Dissertation LSD Patrick Dolder 18.09.17

Download as pdf or txt
Download as pdf or txt
You are on page 1of 130

The Pharmacology of d-Lysergic Acid Diethylamide (LSD)

Inaugural dissertation

to

be awarded the degree of Dr. sc. med.


presented at
the Faculty of Medicine
of the University of Basel

by

Patrick Christian Dolder

from Basel, Switzerland

Basel, 2017

Originaldokument gespeichert auf dem Dokumentenserver der Universität Basel


edoc.unibas.ch

1
Approved by the Faculty of Medicine

On application of

Prof. Dr. med. Matthias Liechti


Prof. Dr. sc. nat. Katharina Rentsch
Prof. Dr. med. Stefan Borgwardt
Prof. Dr. rer. nat. Wolfgang Weinmann
Prof. Dr. med. Dr. pharm. Stephan Krähenbühl

Basel

Prof. Dr. med. Thomas Gasser


Dean

2
Acknowledgment

My doctorate was on the one side very hard and demanding, but on the other side
also a very joyful and instructive time. I had the pleasure to work in and with many
different teams and thereby met many people who not only helped me develop my
scientific skills, but also contributed to my personal development. Finally and
most important, I had the opportunity to build invaluable friendships.

First of all, I want to thank my two advisors Käthi and Matthias, for their
expertise, support, patience, and collaboration during my master thesis and following
doctorate. “It is the supreme art of the teacher to awaken joy in creative
expression and knowledge”.

I want to thank Felix who was my coworker in the LSD studies. We spent many hours
together during study sessions that lasted between 15 and 25 hours and brought us
to our limits. “A sorrow shared was a sorrow halved.”

With all my heart I want to thank my teammates Anna and Vizeli for our numerous
exhilarating days in the office, during the studies, and in our free time that we have
spent together. “A joy shared was a joy doubled.”

I want to extend my thanks to Petra and Claudia for their support and my
masterstudents Friede, Samuel, Toya, Raoul, and Laura for their work during the
studies. “Friends make the good times better and the hard times easier.”

Further I want to thank Orhan, Sophia, and all the lab mates from the Laboratory
Medicine for their knowledge and support during my analytical work.
“The only source of knowledge is experience.”

I want to thank Peter for his collaboration in our new study and his pioneer work with
LSD in general. I hope to learn from you in the next years.

3
“It is my great concern to separate psychedelics from the ongoing
debates about drugs, and to highlight the potential inherent to these
substances for self-awareness, as an adjunct in therapy, and for
fundamental research into the human mind.”

Albert Hofmann

“LSD is a catalyst or amplifier of mental processes. If properly used, it


could become something like the microscope or telescope of psychiatry.
Whether or not LSD research and therapy will return to society, the
discoveries that psychedelics made possible have revolutionary
implications for our understanding of the psyche, human nature, and the
nature of reality.”

Stanislav Grof

4
Content

Acknowledgment ........................................................................................................ 3

1. Overview .............................................................................................................. 6

2. Introduction .......................................................................................................... 7

2.1 Background ...................................................................................................... 7

2.2 The History of LSD ........................................................................................... 9

2.3 The Future of LSD or Aims of the PhD Project ............................................... 15

3. Publications ........................................................................................................ 17

3.1 Publication 1 ................................................................................................... 17

3.2 Publication 2 ................................................................................................... 26

3.3 Publication 3 ................................................................................................... 34

3.4 Publication 4 ................................................................................................... 48

3.5 Publication 5 ................................................................................................... 63

3.6 Publication 6 ................................................................................................... 69

3.7 Publication 7 ................................................................................................... 78

4. Discussion................................ ..........................................................................111

4.1 Pharmacokinetics................................ ...........................................................111

4.2 Pharmacodynamics ........................................................................................114

4.3 Pharmacokinetics - Pharmacodynamics .........................................................116

4.4 Emotion Recognition and Empathy.................................................................118

5. Summary and Outlook .......................................................................................119

References................................ ...............................................................................120

5
1. Overview

My PhD thesis consisted of two different scientific parts, each supervised by one of
my advisors, Prof. Dr. med. Matthias Liechti and Prof. Dr. sc. nat. Katharina Rentsch.

One part was to develop liquid chromatography tandem mass spectrometry (LC-
MS/MS) methods to measure d-lysergic acid diethylamide (LSD) and its main
metabolites in plasma, serum, and urine samples. We established the
pharmacokinetics of LSD and collected data from emergency toxicological cases.
Therefore we have developed and validated two analytical methods using LC-MS/MS
which resulted in several publications. All analytical work was performed in the
Toxicology Lab of the Laboratory Medicine at the University Hospital Basel under the
supervision of Prof. Dr. sc. nat. Katharina Rentsch.

