task [DSST]) require “eagerness” (promotion). It is possible that the

amphetamines might produce differential effects on these more nuanced subjective measures of

regulatory focus and engagement on relevant psychomotor tasks.

Page 33: Acute, Repeated-Dose and Residual Effects of Amphetamines

22

The present investigation was conducted in an effort to understand further the comparative

effects of d-amphetamine and methamphetamine on well-validated measures that are designed to

assess more sensitive behavioral measures, such as regulatory focus and regulatory engagement

effects (e.g., Regulatory Focus Questionnaire [RFQ], etc.). Single, intranasal doses of

methamphetamine and d-amphetamine (0, 12, 50mg/70kg each) were administered in current

methamphetamine users. We hypothesized that: 1) d-amphetamine and methamphetamine would

dose-dependently increase “prevention” focus because previous data indicated that intranasal

methamphetamine improved performance on tasks of “vigilance” (e.g., DAT) and produced no

significant performance effects on tasks relating to promotion system (e.g., DSST) (Hart et al.,

2008), and 2) d-amphetamine and methamphetamine would dose-dependently increase

engagement ratings on a task involving the use of “vigilant” strategies (DAT), suggesting a

possible “regulatory fit” with the prevention system. Because methamphetamine produced

greater increases in various subjective ratings compared to d-amphetamine (e.g., “high”) in the

larger behavioral study (Kirkpatrick et al., 2011), we further predicted that methamphetamine

would dose-dependently produce greater increases in subjective ratings of “prevention” focus

and “task engagement” in comparison to d-amphetamine.

2.2 Methods and Materials

2.2.1 Participants

Thirteen male research volunteers completed this 10-session outpatient study (mean age

= 37.4 ± 7.3 [mean ± standard deviation (SD)]): one was Asian, six were Black, two Hispanic,

and four were White. Participants had completed 14.8 ± 2.0 (mean ± SD) years of formal

education. All participants were solicited via word-of-mouth referral, and various online and

newspaper advertisements in New York City. Prior to study enrollment, participants signed a

Page 34: Acute, Repeated-Dose and Residual Effects of Amphetamines

23

consent form that was approved by the Institutional Review Board of the New York State

Psychiatric Institute (NYSPI); each passed comprehensive medical and psychiatric evaluations

and were in within normal weight ranges according to the 1983 Metropolitan Life Insurance

Company height/weight table (body mass index: 24.9 ± 2.7 [mean ± SD]). Three participants

met criteria for current methamphetamine dependence. All participants stated that they were not

seeking treatment for his drug use and no one met criteria for any other Axis I disorders. On

average, participants reported using methamphetamine 9.4 ± 4.7 days/month. Seven reported

current alcohol use (4-10 drinks/week), seven reported current cocaine use (1-8 days/month),

three reported current marijuana use (4-12 days/month), and four smoked three to 20 tobacco

cigarettes per day.

2.2.2 Pre-study training

Prior to starting the study, participants completed two training sessions (3-4 hours per

session) on computerized psychomotor tasks that would be used in the study in order to

minimize the effects of learning on task performance. On a separate day, participants received

the largest single dose of methamphetamine (50 mg/70kg) to be tested in order to monitor any

adverse reactions and provide them with experience with the study drug in the laboratory setting.

No adverse cardiovascular effects were noted. Participants were not informed of the dose until

study debriefing.

2.2.3 Design

This within-participants study examined two intranasal methamphetamine and two

amphetamine doses (12, 50 mg/70kg each drug) and placebo over the course of two weeks.

Table 1 shows that the study consisted of five 1-day blocks of sessions during which one drug

was administered. The present study was part of a larger study examining the reinforcing, mood

Page 35: Acute, Repeated-Dose and Residual Effects of Amphetamines

24

and psychomotor effects of methamphetamine and d-amphetamine. For the purposes of this

paper, such effects will not be discussed (Kirkpatrick et al., 2011).

2.2.4 Procedure

All laboratory sessions began at approximately 0900 hours and lasted for 6 hours.

Following a light breakfast, volunteers were required to pass a field sobriety test and give a urine

sample that tested negative for several drug metabolites (excluding amphetamines and THC).

They also completed a baseline psychomotor battery (see below). After baseline assessments,

participants insufflated the drug. All study measures were assessed 30 minutes post drug

administration. Upon study completion, participants were evaluated for signs of intoxication and

required to pass a field sobriety test after which they were excused and given compensation for

public transportation.

2.2.5 Psychomotor Task

The computerized 10-min Divided Attention Task consisted of simultaneous pursuit-

tracking and “vigilance” tasks (DAT; Miller et al., 1988). In this task, participants were required

to track a moving circle on the video screen using the mouse, and also signal when a small black

square appeared at any of the four corners of the screen. Accurate tracking of the moving

stimulus increased its speed proportionately. Data from the previous, larger study have already

been published (Kirkpatrick et al., 2011).

2.2.6 Regulatory Focus Questionnaire (RFQ)

The 11-item Regulatory Focus Questionnaire (RFQ; Higgins et al., 2001) consisted of a

series of 5-point lines labeled “never or seldom” at one end and “very often” at the other end.

Participants were required to rate their subjective histories of “promotion” and “prevention”

success (e.g., promotion success: “I feel like I have made progress toward being successful in my

Page 36: Acute, Repeated-Dose and Residual Effects of Amphetamines

25

life”; prevention success: “Not being careful enough has gotten me into trouble at times

(reversed scored)”). This measure is designed to assess subjective experiences of being effective

in each motivational system. A higher score on the promotion items reflects the “promotion

system working,” or a subjective history of effectiveness with promotion-related “eagerness”

(i.e., being bold and optimistic; being willing to take chances). A higher score on “prevention”

pride reflects the "prevention system working," or a subjective history of effectiveness with

“prevention”-related “vigilance” (i.e., being careful and accurate; avoiding mistakes).

2.2.7 State Regulatory Focus Questionnaire

The 3-item State Regulatory Focus Questionnaire (see Zhou and Pham, 2004) consisted

of a series of 9-point lines labeled with two consumer options (option A and B), one relating to

the promotion system (e.g., a product emphasizing advancement and achievement) and the other

relating to the “prevention” system (e.g., a product emphasizing safety and security) at each end.

Participants were required to indicate their preference using a 9-point scale: 1= option A and 9=

option B. Higher numbers indicate a higher preference for “prevention”-related strategies (e.g.,

being “vigilant”).

2.2.8 Task Engagement Questionnaire

The 11-item engagement strength questionnaire consisted of a series of 9-point lines

labeled “disagree strongly” at one end, “neutral” in the middle, and “agree strongly” at the other

end (Higgins, 2006). Immediately after the end of the DAT task, participants were required to

rate the extent to which they felt engaged in the task (e.g., “I was completely focused on the

task”) and their mood during the task (e.g., “I felt good during the task”). Participants were also

asked to rate “motivation to take the drug again” on a 9-point scale: 0= “not at all motivated,” 4=

neutral, and 8= “very motivated.”

Page 37: Acute, Repeated-Dose and Residual Effects of Amphetamines

26

2.2.9 Drug

The New York State Psychiatric Institute (NYSPI) Pharmacy provided methamphetamine

HCL [provided by the National Institute on Drug Addiction (NIDA)] and d-amphetamine sulfate

(provided by Cambrex, Charles City, IA, USA). Placebo consisted of powder containing only

lactose and was added to each active amphetamine dose (12 and 50 mg/70 kg) to achieve a final

weight of 60 mg/70 kg. A research nurse placed each dose in a small medicine cup along with a

straw. Participants were required to insufflate the full dose within a 30 second period in either

one or both nostrils. All drug doses were administered

2.2.10 Data Analysis

Repeated-measures analyses of variance (ANOVA) with planned comparisons were used

to determine the effects of d-amphetamine and methamphetamine on subjective ratings of

regulatory focus state and task engagement. The dependent measures were analyzed using a two-

factor repeated-measures ANOVA: the first factor was dose (0, 12, and 50 mg/70 kg), and the

second factor was drug condition (methamphetamine and d-amphetamine). For all analyses,

ANOVAs provided the error terms needed to calculate planned comparisons that were designed

to determine the effects of drug (methamphetamine vs d-amphetamine) and dose level (e.g., 0 mg

vs two active methamphetamine doses and 12 mg vs 50 mg methamphetamine dose). Data were

considered statistically significant at p < 0.05, using Huynh–Feldt corrections where appropriate.

2.3 Results

2.3.1 Regulatory Focus Questionnaire (RFQ)

Figure 2.1 shows the effects of dosing condition on participants’ responses to the

regulatory focus pride questionnaire items. Relative to placebo, both large doses and the 12-mg

Page 38: Acute, Repeated-Dose and Residual Effects of Amphetamines

27

methamphetamine dose increased participants’ subjective ratings of their “prevention” focus (p <

.05). No other regulatory focus effects were observed.

2.3.2 State Regulatory Focus Questionnaire

Figure 2.2 shows the effects of dosing condition on state regulatory focus ratings.

Compared to placebo, both large doses of amphetamines and the 12-mg dose of

methamphetamine increased self-reported preference for “prevention” focus strategies (p < .05).

No other significant drug effects on state regulatory focus were noted.

2.3.3 Task Engagement Questionnaire

Figure 2.3 shows the effects of dosing condition on selected task engagement ratings.

Relative to placebo, all active doses of amphetamines significantly increased ratings “completely

focused on the task,” and “felt involved during task,” (p < .05). Both large doses and the 12-mg

dose of methamphetamine significantly increased ratings of “motivated to get same drug dose”

compared to placebo (p < .05). Table 2.1 summarizes other significant effects observed on the

task engagement questionnaire.

2.4 Discussion

As predicted, the current findings indicate that both large doses of the drugs enhanced

ratings of “prevention” focus on both the state and chronic effectiveness measures of regulatory

focus. That is, both drugs enhanced participants’ preferences for “reliable” (over “luxurious”)

consumer options and effectiveness in being “vigilant.” Consistent with these results, d-

amphetamine and methamphetamine produced similar effects on task engagement: both drugs

increased ratings of “involvement” and “focus” during the task in all active dose conditions. In

general, these results are consistent with previous studies showing that d-amphetamine and

methamphetamine produce overlapping effects (Martin et al., 1971; Sevak et al., 2009;

Page 39: Acute, Repeated-Dose and Residual Effects of Amphetamines

28

Kirkpatrick et al., 2011). The current study extends earlier findings by providing the first data

directly comparing the regulatory focus effects of the two drugs in humans. These findings are

important because they expand the limited database documenting the social psychological effects

of amphetamines on motivational states.

The overall pattern of d-amphetamine- and methamphetamine-related effects suggests

that participants experienced a similar regulatory “fit” between their temporary motivational

state (e.g., “prevention” focus) and the psychomotor task designed to measure “vigilance” (e.g.,

DAT). Regulatory “fit” is experienced when an individual’s temporary motivational orientation

(e.g. “prevention”) is sustained by a task that requires the use of preferred strategies (e.g.,

“vigilant” strategies), and results in greater levels of engagement and enjoyment during a task. In

the current study, participants reported higher levels of engagement on the psychomotor task

under all active drug conditions (e.g, under high “prevention” focus conditions) relative to

placebo. This comment is speculative, however, and suggests avenues for further investigation.

For example, future research should include 1) a regulatory “nonfit” condition, which would

consist of a task, such as the Rapid Information Task (RIT), that requires the use of “eager”

strategies (i.e., making “hits”), and 2) additional measures that are designed to assess

characteristic emotions associated with regulatory “fit” (e.g., feeling “right,” “effective”).

We hypothesized that methamphetamine would produce greater increases in ratings of

regulatory focus and task engagement compared to d-amphetamine. This prediction was not

borne out. The drugs produced equipotent motivational effects. This prediction was based, in

part, on the previously published behavioral study indicating that methamphetamine produced a

more prominent effect on a subjective measure (e.g., in ratings of “high”) compared to d-

amphetamine (Kirkpatrick et al., 2011). It is important to note, however, that Kirkpatrick et al.

Page 40: Acute, Repeated-Dose and Residual Effects of Amphetamines

29

(2011) observed that d-amphetamine and methamphetamine produced nearly identical effects on

the majority of subjective measures. In general, the current observations are consistent with

these data from the previous study.

However, one potential explanation for the inconsistency with results from Kirkpatrick et

al. (2011) indicating that methamphetamine produces greater subjective effects (e.g., on ratings

of “high”) is that the previous investigation used a measure (e.g., Visual Analogue Scale [VAS])

that is designed to assess different, more general psychological concepts than those tested in the

current experiment (i.e., subjective mood, drug effects, and physical symptoms). The current

measures are designed to investigate temporary motivational states and involvement in a task

(e.g., of “eagerness” and “vigilance”) and allow for unique predications about distinct types of

desired end-states and “pleasure” (euphoria). It is possible that measures designed to assess

specific “prevention”—related behavioral tendencies (e.g., conservative bias in decision-making,

preference for stability over change, and concrete ways of thinking etc.) and other motivational

states (e.g., Regulatory Mode) would be more effective in differentiating between the two drugs.

Another potential explanation for the apparent incongruence between the present data and

previous results is related to different data collection/analysis methods. The current study

assessed amphetamine-related subjective effects at one time point (e.g., 30 minutes), whereas the

previous study included a wide range of time points and subjective-effect ratings data were

summed across these time points. It is unclear whether divergent regulatory focus and

engagement effects of the drugs would emerge at other points along the time course. Future

studies on amphetamine-related regulatory focus effects should include multiple data collection

times of observation (e.g., baseline-90 minutes)

Page 41: Acute, Repeated-Dose and Residual Effects of Amphetamines

30

It is intriguing that methamphetamine enhanced ratings on “prevention” focus and certain

engagement measures (e.g., “motivated to get same drug dose,” “felt good during task”) in the

low dose condition (12 mg) while d-amphetamine did not. It is important to note, however, that

there was a nonsignificant trend in the data indicating that the smaller amphetamine dose

increased “prevention focus” (p = .08). These data suggest a subtle quantitative difference

between both drugs and highlight the importance of considering dose potency when comparing

drug effects: it is possible that d-amphetamine would produce similar qualitative effects

compared to a 12-mg dose of methamphetamine at slightly higher doses (e.g., 20-30 mg). Future

research investigating the motivational effects of amphetamines should include a wider range of

doses.

In general, these results support our predictions. The present findings show that

relatively large intranasal doses of d-amphetamine and methamphetamine produce nearly

identical effects on regulatory focus and task engagement measures: both drugs increased ratings

of “prevention” focus and engagement in a task that was designed to assess “vigilance” and

inhibitory control. While the current data lend support to the idea that similar regulatory “fit”

experiences were produced between both d-amphetamine- and methamphetamine-related states

of “prevention” and the psychomotor task, future research is necessary. Another important

finding was that only methamphetamine produced self-reported increases in prevention focus

under the low dose condition (12-mg dose). This result highlights the importance of assessing

more sensitive self-report measures (e.g., regulatory focus) at a wider range of doses when

determining potential differences between the amphetamines.

Page 42: Acute, Repeated-Dose and Residual Effects of Amphetamines

31

Chapter 3: Acute, Repeated-dose and Residual Effects of 3,4-methylenedioxymethamphetamine MDMA in Humans 3.1 Introduction

Recreationally, 3,4-methylenedioxymethamphetamine (MDMA) is a popular

amphetamine that has attracted considerable attention in recent years. According to the United

Nations Office on Drugs and Crime (UNODC), amphetamine-type stimulants (ATS), which

include MDMA, are the second most-widely used drugs globally, only exceeded by cannabis use

(UNODC, 2012). Because multiple drugs are included in the ATS grouping - including

methamphetamine, d-amphetamine, and MDMA – it is difficult to distinguish the extent of

MDMA use from that of other drugs in this category. In United States, however, MDMA is

differentiated from other ATS on surveys of drug use. The U.S. National Survey on Drug Use

and Health, for example, provides estimates of individual drug use by people aged 12 years or

older. When asked about illicit drug use within the past 30 days (or past year), respondents

reported greater amounts of MDMA use than other substances including methamphetamine and

crack cocaine (SAMSHA, 2011). Together, these findings indicate that recreational MDMA use

is substantial.

MDMA is used in a variety of social settings, including at night and on weekends. During

this time, multiple doses may be taken within a 48-hour period, which could lead to dangerous

additive effects, residual effects and impact next-day functioning. Indeed, in the U.S., there have

been at least two recent deaths attributed to repeated-dosing effects on MDMA (also known as

Molly) (Pareles, 2013). In addition, some anecdotal reports suggest that MDMA users may

experience negative mood states (e.g., "Lethargic," "Depressed") in the days immediately

following MDMA administration (Curran & Travill 1997; Parrott & Lasky 1998).

Page 43: Acute, Repeated-Dose and Residual Effects of Amphetamines

32

The available data from laboratory investigations of the drug in humans have focused

almost exclusively on the acute effects of MDMA following single-dose administrations (e.g.,

Cami et al. 2000; Kuypers & Ramaekers, 2005, 2007; Tancer & Johanson 2001). In general,

MDMA, at doses ranging from ~75 to 145 mg, produces effects similar to those produced by d-

amphetamine and methamphetamine: increases in positive subjective effects, blood pressure, and

heart rate (Bedi et al., 2010; Cami et al. 2000; Harris et al. 2002; Tancer & Johanson, 2003,

2007). Unlike the amphetamines mentioned above, MDMA has also been shown to either

disrupt performance (e.g., Cami et al. 2000) or produce no performance alterations (e.g.,

Marrone et al. 2010; Kirkpatrick et al., 2012). These data, however, do not provide information

about next-day consequences of MDMA use; nor do they provide any information about

repeated-dose administration.

Only a few laboratory studies have assessed amphetamine-related effects following

repeated administration (Comer et al. 2001; Farré et al., 2004; Peiró et al., 2012). In one study,

Farre et al. (2004) investigated the effects of two consecutive oral doses of MDMA (100 mg

each) administered 24 hours apart. The researchers found that the second dose produced further

significant elevations in heart rate, blood pressure and ratings of euphoria compared to the first

dose. In contrast to these findings, tolerance appears to develop after repeated administration of

methamphetamine. For example, Comer et al. (2001) administered low oral doses (5-10 mg) of

the drug twice daily for 3 consecutive days and reported that by day 2 some positive subjective

effects were not significantly different from placebo. They also observed that more negative

subjective effects (e.g., dizzy, bad drug effect) emerged following repeated administration of the

drug. Of course, the apparent inconsistencies between the findings could be explained by the fact

that the amphetamines under investigation were different drugs with different pharmacological

Page 44: Acute, Repeated-Dose and Residual Effects of Amphetamines

33

profiles. For example, the half-life for MDMA is approximately 6 hours, whereas the half-life is

about 12 hours for methamphetamine. Another possibility is related to the dosing regimens

employed; Farre and colleagues administered two MDMA doses (100 mg each) separated by 24

hours, whereas Comer et al. (2001) gave methamphetamine (5, 10 mg) twice daily, at an 8-hour

interval, for 3 consecutive days.

In a more recent study of repeated MDMA administration, Peiró et al. (2012) gave two

doses separated by 2 hours: the first was 50 mg and the second was 100 mg. Relative to the

comparator condition (a single 100-mg dose), repeated MDMA administration significantly

increased ratings of euphoria but disrupted performance (i.e., increased response time on a

reaction time task). These findings indicate that the effects of an initial small dose of MDMA is

enhanced by a larger dose administered a couple of hours later. While the study by Peiró and

colleagues provided important information about repeated-dose administration of MDMA, it did

not include conditions during which the same dose was administered repeatedly. Another issue

that was not addressed in the two earlier MDMA studies is whether residual effects emerge

following repeated-dose administration, as was the case for methamphetamine.

In an effort to address limitations in the existing literature, we undertook a double-blind,

inpatient, within-participant study to evaluate the acute, repeated-dose and residual effects of oral

MDMA administration (0, 50, and 100 mg) on several dependent variables, including

cardiovascular, mood, and psychomotor performance effects. Placebo or MDMA was

administered at three different time points within a 36-hour period to measure acute and

repeated-dose effects. Following this period, placebo was administered over the next two days in

order to assess residual effects. We hypothesized that: 1) repeated-dose administration would

produce tolerance to “positive” subjective effects but additive cardiovascular effects; and 2)

Page 45: Acute, Repeated-Dose and Residual Effects of Amphetamines

34

“negative” mood effects would emerge in the days following three consecutive drug

administrations. We made no predictions about MDMA-related effects on performance, because

previous findings have been mixed.

3.2 Methods and Materials

3.2.1. Participants

Twelve research volunteers (mean age was 28.9 ± 7.2 [± SD]) completed this 15-day

inpatient study. Three participants were females (one Black and two Hispanic) and nine were

males (three Black, three Hispanic, three White). Participants were recruited by word-of-mouth

referrals, various online and newspaper advertisements in New York City newspapers and poster

flyers. On average, volunteers had completed 13.0 ± 1.7 [(mean ± standard deviation (SD)] years

of formal education. Prior to study enrollment, participants passed comprehensive medical and

psychiatric evaluations and were within normal weight ranges according to the Metropolitan Life

Insurance Company height/weight table (body mass index: 24.4 ± 3.8). All participants reported

current MDMA use (1.28 ± 0.95 times/week). Nine participants reported current cocaine use (1-

3 times/week), eleven reported current alcohol use (0.25-5 drinks/week) and marijuana use (1-7

times/week), and five smoked five to 30 tobacco cigarettes/day. No participant met criteria for

an Axis I disorder and no one was seeking treatment for her/his drug use.

Prior to study enrollment, each participant signed a consent form that was approved by

the Institutional Review Board of the New York Psychiatric Institute (NYSPI); upon discharge,

they were informed about experimental and drug conditions and were compensated at a rate of

$35/day for their participation. They were also paid an additional bonus of $35/day for

completing the entire 15-day study.

Page 46: Acute, Repeated-Dose and Residual Effects of Amphetamines

35

3.2.2. Pre-study training

Prior to starting the study, participants completed two training sessions (3-4 hours per

session) on computerized psychomotor tasks that would be used in the study in order to

minimize the effects of learning on task performance. They were also familiarized with the

residential laboratory and study procedures. On a separate day, participants received the largest

single dose of MDMA (100 mg) to be tested inpatient in order to monitor any adverse reactions

and provide them with experience with the study drug in the laboratory setting. Participants were

not informed of the dose until study debriefing.

3.2.3. Design

Three groups of four participants lived in a residential laboratory located in the New

York Psychiatric Institute for 15 days. Table 1 shows that the study consisted of 3 five-day

blocks of sessions, during which participants completed visual analog mood scales and

psychomotor task batteries before and after MDMA administration. Drug administrations

occurred twice daily - 0900 and 2100 hours – and were counterbalanced across as well as within

participant groups. On the first day of each block, only placebo was administered as this served

as a baseline period. On the second day of each block, MDMA (0, 50, or 100 mg) was

administered both in the morning and evening. On the third day of each block, placebo was

administered at 0900 hours and MDMA was administered at 2100 hours, followed by two

consecutive days of placebo administration at both dosing times. The three consecutive mornings

of placebo administration were included in order to examine residual MDMA effects and as a

washout period.

Page 47: Acute, Repeated-Dose and Residual Effects of Amphetamines

36

3.2.4. Procedure

Participants moved into the laboratory a day before the study began in order to further

orient them to the laboratory and practice experimental procedures. Each day volunteers were

awakened at 0800 hours. After waking, participants completed a visual analog sleep

questionnaire and baseline performance and subjective effects batteries beginning at 0815 hours.

Subsequently, each participant was administered a capsule containing drug or placebo and

weighed in a private vestibule. Then, they were given time to eat breakfast. Following breakfast,

from 1000 to 1715 hours, participants engaged in work activities (i.e., completed various

performance and subjective effects batteries) with an hour and a half lunch break (1300-1430

hours), during which the social area was available. In short, performance and subjective effects

measures were assessed at baseline and 60, 110, 160, 210, 335, 375, 420, and 465 minutes after

the first dose. Cardiovascular measures and oral temperature were obtained at baseline and 25,

50, 100, 150, 200, 250, 300, 325, 350, and 500 minutes post the first capsule administration.

Subjective-effects ratings were also collected 150 minutes after the second dose and

cardiovascular measures were assessed before and 25, 50, 100, and 150 minutes after the second

capsule dosing.

The social area was available to participants from 1715 to 2330 hours. During this

period, participants could interact with other study participants and engage in recreational

activities such as playing video games or watching videotaped films. Two films were shown

daily, beginning at 1900 and 2130 hours. Social behavior was collected using a computerized

observation program that prompted recording of each participant’s behavior every 2.5 minutes

(e.g., Haney et al., 2007). Behaviors were classified into private (time spent in

bathroom/bedroom) and social (time spent in the recreational room). Time spent in the social

Page 48: Acute, Repeated-Dose and Residual Effects of Amphetamines

37

area was further divided into time spent talking and time spent in silence. Outcomes were total

minutes spent engaging in each behavior per day. At 2100 hours, participants received their

second capsule. Lights were turned out at 2400 hours for an 8-hour sleep period. Participants

could smoke tobacco cigarettes and eat meals ad libitum from 0800 to 2330 hours.

3.2.5 Subjective effects and psychomotor battery

The computerized visual analog questionnaire consisted of 100-mm lines labeled “not at

all” at one end and “extremely” at the other end (described in Hart et al., 2003). The lines were

labeled with adjectives describing a mood (e.g. “Anxious,” “Talkative,” “Unmotivated”), a drug

effect (e.g. “Bad Drug Effect,” “Good Drug Effect,” “High”), or a physical symptom (e.g.,

“Headache,” “Muscle Pain,” “My heart is beating faster than usual”). Participants completed a

drug-effect questionnaire (DEQ), during which they were required to rate “good effects” and

“bad effects” on a five-point scale: 0 = “not at all” and 4 = “very much.” Ratings of drug

strength and desire to “take the drug again” were collected as well. At 2240 hours, participants

were asked to rate how “close [they feel] to those around me” and how much they “prefer to be

alone” on 100-mm lines labeled “not at all” on one end and “extremely” on the other end.

Computerized psychomotor tasks consisted of a 5 tasks: 1) the Digit-Symbol Substitution

task (DSST; McLeod et al., 1982), designed to assess changes in visuospatial processing, 2) the

Divided Attention Task (DAT), designed to assess changes in vigilance and inhibitory control

(Miller et al., 1988); 3) the Digit Recall Task, designed to assess changes in immediate and

delayed recall (Hart et al., 2001); 4) the Rapid Information Task (RIT), designed to assess

changes in sustained concentration and inhibitory control (Wesnes and Warburton, 1983); and 5)

the Repeated Acquisition Task, designed to assess changes in learning and memory (Kelly et al.,

1993).

Page 49: Acute, Repeated-Dose and Residual Effects of Amphetamines

38

3.2.6. Sleep monitoring

Objective sleep was measured by tracking gross motor activity using the Actiwatch®

Activity monitoring System (Actiwatch® ; Respironics Company, Bend, OR). This system

provided measures of total sleep time, sleep onset latency, sleep efficiency (total sleep time as a

percentage of time in bed), and number of wake bouts (Kushida et al., 2001). Subjective sleep

experience from the immediately preceding sleep period was measured by a visual analog sleep

questionnaire each morning. This questionnaire consisted of a series of 100-mm lines labeled

"not at all" at one end and "extremely" at the other end. The lines were labeled: "I slept well last

night," "I woke up early this morning," "I fell asleep easily last night," "I feel clear-headed this

morning," "I woke up often last night," "I am satisfied with my sleep last night," and a fill-in

question in which participants were asked to estimate the number of hours they thought they

slept the previous night (Haney et al. 2001).

3.2.7 Drug

Placebo and two active MDMA doses (50 and 100 mg) were tested. MDMA

hydrochloride was provided by Dr. David Nichols of Purdue University and packaged by the

Pharmacy Department of the New York State Psychiatric Institute. The appropriate amount of

MDMA hydrochloride was placed into a white #00 opaque capsule along with lactose filler.

Placebo consisted of white #00 capsules containing only lactose.

3.2.8 Data Analysis

Acute cardiovascular activity, subjective effects and psychomotor performance were

analyzed using two-factor repeated-measures analyses of variance (ANOVAs): the first factor

was drug condition (placebo, 50, and 100 mg MDMA), and the second factor was time (time and

number of assessments varied depending on the measure, e.g. cardiovascular effects were

Page 50: Acute, Repeated-Dose and Residual Effects of Amphetamines

39

assessed at time points baseline, 25, 50, 100, 150, 200, 250, and 300 minutes post drug

administration). Repeated-dose and residual effects analyses included an additional third factor

of day. Repeated-dose effects were assessed by comparing peak effects produced by the first

drug administration within each dosing to the third drug administration. The residual effects of

repeated drug administration was examined using planned contrasts that compared performance

and mood upon awakening the first placebo day following active drug administration to the

corresponding first placebo day following the period of placebo administration. Sleep and

cigarette intake data were analyzed using a two-factor ANOVA: the first factor was drug

condition and the second factor was day. For all analyses, ANOVAs provided the error terms

needed to calculate the following planned comparisons: acute and residual 1) placebo vs. active

doses and 2) 50 vs. 100 mg MDMA; repeated dose 1) 50 mg: (first versus third administration)

and 2) 100 mg: (first versus third administration). p Values were considered statistically

significant at less than 0.05, using Huynh-Feldt corrections when appropriate.

3.3 Results

3.3.1 Cardiovascular and oral temperature effects

Acute effects

Figure 3.1 shows that MDMA increased heart rate over time. Active doses produced

significant increases in heart rate and blood pressure compared to placebo (p < 0.01). The 100-

mg dose produced significantly larger increases in systolic and diastolic blood pressure than the

50 mg dose (p < 0.05). There were no significant drug effects on oral temperature.

Repeated-dose effects

The only cardiovascular measure that varied significantly as a function of repeated-

dosing was heart rate. By the third administration, heart rate no longer was significantly elevated

Page 51: Acute, Repeated-Dose and Residual Effects of Amphetamines

40

above placebo (see Figure 3.2). Under the 50-mg condition, peak heart rate was 100.75 bpm

following the first administration but only 90.67 bpm following the third administration (p <

0.02). Under the 100-mg condition, heart rate peaked at 105.67 bpm following the first drug

administration but was down to 89.67 after the third administration (p < 0.001). Following the

third administration, active drug conditions significantly increased systolic and diastolic blood

pressure relative to placebo (p < .001). No other significant cardiovascular effects were

observed.

Residual effects

There were no significant residual cardiovascular effects.

3.3.2 Subjective effects

Acute effects

Figure 3.1 shows the effects of MDMA on selected subjective-effect ratings over time.

Peak effects for both doses were observed 1 hour after ingestion. Relative to placebo, MDMA

(50 and 100 mg) significantly increased ratings of “good drug effect” and “stimulated” (p <

0.0001), and the larger dose produced greater increases than the smaller dose (p < 0.001). In

addition, while both active doses produced significantly larger increases on ratings of “sedated”

compared to placebo (p < 0.05), only the 100-mg dose significantly increased ratings of “jittery”

(p < 0.001 for all comparisons). On the DEQ, both MDMA doses increased ratings of “good

effect” and “drug strength” compared to placebo (p = 0.001); the 100-mg dose produced greater

increases on these measures than the 50-mg dose (p < 0.05). Table 3.2 summarizes other

significant acute effects observed on the visual analog questionnaire and the DEQ. In general,

positive subjective effects were dose-dependently increased.

Page 52: Acute, Repeated-Dose and Residual Effects of Amphetamines

41

Repeated-dose effects

Figure 3.2 shows the repeated-dose effects of MDMA on selected subjective-effect

ratings. Rating of “good drug effect” and “jittery” were attenuated with repeated drug

administration, though still remained significantly elevated relative to placebo. Under the 100-

mg condition, both ratings were significantly decreased following the third administration

compared to the first administration (p < 0.05). Following the third dosing, active drug

conditions significantly increased ratings of “good drug effect” and “stimulated” compared to

placebo, (p = 0.001). Only the larger dose increased ratings of “sedated” and “jittery” (p <

0.05). Following the second administration (Day 2), ratings of “I feel close to those around me”

were significantly increased by both MDMA doses compared to placebo (p < 0.05). Following

the third administration (Day 3), closeness ratings remained significantly increased by the larger

MDMA dose compared to placebo (p < 0.05). Relative to placebo, both doses of MDMA

significantly decreased ratings of “I would prefer to be alone” after the third dosing (p < 0.01).

Other significant VAS and DEQ effects following repeated-dosing are summarized in Tables 3.3

and 3.4.

Residual effects

No significant residual subjective effects were noted.

3.3.3 Psychomotor performance effects

Acute effects

There were no significant acute drug effects on psychomotor performance.

Residual effects

No significant residual performance effects were observed.

Page 53: Acute, Repeated-Dose and Residual Effects of Amphetamines

42

3.3.4 Sleep effects

Figure 3.3 illustrates the effects of MDMA administration on objective and subjective

sleep measures during the sleep period that began 3 hours after the second drug administration

(Day 2). During this period, the larger MDMA dose decreased both the objective and subjective

number of hours participants slept relative to placebo (p < 0.05). While both MDMA doses

decreased participants’ ratings of “fell asleep easily” compared to placebo (p < 0.05), only the

larger MDMA dose increased objective measures of sleep onset latency (p < 0.01 for all

comparisons). Under both active drug conditions, subjective ratings of “fell asleep easily” were

significantly increased on Day 3 compared to Day 2 (p = 0.001). No other significant sleep

effects were observed.

3.3.5 Effects on cigarette smoking

For the six participants who smoked cigarettes during the study, both MDMA doses

increased the number of cigarettes smoked compared to placebo on Day 2 (p < .05). The average

increase was approximately four to five cigarettes (15.7±2.7 [50 mg] and 15.3±2.8 [100 mg], vs.

10.8±2.2 [placebo]). No other significant effects on cigarette intake were observed.

3.3.6 Effects on social interactions

Only the larger MDMA dose increased the amount of time participants engaged in verbal

conversation in the social area compared to placebo following the third administration (Day 3) (P

< 0.05). No other significant effects were noted.

Page 54: Acute, Repeated-Dose and Residual Effects of Amphetamines

43

3.4 Discussion

The current findings show that repeated-dosing administration of MDMA produced

mixed cardiovascular and subjective effects. Blood pressure was significantly increased after

each of the three successive MDMA administrations, but heart rate increases were no longer

significant by the third administration. Subjective-effect ratings following repeated dosing were

also complex. Both positive (e.g., “good drug effect”) and negative (e.g., “jittery”) ratings were

significantly elevated across all drug dosings. However, even though ratings were significantly

increased compared to placebo, elevations produced by the larger MDMA dose were markedly

smaller after the third administration compared to the first, indicating partial tolerance. In

general, these data replicate previous results from investigations assessing acute MDMA-related

effects (e.g., Camí et al., 2000; Kirkpatrick et al., 2012); they extend such findings by

demonstrating attenuation of some effects following repeated MDMA administration.

The current result showing that repeated MDMA dosing produced tolerance to heart rate

elevations and the finding that repeated drug administration did not produce additive blood

pressure effects were unexpected. Anecdotally, MDMA is sometimes used in multiple doses

within a short period, which has raised concerns about potential dangerous additive

cardiovascular effects. Previously, researchers investigated this issue and reported data consistent

with this concern. In the first study, Farré and colleagues (2004) administered two 100-mg

MDMA doses separated by 24 hours and assessed multiple measures, including cardiovascular

effects. They found that the second MDMA administration produced significantly larger

increases in heart rate and blood pressure than did the first. In another study, Peiró et al. (2012)

assessed MDMA repeated-dose effects by administering a smaller dose (50 mg) followed 2

hours later by a larger dose (100 mg) using a single 100-mg dose as a comparator. These

Page 55: Acute, Repeated-Dose and Residual Effects of Amphetamines

44

researchers also reported significant cardiovascular elevations under the two active drug

conditions. One potential reason for the apparent inconsistent findings between the present data

and previous results is related to the different study designs. The current study assessed MDMA-

related repeated effects following three drug administrations at 12- and 24- hour intervals,

respectively, whereas previous investigations of MDMA repeated effects only included 2

administrations at 24- and 2- hour intervals, respectively (Farré et al., 2004; Peiró et al., 2012).

It is important to also note that while the studies by Farré et al. (2004) and Peiró et al. (2012)

found additive cardiovascular activity, these effects were small and unlikely to prompt clinical

concerns.

Our results indicate a diminution but not absence of positive and negative subjective

effects with repeated dosing. Despite some evidence for partial tolerance, it should be noted that

participants still reported substantial amounts of euphoria after the third MDMA dose relative to

placebo. Previous research examining two MDMA administrations did not demonstrate any

attenuation of subjective effects (Farré et al. 2004; Peiró et al. (2012). However, when another

amphetamine—methamphetamine (5, 10 mg)—was assessed using a 3-dose study design similar

to the current study, tolerance to some positive effects was observed (Comer et al., 2001).

Comer and colleagues (2001) reported a significant diminution of “good drug effect” and “high”

by the third methamphetamine administration compared the first administration. Decreased

negative mood effects (e.g., “jittery”) were also observed with repeated dosing in the current

study. This is an important finding because negative mood effects can sometimes decrease drug

intake. Future studies should investigate this issue by measuring actual drug intake and variation

in positive and negative subjective effects overtime.

Page 56: Acute, Repeated-Dose and Residual Effects of Amphetamines

45

The current study also sought to expand upon the limited database documenting residual

mood effects of MDMA administration. Anecdotal reports indicate the emergence of a short-

term depressive state in the days after MDMA use, a phenomenon colloquially referred to as

“Suicide Tuesday” (e.g., Parrott and Lasky, 1998). Accordingly, we hypothesized that negative

subjective effects would emerge in the days following three successive drug administrations.

The current data do not lend support for the hypothesis, as negative mood ratings (i.e.,

“depressed,” “miserable,” and “anxious”) were not significantly altered 12- to 48- hours after the

third MDMA dosing. Multiple factors could potentially account for the apparent discrepant

laboratory data and anecdotal observations. In the natural ecology, individuals frequently take

multiple drugs (e.g., alcohol and other sedatives) and may not get sufficient amounts of sleep

following an evening of drug use. In the current study, only one drug was tested (MDMA), and

each evening lights were turned out for a required 8-hour sleep period.

Objective and subjective measures of sleep were disrupted by the larger MDMA dose

administered three hours before the sleep period (Day 2). Specifically, participants slept

approximately two fewer hours and reported great difficulties falling asleep when the 100-mg

MDMA dose was administered compared to placebo. Under the 50-mg dose condition, the only

effect observed on sleep was that participants’ reported greater difficulties falling asleep. In

general, these findings are in agreement with data from a previous investigation evaluating

MDMA-related sleep effects (Randall et al., 2009). Randall and colleagues (2009) administered

a single MDMA dose (2 mg/kg) five hours prior to bedtime. They showed that the drug

increased sleep latency and decreased total sleep time without producing next-day sleepiness.

Similarly, we did not observe next-day mood or psychomotor performance alterations.

Page 57: Acute, Repeated-Dose and Residual Effects of Amphetamines

46

In conclusion, the current findings show that cardiovascular effects following repeated

MDMA administrations diminished (heart rate) or remained the same (blood pressure). These

findings are important because of anecdotal concerns about dangerous additive cardiovascular

effects after multiple doses of the drug administered within a relatively short time frame. We

found no evidence supporting such concerns. Another important finding was that negative mood

following repeated MDMA dosing was unaltered. The drug produced few residual effects, even

after repeated dosing. Future studies assessing the repeated-dosing effects of MDMA should

decrease the dosing interval to further characterize drug-related effects on a range of human

behaviors.

Page 58: Acute, Repeated-Dose and Residual Effects of Amphetamines

47

Chapter 4: General Discussion

Concern about illicit use and abuse of amphetamines has increased dramatically over the

past decade. Yet, much of our knowledge about amphetamine-related effects in humans is

anecdotal. The current studies addressed two gaps in our knowledge relating to effects of these

drugs in humans: 1) the overlapping and divergent effects of d-amphetamine and

methamphetamine, and 2) MDMA-related repeated-dose and residual effects.

4.1 d-amphetamine and methamphetamine Although d-amphetamine and methamphetamine are commonly regarded as distinct in

the U.S., there are few empirical studies that directly compare the effects of these drugs in

humans. Study 1 addresses this issue by comparing the effects of the amphetamines on well-

validated measures of regulatory focus and task engagement. Data from this study showed that

d-amphetamine and methamphetamine produce nearly identical effects: both drugs increased

both “prevention” focus and enhanced engagement on a task assessing vigilance, suggesting a

regulatory “fit.” In general, these results are consistent with previous data from investigations on

the subjective effects of d-amphetamine and methamphetamine (Kirkpatrick et al., 2011; Martin

et al., 1971; Sevak et al., 2009). These data are the first to demonstrate that intranasal d-

amphetamine and methamphetamine produce nearly identical stimulant effects on more sensitive

psychological measures of regulatory focus and engagement.

It is clear from findings from early and current research that d-amphetamine and

methamphetamine produce a similar profile of prototypical stimulant effects. This empirical

evidence suggests that the drugs are essentially the same, which is consistent with the way in

which amphetamines were viewed for most of the twentieth century. The drugs were used

interchangeably to treat a wide range of medical conditions, including narcolepsy and obesity,

Page 59: Acute, Repeated-Dose and Residual Effects of Amphetamines

48

and considered panaceas. Despite the fact that the amphetamines were perceived as similar for

over four decades, the general public currently views d-amphetamine and methamphetamine as

markedly different. For example, d-amphetamine is considered a safe and effective therapeutic

for the treatment of ADHD; in 2012, Adderall (e.g., d-amphetamine) was ranked 70 out of 200 of

the most widely prescribed medications (Bartholow, 2012). In contrast, methamphetamine is

perceived as a more potent, addictive drug and is rarely prescribed as an ADHD medication

(Bartholow, 2012).

As in the past, considerable national concern regarding the purported deleterious effects of

methamphetamine abuse has encouraged lawmakers to pass restrictive legislation. In 2005, the

U.S. federal government passed the Combat Methamphetamine Epidemic Act which requires

identification and a signature for sales of over-the-counter medications containing ingredients

that are involved in the illicit synthesis of methamphetamine (e.g., pseudoephedrine and

ephedrine). It is important to note, however, that data from early and current empirical studies

are inconsistent with recent public concerns and legislation relating to methamphetamine.

4.2 3,4-methylenedioxymethamphetamine

An important gap addressed by the current study was the lack of information about the

repeated-dose and residual effects of MDMA. The major scientific contribution of the current

study is that it was the first investigation of the cardiovascular, subjective and performance

effects of repeated administration of a wide range of MDMA doses (0, 50, 100 mg). Following

repeated dosing, heart rate elevations were no longer statistically significant and blood pressure

increases remained the same. Subjective-effect ratings remained significantly increased, but

such increases were diminished relative to acute drug effects. Measures of sleep were decreased

only on the evening following two active MDMA administrations and no performance alterations

Page 60: Acute, Repeated-Dose and Residual Effects of Amphetamines

49

or residual effects were observed.

These findings provide important public health information regarding the consequences

of MDMA use. Users of illicit MDMA reportedly administer multiple doses of the drug within a

relatively short time frame, which has sparked concerns about dangerous additive cardiovascular

effects. However, data from the present study do not support anecdotal reports of toxic effects

associated with this common recreational MDMA use pattern. Furthermore, the finding that

MDMA produced no residual mood effects is important given popular reports about depressive

states in the days following repeated dosing.

While there is a plethora of research on amphetamine- and methamphetamine-related

effects, the database documenting MDMA-related effects is limited. Questions still remain about

the effects of this amphetamine derivative in humans. For example, it is possible that additive

cardiovascular and negative mood effects may emerge following multiple dosing of MDMA at

shorter intervals. Future studies on MDMA-related repeated-dosing effects are needed and

should decrease the dosing interval.

4.3 Concluding Remarks

As in the past, findings from empirical research are inconsistent with conventional

perceptions of amphetamines in today’s society. The present results indicate that d-amphetamine

and methamphetamine produce a similar profile of effects. Yet, d-amphetamine and

methamphetamine are viewed as distinct drugs. Data from study 2 show no dangerous additive

cardiovascular or residual effects of MDMA. Abuse of this drug, however, is commonly

associated with deleterious cardiovascular and mood effects. As a result of public concern,

methamphetamine is often ignored as an effective medication and MDMA remains categorized

as a dangerous Schedule I drug with no acceptable medical use.

Page 61: Acute, Repeated-Dose and Residual Effects of Amphetamines

50

History relating to amphetamines appears to be repeating itself. Since the early twentieth

century, empirical evidence indicates that amphetamines are nearly identical drugs. However,

the best available scientific data are often ignored in the face of sensational media claims about

the dangerous effects associated with abuse of a particular amphetamine. Informed by anecdotal

reports, lawmakers pass legislation distinguishing various amphetamines. In the future, it is

likely that similar statements will be made in the popular press about an unfamiliar “new” type of

amphetamine and, in turn, legislation will hastily be passed. However, the present thesis aims to

highlight the importance of a critical examination of available early and current empirical

evidence on the effects of amphetamines in humans. This scientific approach will hopefully

prevent the enactment of premature public policies.

Page 62: Acute, Repeated-Dose and Residual Effects of Amphetamines

51

References

Allott K., & Redman J. (2006). Patterns of use and harm reduction practices of ecstasy users in Australia. Drug Alcohol Depend., 82, 168–176.

AMA Council on Pharmacy and Chemistry. (1937). Present Status of Benzedrine Sulfate.

Journal of the American Medical Association, 109, 2064–2069. AMA Council on Pharmacy and Chemistry. (1947). Drugs for Obesity. Journal of the American

Medical Association, 134, 527–529. Anglin, M.D., Burke, C., Perrochet, B., Stamper, E., & Dawud-Noursi, S. (2000). The History of

the Methamphetamine Problem. Journal of Psychoactive Drugs, 32, 2, 137-141. Anon. (May 10, 1937). Pep-Pill Poisoning. Time. Retrieved from http://time.com/ Anon. (1937). Students warned against pep pills at examinations. Science Newsletter, 31, 377. Anon. (September 14, 1942). Medicine: Nazi Pep-Pills. Time. Retrieved from http://time.com/ Anon. (February 21, 1944). Benzedrine Alert. Time. Retrieved from http://time.com/ Bakst, H.J. (1944). Daily Use of Benzedrine Sulfate Over a Period of Nine Years. United States

Naval Medical Bulletin, 43, 1228-1231. Balster, R.L., & Schuster, C.R. (1973). A comparison of d-amphetamine, 1-amphetamine, and

methamphetamine self-administration in rhesus monkeys. Pharmacol Biochem Behav, 1, 67-71.

Bartholow, (2012). Top 200 prescription drugs of 2011. Pharmacy Times. Retrieved from

http://www.pharmacytimes.com/publications/issue/2013/July2013/Top-200-Drugs-of-2012

Bedi G., Hyman D., & de Wit H. (2010). Is ecstasy an “empathogen”? Effects of 3,4-

methylenedioxymethamphetamine on prosocial feelings and identification of emotional states in others. Biol. Psychiatry, 68, 1134–1140.

Benzenhofer, U., & Passie, T. (2010). Rediscovering MDMA (ecstasy): the role of the American

chemist Alexander T. Shulgin. Addiction, 105, 1355-1361. Bernschneider-Reif, S., Oxler, F., & Freudenmann, R.W. (2006). The origin of MDMA

(“Ecstasy”)- separating the facts from the myth. Pharmazie, 61, 11, 866-962. Bett, W.R. (1946). Benzedrine Sulphate in Clinical Medicine: A Survey of the Literature.

Postgrad Med J, 22, 205-218.

Page 63: Acute, Repeated-Dose and Residual Effects of Amphetamines

52

Black, J. (June 21, 1970). The ‘Speed’ that kills or worse. New York Times. Retrieved from http://www.nytimes.com/

Bloomberg, W. (1939). Effects of Benzedrine in Altering Mental and Emotional Processes.

Proc. Assoc. Res. Ment. Nerv. Dis.,19, 172–79. Bradley, A., & Bowen, M. (1940). School Performance of Children Receiving Amphetamine (Benzedrine) Sulfate. Am. J. Orthopsychiat., 10, 4. Camí J., Farre M., Mas M., Roset P.N., Poudevida S., Mas A., San L., & de la Torre R. (2000).

Human pharmacology of 3,4-methylenedioxymethamphetamine (“ecstasy”): psychomotor performance and subjective effects. J Clin Psychopharmacol., 20, 455–466.

Chen, K. K., & Meek, W. J. (1926). Further Studies of the Effect of Ephedrine on the

Circulation. J. Pharmacol. & Exper. Therap., 28, 31. Cho, A.K., Melega, W.P., Kuczenski R., & Segal D.S. (2001). Relevance of pharmacokinetic

parameters in animal models of methamphetamine abuse. Synapse, 39, 161-6. Comer S.D., Hart C.L., Ward A.S., Haney M., Foltin R.W., & Fischman M.W. (2001). Effects of

repeated oral methamphetamine administration in humans. Psychopharmacology, 155, 397–404.

Curran V.H., & Travill R.A. (1997). Mood and cognitive effects of 3,4-

methylenedioxymethamphetamine (MDMA, `ecstasy’): week-end `high’ followed by mid-week low. Addiction, 92, 821-831.

Davis, P.L., & Stewart, W.B. (1938). The Use of Benzedrine Sulfate in Postencephalitic

Parkinsonism. J. Am. Med. Assoc., 110, 23, 1890-1892. de Wit, Harriet, Susan Dudish, and John Ambre. (1993). Subjective and behavioral effects of

diazepam depend on its rate of onset. Psychopharmacology, 112.2-3, 324-330. Dworkin, S.I., S Mirkis, and J.E. Smith. (1995). Response-dependent versus Response-

Independent Presentation of Cocaine: Differences in the Lethal effects of the Drug. Psychopharmacology, 117, 262-266.

Farber, L. (December 11, 1966). Ours is the Addicted Society. New York Times. Retrieved from

http://www.nytimes.com/ Farre M., de la Torre R., Ó Mathúna B., Roset P.N., Peiró A.M., Torrens M., Ortuño J., Pujadas

M., & Camí J. (2004). Repeated doses administration of MDMA in humans: Pharmacological effects and pharmacokinetics. Psychopharmacology, 173, 364-375.

Page 64: Acute, Repeated-Dose and Residual Effects of Amphetamines

53

Foltin, R.W., & Haney M. (2004). Intranasal cocaine in humans: acute tolerance, cardiovascular and subjective effects. Pharmacol Biochem Be., 78, 93-101.

Fournier, J.C., DeRubeis, R.J., Hollon, S.D., Dimidjian, S., Amsterdam, J.D., Shelton, R.C., &

Fawcett, J. (2010). Antidepressant drug effects and depression severity: a patient-level meta-analysis. JAMA, 303, 1, 47-53.

Garber, M. (May 31, 2013). Pilot’s Salt: The Third Reich Kept Its Soldiers Alert With Meth.

Atlantic Monthly. Retrieved from http://www.theatlantic.com/technology/archive/2013/05/pilots-salt-the-third-reich-kept-its-soldiers-alert-with-meth/276429/

Golden, S.A.O., (August 19, 1967). Hippie Regulars on Haight want Part-Timers to Take a Trip.

New York Times. Retrieved from http://www.nytimes.com/ Goodman, L., & Gilman, A. (1941). The pharmacological basis of Therapeutics: A textbook of

pharmacology, toxicology and therapeutics for physicians and medical students. New York: The Macmillian Company.

Grobler, S.R., Chikte, U., & Westraat, J. (2011). The pH Levels of Different Methamphetamine

Drug Samples on the Street Market in Cape Town. ISRN Dentistry,1. doi:10.5402/2011/974768. PMC 3189445. PMID 21991491.

Gulaboski R., Cordeiro M.N., Milhazes N., Garrido J., Borges F., Jorge, M., Pereira, C.M.,

Bogeski, I., Morales, A.H., Naumoski, B., & Silva, A.F. (2007). Evaluation of the lipophilic properties of opioids, amphetamine-like drugs, and metabolites through electrochemical studies at the interface between two immiscible solutions. Anal Biochem., 361, 236-43.

Guttman, E. (1936). “The Effect of Benzedrine on Depressive States,” J. Ment. Sci., 82, 618-

625.

Guttman, E. & Sargant, W. (1937). Observations on Benzedrine. Brit. Med. J., 1, 1013–15. Hall, D.A., Stanis, J.J., Marquez Avila, H., & Gulley, J.M. (2008). A comparison of

amphetamine and methamphetamine-induced locomotor activity in rats: evidence for qualitative differences in behavior. Psychopharmacology, 195, 469-78.

Haney M., Ward A.S., Comer S.D., Hart C.L., Foltin R.W., & Fischman M.W. (2001).

Bupropion SR worsens mood during marijuana withdrawal in humans. Psychopharmacology, 155, 171–179.

Haney M., Gunderson E.W., Rabkin J., Hart C.L., Vosburg S.K., Comer S.D., & Foltin R.W.

(2007). Dronabinol and marijuana in HIV-positive marijuana smokers. Caloric intake, mood and sleep. JAIDS, 45, 545–554.

Page 65: Acute, Repeated-Dose and Residual Effects of Amphetamines

54

Hardman, H.F., Haavik, C.O., & Seevers, M.H. (1973). Relationship of the structure of mescaline and seven analogs to toxicity and behavior in five species of laboratory animals. Toxicology and Applied Pharmacology, 25, 299-309.

Hart C.L., Ward A.S., Haney M., Foltin R.W., & Fischman M.W. (2001). Methamphetamine

self-administration by humans. Psychopharmacology, 157, 75–81. Hart C.L., Ward A.S., Haney M., Nasser J., & Foltin R.W. (2003). Methamphetamine attenuates

disruptions in performance and mood during simulated night-shift work. Psychopharmacology, 169, 42–51.

Hart, C.L., Gunderson, E.W., Perez, A., Kirkpatrick, M.G., Thurmond, A., Comer, S.D., &

Foltin, R.W. (2008). Acute Physiological and Behavioral Effects of Intranasal Methamphetamine in Humans. Neuropsychopharmacology, 33, 1847–1855.

Hart, C.L. & Ksir, C. (2011). Drugs, Society and Human Behavior. New York: McGraw-Hill

Companies.

Hart, C.L., Marvin, C.B., Silver, R., & Smith, E.E. (2012). Is cognitive functioning impaired in methamphetamine users? A critical review. Neuropsychopharmacology 37, 586-608.

Harris D.S., Baggott M., Mendelson J.H., Mendelson J.E., & Jones R.T. (2002). Subjective and

hormonal effects of 3,4-methylenedioxymethamphetamine (MDMA) in humans. Psychopharmacology, 162, 396-405.

Heal, D.J., Smith, S.L., Gosden, J., & Nutt, D.J. (2013). Amphetamine, past and present – a

pharmacological and clinical perspective. Journal of Psychopharmacology, 27, 6, 479–496.

Higgins, E.T. (1997). Beyond pleasure and pain. American Psychologist, 52, 1280- 1300. Higgins, E.T. (1998). Promotion and prevention: Regulatory focus as a motivational principle. In M. P. Zanna (Ed.). Advances in Experimental Social Psychology, (Vol. 30, pp. 1-46). New York: Academic Press. Higgins, E.T. (2000). Making a good decision: Value from fit. American Psychologist, 55, 1217-1230. Higgins, E.T., Friedman, R.S., Harlow, R.E., Idson, L.C., Ayduk, O.N. & Taylor, A. (2001). Achievement orientations from subjective histories of success: promotion pride versus prevention pride. European Journal of Social Psychology, 31, 3-23. Higgins, E.T. (2005). Value from regulatory fit. Current Directions in Psychological Science,

14, 209 - 213.

Page 66: Acute, Repeated-Dose and Residual Effects of Amphetamines

55

Holland, J. (2001). The History of MDMA. In A. Holland, J. (Ed), Ecstasy: The Complete Guide: A Comprehensive Look at the Risks and Benefits of MDMA. (pp. 11-20). Vermont: Park Street Press.

Iverson, L.L. (2008). Speed, Ecstasy, Ritalin: The Science of Amphetamines. Oxford: Oxford

University Press. Jackson, C.O. (1971). The Amphetamine Inhaler: A Case Study of Medical Abuse. Journal of

the History of Medicine, 26, 187–196. Jackson, C.O. (1975). The Amphetamine Democracy: Medicinal Abuse in the Popular Culture.

The South Atlantic Quarterly, 74, 3, 308-323. Jackson, C.O. (1976). Before the Drug Culture: Barbiturate/Amphetamine Abuse in American

Society. Clio Medica, 11, 47–58. Karch, S.B. (2011). A Historical Review of MDMA. The Open Forensic Science Journal, 4, 20-

24. Kelly T.H., Foltin R.W., Emurian C.S., & Fischman M.W. (1993). Performance-based testing for

drugs of abuse: dose and time profiles of marijuana, amphetamine, alcohol, and diazepam. J Anal Toxicol., 17, 264–272.

Kifner, J. (October 11, 1967). The East Village: A Changing Scene for Hippes. New York Times.

Retrieved from http://www.nytimes.com/ Kirkpatrick M.G., Gunderson E.W., Johanson C.E., Levin F.R., Foltin R.W., & Hart C.L.

(2011). Comparison of intranasal methamphetamine and d-amphetamine self-administration by humans. Addiction, 107, 783-791.

Kirkpatrick M.G., Gunderson E.W., Perez A.Y., Haney M., Foltin R.W., & Hart C.L. (2012). A

direct comparison of the behavioral and physiological effects of methamphetamine and 3,4-methylenedioxymethamphetamine (MDMA) in humans. Psychopharmacology, Berl. 219, 109-22.

Kremers, E. & Sonnedecker, G. (1976). Kremers and Urdang's History of Pharmacy. 4th rev. ed.

Madison: WI Amer. Inst. History of Pharmacy. Kunstadter, R.H. (1940). Experience with Benzedrine Sulfate in the Management of Obesity in

Children. Journal of Pediatrics, 17, 4, 490-501. Kushida C.A., Chang A., Gadkary C., Guilleminault C., Carrillo O., & Dement W.C. (2001).

Comparison of actigraphic, polysomnographic, and subjective assessment of sleep parameters in sleep-disordered patients. Sleep Med., 2, 389–396.

Page 67: Acute, Repeated-Dose and Residual Effects of Amphetamines

56

Kuypers K.P., & Ramaekers J.G. (2005). Transient memory impairment after acute dose of 75 mg 3.4-Methylene-dioxymethamphetamine. J Psychopharmacol., 19, 633–639.

Kuypers K.P., & Ramaekers J.G. (2007). Acute dose of MDMA (75 mg) impairs spatial memory

for location but leaves contextual processing of visuospatial information unaffected. Psychopharmacology, 189, 557-563.

Lamb R.J., & Henningfield J.E. (1994). Human d-amphetamine drug discrimination:

methamphetamine and hydromorphone. J Exp Anal Behav., 61, 169–80. Marrone G.F., Pardo J.S., Krauss R.M., & Hart C.L. (2010). Amphetamine analogs

methamphetamine and 3,4-methylenedioxymethamphetamine (MDMA) differentially affect speech. Psychopharmacology, 208, 169–177.

Martin, W.R., Sloan, J.W., Sapira, J.D., & Jasinski, D.R. (1971). Physiologic, subjective, and

behavioral effects of amphetamine, methamphetamine, ephedrine, phenmetrazine, and methylphenidate in man. Clin Pharmacol Ther., 12, 245–58.

McLeod D.R., Griffiths R.R., Bigelow G. E., & Yingling J. (1982). An automated version of the

digit symbol substitution test (DSST). Behav Res Methods Instrum., 14, 463–6. Michale-Robinet. (1994). Declassified: Beyond MK-Ultra: The Technology of Psychological

Conditioning in the 21st Century. Retrieved from http://www.cronacheterrestri.com/images/stories/mkultra/michael.robinett.mk_ultra.pdf

Miller, T. G. (1925). A Consideration of the Clinical Value of Ephedrine, Am. J. M. Sc., 170,

157. Miller T.P., Taylor J.L., & Tinklenberg J.R. (1988). A comparison of assessment techniques

measuring the effects of methylphenidate, secobarbital, diazepam and diphenhydramine in abstinent alcoholics. Neuropsychobiology, 19, 90–96.

Miller M.A., & Hughes A.L. (1994). Epidemiology of amphetamine use in the United States.

In: Cho AK, Segal DS (eds). Amphetamine and its Analogs. Academic Press: San Diego, CA. pp 439^157

Monroe, R.R., & Drell, H.J. (1947). Oral Use of Stimulants Obtained From Inhalers. JAMA, 135,

14, 909-915. Myerson, J.L. (1936). Effect of Benzedrine Sulfate on mood and fatigue in normal and neurotic

patients. Arch NeurPsych., 36, 4, 816-822. Myerson, J.L., & Damashek, W. (1936). Physiologic Effect of Benzedrine and Its Relationship to

Other Drugs of the Autonomic Nervous System. Am. J. Med. Sci., 192, 560–74. Nichols, D.E. (1994). Medicinal chemistry and structure-activity relationships. In: Cho AK,

Page 68: Acute, Repeated-Dose and Residual Effects of Amphetamines

57

Segal DS (eds). Amphetamine and its Analogs. Academic Press: San Diego, CA. Norman, J., & Shea, J.T. (1945). Acute hallucinosis as a complication of addiction to

amphetamine sulfate. New England Journal of Medicine, 233, 9, 270-271. Parascandola, J. (2010). Abel, Takamine, and the isolation of epinephrine. The Journal of

Allergy and Clinical Immunology, 125, 2, 514. Pareles, J. (2013, September 1). Dancing in the Eternal Present, Before Harsh Reality Intervened:

Electric Zoo Festival Cut Short by Two Deaths. The New York Times. Retrieved from http://www.nytimes.com

Parrott A.C., & Lasky J. (1998). Ecstasy (MDMA) effects upon mood and cognition: before,

during and after a Saturday night dance. Psychopharmacology, 139, 261–268. Peiró, A.M., Farré M., Roset P.N., Carbó M., Pujadas M., Torrens M., Camí J., & de la Torre R.

(2012). Human pharmacology of 3,4 methylenedioxymethamphetamine (MDMA, ecstasy) after repeated doses taken 2 h apart. Psychopharmacology Berl., 225, 883-93.

Perez-Reyes M., White W.R., McDonald S.A., Hicks R.E., Jeffcoat R.A., Hill J.M., & Cook C.E.

(1991). Clinical effects of daily methamphetamine administration. J Clin Neuropsychol., 14, 352-358.

Piness, G., Miller, H. & Alles, G. (1930) “Clinical Observations on Phenylethanolamine

Sulfate,” Journal of the American Medical Association, 94, 790–791. Prinzmetal, M. & Bloomberg, W. (1935). The use of Benzedrine for the treatment of narcolepsy.

J. Amer. Med. Ass.,105, 2051-2054. Randall, S., Johanson, C.E., Tancer M., & Roehrs, T. (2009). Effects of acute 3,4-

methylenedioxymethamphetamine on sleep and daytime sleepiness in MDMA users: a preliminary study. Sleep, 32, 1513–1519.

Rasmussen, N. (2006) Making the First Anti-Depressant: Amphetamine in American Medicine,

1929–1950,” Journal of the History of Medicine and Allied Sciences, 61, 288–323. Rasmussen, N. (2008). On Speed: The Many Lives of Amphetamine. New York: New York

University Press. Rasmussen, N. (2011) Medical science and the military: The Allies use of amphetamine during

World War II. Journal of Interdisciplinary History, 42, 205-233. Ricaurte, G.A., Schuster, C.R., & Seiden, L.S. (1980). Long-term effects of repeated

methylamphetamine administration on dopamine and serotonin neurons in the rat brain: a regional study. Brain Res, 193, 153-63.

Page 69: Acute, Repeated-Dose and Residual Effects of Amphetamines

58

Robinson, D. (November 3, 1967). New Westport Arrests Expected in Teen-Age Narcotics

Scandal, New York Times. Rosenthal, G. & Solomon, H.A. (1940). Benzedrine Sulfate in Obesity. Endocrinology, 26, 5, Sevak R.J., Stoops, W.W., Hays L.R., & Rush C.R. (2009). Discriminative stimulus and subject-

rated effects of methamphetamine, d-amphetamine, methylphenidate, and triazolam in methamphetamine-trained humans. J Pharmacol Exp Ther., 328, 1007–18.

Severo, R. (June 24, 1971). State Finds Drug Abuse is Rising. New York Times. Retrieved from

http://www.nytimes.com/ Simon S. L., Richardson K., Dacey J., Glynn S., Domier C. P., Rawson R. A. et al. (2002). A

comparison of patterns of methamphetamine and cocaine use. J Addict Dis., 21, 35–44. Substance Abuse and Mental Health Services Administration (SAMHSA) (2011). National

Household Survey on Drug Abuse: Summary of national findings. Office of Applied Studies, NHSDA Series H-44, HHS Publication No. (SMA) 12-4713. Rockville, MD.

Substance Abuse and Mental Health Services Administration, Center for Behavioral Health

Statistics and Quality. Treatment Episode Data Set (TEDS): 2001-2011. National Admissions to Substance Abuse Treatment Services. BHSIS Series S-65, HHS Publication No. (SMA) 13-4772. Rockville, MD: Substance Abuse and Mental Health Services Administration, 2013.

Substance Abuse and Mental Health Services Administration, Drug Abuse Warning Network,

2011: National Estimates of Drug-Related Emergency Department Visits. HHS Publication No. (SMA) 13-4760, DAWN Series D-39. Rockville, MD: Substance Abuse and Mental Health Services Administration, 2013.

Tancer M., & Johanson C.E. (2001). The subjective effects of MDMA and mCPP in moderate

MDMA users. Drug Alcohol Depend., 65, 97-101. Tancer M., & Johanson C.E. (2003). Reinforcing, subjective, and physiological effects of

MDMA in humans: a comparison with D-amphetamine and mCPP. Drug Alcohol Depend., 72, 33–44.

Tancer M., & Johanson C.E. (2007). The effects of fluoxetine on the subjective and

physiological effects of 3,4-methylenedioxymethamphetamine (MDMA) in humans. Psychopharmacology,189, 565-573.

Tidy, H.L. (1938). Discussion on Benzedrine: Uses and abuses. Proc. R. Soc. Med., 3, 2, 385-

398.

Page 70: Acute, Repeated-Dose and Residual Effects of Amphetamines

59

Ulrich A. (May 5, 2005). Hitler's drugged soldiers. Der Spiegel. Retrieved from

http://www.spiegel.de/international/the-nazi-death-machine-hitler-s-drugged-soldiers-a- 354606.html

United Nations Office of Drugs and Crime. (2012). World drug report 2012. Vienna: United

Nations. Retrieved from: http://www.unodc.org/documents/data-and analysis/WDR2012/WDR_2012_web_small.pdf

Ward, A.S., Haney M., Fischman M.W., & Foltin, R.W. (1997). Binge cocaine self-

administration by humans: smoked cocaine. Behav Pharmacol., 8, 736-744. Weisheit, R. & White, L.W (2009). Methamphetamine: It’s history, pharmacology and treatment.

Minnesota: Hazelden. Wesnes K., & Warburton D.M. (1983). Effects of smoking on rapid information processing

performance. Neuropsychobiology, 9, 223–229. Wilbur, D.L., MacLean, A.R. & Allen, E.V. (1937). Clinical Observations on the Effect of

Benzedrine Sulfate. J. Am. Med. Assoc., 109, 549–54. W.L.L (December 7, 1947). Notes on Science: Drug Hazards. New York Times. Retrieved from

http://www.nytimes.com/ Yokel, R.A., & Pickens, R. (1973). Self-administration of optical isomers of amphetamine and

methylamphetamine by rats. J Pharmacol Exp Ther. 187, 27-33. Young, D. & Scoville, W.B. (1938). Paranoid Psychoses in Narcolepsy and Possible Danger of

Benzedrine Treatment. Medical Clinics of North America, 22, 637–646. Zhou, R., & Pham, M.T. (2004). Promotion and prevention across mental accounts: when

financial products dictate consumers’ investment goals. Journal of Consumer Research, 31, 125-135.

Page 71: Acute, Repeated-Dose and Residual Effects of Amphetamines

60

Appendix A

Table 1.1 Early medical uses of amphetamines

Medical Use 1. Narcolepsy 2. Epilepsy 3. Fatigue 4. Depression 5. Schizophrenia 6. Alcoholism 7. Barbiturate Intoxication and Narcosis 8. Avertin Anesthesia 9. Morphine Addiction 10. Codeine Addiction 11. Caffeine Mania 12. Nicotinism 13. Disseminated Sclerosis 14. Myathenia Gravis 15. Myotonia 16. Electric Convulsion Therapy 17. Head Injuries 18. Infantile Cerebral Palsy 19. Problem Children and Enuresis 20. Migraine 21. Urticaria 22. Meniere’s Syndrome 23. Sea Sickness 24. Persistent Hiccup 25. Dysmenorrhoea 26. Ureteral Colic 27. Obesity 28. The Irritable Colon 29. X-Ray Visualization 30. Irradiation Sickness 31. “Restless Legs” 32. Carotid Sinus Syndrome 33. Hypotension 34. Heart Block 35. Analgesia 36. Malingering 37. Ophthalmoligical 38. Night Blindness 39. Libido

Source: Bett (1946)

Page 72: Acute, Repeated-Dose and Residual Effects of Amphetamines

61

Table 1.2 Summary of controlled substance schedules

Schedule Criteria Examples Schedule I a. High potential for abuse

b. No currently acceptable medical use in treatment in the U.S. c. Lack of accepted safety for use under medical supervision.

Heroin Marijuana MDMA (Ecstasy)

Schedule II a. High potential for abuse b. Currently accepted medical use c. Abuse may lead to severe psychological or physical

dependence.

Morphine Cocaine Methamphetamine

Schedule III a. Potential for abuse less than I and II b. Currently accepted medical use c. Abuse may lead to moderate physical dependence or high

psychological dependence.

Anabolic steroids Most barbituates Dronabinol (THC)

Schedule IV a. Low potential for abuse relative to III b. Currently accepted medical use c. Abuse may lead to limited physical dependence or

psychological dependence relative to III.

Alprazolam (Xanax) Fenfluramine Zolpidem (Ambien)

Source: Hart and Ksir (2011)

Figure 1.1 The effects of subcutaneous and oral amphetamine on cardiovascular measures in humans (Piness et al., 1930)

Page 73: Acute, Repeated-Dose and Residual Effects of Amphetamines

62

Figure 1.2 Benzedrine inhalers sold over-the-counter.

Figure 1.3 A Smith, Klein and French advertisement for Benzedrine Sulfate (e.g., amphetamine) in the American Journal of Psychiatry, March 1945, and California and Western Medicine, April 1945.

Page 74: Acute, Repeated-Dose and Residual Effects of Amphetamines

63

Figure 1.4 Smith, Klein and French advertisements for military use of Benzedrine inhaler in the Journal of the American Medical Association, Vol. 123, No. 10, 1943 and Vol. 124, No. 12., 1944.

Figure 1.5 A Burroughs Wellcome & Co. advertisement for Methedrine

Page 75: Acute, Repeated-Dose and Residual Effects of Amphetamines

64

Figure 1.6 Chemical structure of dextro- and levo-amphetamine (Prinzmetal and Alles, 1940)

Figure 1.7 A Burroughs Wellcome & Co. advertisement for Methedrine ampoules

Page 76: Acute, Repeated-Dose and Residual Effects of Amphetamines

65

Appendix B Table 2.1 Study design

Week Drug Monday Tuesday Wednesday Thursday Friday

1 MA (mg/70 kg) (50) (12)

2 AMPH (mg/70kg) (12) (50)

3 (0)

Table 2.2 d-amphetamine- and methamphetamine-related effects on task engagement ratings

Placebo 12 mg AMPH 12 mg MA 50 mg AMPH 50 mg MA Measure Mean (SEM) Mean (SEM) Mean (SEM) Mean (SEM) Mean (SEM)

Task engagement ratings (max = 9)

“Felt bad during task” 2.07 (0.47) *1.54 (0.10) *1.54 (0.10) *1.07 (0.07) *1.0 (0.00) “Felt good during task” 5.85 (0.56) 6.30 (0.62) *7.50 (0.40) *7.0 (0.50) *7.10 (0.50) “Felt happy during task”

5.46 (0.31) 6.07 (0.33) *6.50 (0.05) *6.70 (0.44) *6.7 (0.43)

* p < .05, significantly different from placebo. VAS: visual analog scale. DEQ: drug-effect questionnaire. SEM: standard error of the mean

Figure 2.1 “Prevention” focus pride ratings as a function of drug condition.

8

10

12

14

16

Pbo 12AMPH

12 MA

50AMPH

50 MA

RFQ: Prevention Score

Drug Condition (mg/70 kg)

Rat

ing

(Max

=25)

Page 77: Acute, Repeated-Dose and Residual Effects of Amphetamines

66

Figure 2.2 “Prevention” focus state ratings as a function of drug condition.

Figure 2.3 Engagement ratings on selected items as a function of drug condition.

8

10

12

14

16

Pbo 12AMPH

12 MA

50AMPH

50 MA

RFQ: Prevention Score

Drug Condition (mg/70 kg)

Rat

ing

(Max

=25)

Pbo 12AMPH

12 MA

50AMPH

50 MA

0

3

6

9

0

3

6

9

Pbo 12AMPH

12 MA

50AMPH

50 MA

50 MA

0

3

6

9

Pbo 12AMPH

12 MA

50AMPH

"Completely Focused onTask"

Drug Condition (mg/70 kg)

Rat

ing

(Max

=9)

"Involved in Task" "Motivated to get same drugdose"

Page 78: Acute, Repeated-Dose and Residual Effects of Amphetamines

67

Appendix C Table 3.1 Study Design Time Day 1 Day 2 Day 3 Day 4 Day 5 Day 6 Day 7 Day 8 Day

9 Day 10

0900 P MDMA1 P P P P MDMA2 P P P

2100 P MDMA1 MDMA1 P P P MDMA2 MDMA2 P P

Time Day 11 Day 12 Day 13 Day

14 Day 15

0900 P P P P P

0900 P P P P P

Table 3.2 Acute (Administration 1) MDMA-related effects on subjective-effect ratings

Placebo 50 mg MDMA 100 mg MDMA Measure Mean (SEM) Mean (SEM) F Value Mean (SEM) F Value

VAS ratings (max = 100)

Anxious 1.92 (1.66) 18.58 (9.82) 7.13 25.67 (10.30) *14.47 Blurred vision 3.83 (3.66) 16.58 (7.94) 88.84 42.50 (10.13) *§36.50 Can’t concentrate 20.08 (6.86) 41.50 (11.01) *8.12 45.58 (10.56) *11.51 Chills 10.70 (7.31) 33.80 (11.40) *11.93 50.50 (12.41) *§35.28 Confused 1.67 (1.67) 17.50 (8.87) *7.57 17.75 (7.0) *7.81 Content 58.83 (9.07) 66.74 (9.22) 0.89 79.92 (7.53) *6.33 Dizzy 4.83 (3.60) 18.33 (9.60) *9.01 22.67 (9.41) *15.72 Friendly 55.50 (6.69) 70.50 (5.05) *15.19 79.42 (6.86) *2.21 Heart pounding 6.33 (5.10) 32.41 (12.02) *17.96 39.08 (12.81) *28.32 High 8.33 (4.10) 61.83 (11.65) *63.24 85.83 (6.78) *§132.71 Noises seem loud 7.08 (5.80) 23.33 (10.11) *8.79 42.33 (12.45) *§41.36 Restless 10.58 (5.57) 20.67 (7.88) 2.06 31.58 (10.85) *8.96 Social 57.58 (5.52) 64.67 (7.90) 1.02 72.92 (9.0) *4.76 Stimulated 5.583 (3.05) 55.75 (12.93) *48.04 86.25 (6.56) *§124.21 Sweating 0.25 (0.25) 9.50 (6.48) 2.49 30.08 (12.62) *§25.89 Talkative 45.25 (6.16) 55.42 (7.67) 1.79 67.58 (11.77) *8.62

DEQ ratings

Bad drug effect 0.08 (0.08) 0.33 (0.14) *3.04 0.75 (0.37) *§21.63 Desire to take again 1.17 (0.47) 2.75 (0.31) *13.25 3.42 (0.29) *39.97 Like drug 0.33 (0.59) 2.58 (0.21) *32.93 3.25 (0.38) *61.84

* p < .05, significantly different from placebo. § p < .05, significantly different from 50 mg MDMA. VAS: visual analog scale. DEQ: drug-effect questionnaire. SEM: standard error of the mean

Page 79: Acute, Repeated-Dose and Residual Effects of Amphetamines

68

Table 3.3 MDMA-related effects on subjective-effect ratings (Administration 3) Placebo 50 mg MDMA 100 mg MDMA Measure Mean (SEM) Mean (SEM) F Value Mean (SEM) F Value

VAS ratings (max = 100)

Heart pounding 1.83 (1.83) 31.17 (9.13) *39.95 32.83 (11.45) *44.62 High 7.25 (3.87) 47.25 (8.76) *71.52 65.58 (8.75) *§152.12 Noises seem loud 10.33 (6.31) 19.75 (9.72) *5.65 26.17 (9.50) *15.96

DEQ ratings (max = 4)

Drug strength 0.50 (0.20) 2.25 (0.13) *66.38 2.33 (0.40) *72.85 Desire to take again 1.33 (0.51) 2.08 (0.33) *2.97 2.83 (0.41) *11.89 Good drug effect 0.42 (0.19) 2.25 (0.25) *49.67 2.42 (0.43) *59.11 Like drug 0.17 (0.56) 1.92 (0.42) *19.92 2.17 (0.71) *26.02

* p < .05, significantly different from placebo. § p < .05, significantly different from 50 mg MDMA. Table 3.4 Repeated-dose effects on subjective ratings

VAS ratings (max = 100)

Anxious 13.67 (7.62) 16.92 (9.92) 0.33 24.75 (9.14) 10.92 (6.22) *5.92 Heart pounding 30.67 (9.61) 31.17 (9.13) 0.01 43.50 (11.17) 32.83 (11.45) †5.28 Noises seem loud 16.58 (8.60) 19.75 (9.72) 0.64 39.75 (11.24) 26.17 (9.47) * 11.75

DEQ ratings (max = 4)

Drug strength 2.58 (0.15) 2.25 (0.13) 2.41 3.58 (0.19) 2.33 (0.40) *33.87 Good drug effect 2.75 (0.25) 2.25 (0.25) 3.69 3.42 (0.19) 2.42 (0.43) *14.78 Like drug 2.58 (0.19) 1.92 (0.42) 2.90 3.25 (0.25) 2.17 (0.71) *7.63

* p < .05, significantly different from 100 mg MDMA (Administration 1). † p = .055.

50 mg MDMA 100 mg MDMA Admin. 1

(Day 1 AM) Admin. 3 (Day 2 PM)

Admin. 1 (Day 1 AM)

Admin. 3 (Day 2 PM)

Measure Mean (SEM) Mean (SEM) F Value Mean (SEM) Mean (SEM) F Value

Page 80: Acute, Repeated-Dose and Residual Effects of Amphetamines

69

Figure 3.1. Selected acute (administration 1) cardiovascular and subjective-effect ratings as a function of drug dose and time. Error bars represent 1 SEM. Overlapping error bars were omitted for clarity.

Figure 3.2 Selected repeated dose cardiovascular and subjective-effects ratings as a function of drug condition and administration. Error bars represent 1 SEM. Overlapping error bars were omitted for clarity. Admin. 1= administration 1. Admin 3= administration 3.

Figure 3.3 Sleep measures on Day 2 (i.e., sleep period that began 3 hours after the second drug administration) as a function of drug dose.

Rat

ings

(max

=100

)

60 210 375

bpm

"Jittery"

100 mg

50 mg

0 mg"Good Drug Effect"

Time (min)

Heart Rate

75

50

25

00

110 4650 60 110 210 375 465

25

50

75

100100110

100

90

80

70

60500 100 200 300

0

Rati n

gs (m

ax=1

00)

bpm

Heart Rate

1 3

120

110

100

90

80

70

Drug Administration Number

"Good Drug Effect"

0 mg

50 mg

100 mg

100

80

60

40

20

01 13 3

80

60

40

20

0

Ratin

gs (m

ax=1

00)

bpm

Heart Rate

1 3

120

110

100

90

80

70

Drug Administration Number

"Good Drug Effect"

0 mg

50 mg

100 mg

100

80

60

40

20

01 13 3

80

60

40

20

0

"Jittery"

Tim

e (h

r)

Subjective Hours SleptObjective Hours Slept

Tim

e (m

in)

Pbo Pbo Pbo50mg50mg 50mg100 mg 100 mg 100 mg

8

6

0

2

4

0

2

4

6

8

Drug Condition

100

150

200

50

0

Sleep Onset Latency