The second part included planning, conduction, and analysis of clinical phase I trials
with LSD. We investigated the acute psychological and physiological effects of LSD
in healthy humans what resulted in several publication. These projects were
supervised by Prof. Dr. med. Matthias Liechti at the Department of Clinical
Pharmacology and Toxicology of the University Hospital Basel. One LSD study
included a functional magnetic resonance imaging (fMRI) assessment, to investigate
the neural correlates of altered states of consciousness and emotion processing
under the influence of LSD. The fMRI study was done in close collaboration with the
team of Prof. Dr. med. Stefan Borgwardt from the Department of Psychiatry of the
University of Basel.

6
2. Introduction

2.1 Background

Lysergic acid diethylamide (LSD) is a semisynthetic compound related to its


precursors lysergic acid and lysergic acid amide which are naturally occurring in
different fungi (e.g. claviceps purpurea) and plant seeds (e.g. argyreia nervosa). The
chemical structure of LSD is related to the endogenous neurotransmitter serotonin
and other psychedelic drugs such as psilocin, the active compound of the “magic
mushrooms” (e.g. psilocybe cubensis), and dimethyltryptamine, the active compound
of “ayahuasca” (Figure 1). The synthesis of LSD yields four stereoisomeric alkaloids,
d- and l-LSD, and d-iso-LSD and l-iso-LSD, whereof only d-LSD possesses the
powerful mind-altering effects in animals and humans (1-3). d-LSD is one of the most
potent substances, doses above 0.01 mg (10 µg) already produce measurable
effects, and from 40 µg upwards induce intense behavioral and perceptual alterations
(4, 5). LSD interacts with several brain receptors. Specifically, LSD binds to several
subtypes of the serotonin receptor (5-HT2A, 5-HT1A, 5-HT2C), has additional affinity for
dopamine D1 and D2 receptors (6-8), and indirectly alters glutamatergic
neurotransmission via the 5-HT2A receptor (9). The 5-HT2A receptor is also
considered the receptor that primarily mediates the hallucinogenic effects of LSD and
other serotonergic hallucinogens including psilocin and dimethyltryptamine (10-13).
Research with a hallucinogen like LSD always raises some safety concerns.
However, LSD possesses little if any abuse liability, is not self-administered by
animals, and there is no human LSD dependence syndrome (14). Repeated LSD
administration leads to pronounced tolerance to its psychological and physiological
effects in less than seven days (15, 16). Further, there is cross-tolerance after
repeated administration of psilocybin and other LSD derivatives in humans (17, 18).
The tolerance is transient and absent three days after discontinuation. Long-term use
in humans is not associated with any evidence of generalized brain damage related
to the number of LSD consumptions (19). The chance of precipitating a long-term
psychotic reaction is limited to subjects with a personal or familiar history of psychotic
disorders (20). Under controlled and supportive conditions, the LSD experience may
even have lasting positive effects on attitude and personality (21).

7
Dimethyltryptamine

Serotonin

d-lysergic acid diethylamide

Psilocin

Figure 1 gives the structure of d-lysergic acid diethylamide, the neurotransmitter


serotonin (embedded), and the hallucinogens dimethyltryptamine and psilocin.

8
2.2 The History of LSD

LSD was first synthesized in 1938 here in Basel, and its highly specific actions on the
brain and human consciousness were discovered by chance by Albert Hofmann. On
the 16th of April in 1943, he decided to resynthesize LSD to repeat tests at the
pharmacological department of Sandoz. He got contaminated by accident, and
suddenly felt a strong restlessness combined with a slight dizziness whereon he
interrupted his work and returned home. He described the following hours as a “not-
unpleasant intoxicated dreamlike state, with very stimulated imagination and
kaleidoscope-like play of colors” (22). On the 19th of April 1943 he decided to do a
self-experiment with 250 µg of the d-LSD tartrate salt. He described the following trip
as follows: “A demon had invaded me and had taken possession of my body, mind
and soul. I jumped up and screamed in order to free myself from him, but then sank
down again powerless on the sofa. A dreadful fear grasped me that I was becoming
insane. I was taken to another world, another place, another time. My body seemed
to me to be without sensation, lifeless, strange. Was I dying? Was this the transition?
Then, the horror softened and gave way to a feeling of fortune and gratitude, the
more normal perceptions and thoughts returned and my assurance increased that the
danger of insanity was conclusively past. Now I gradually began to enjoy the
unprecedented colors and plays of shapes that persisted behind my closed eyes. It
was particularly remarkable how every acoustic perception…became transformed
into optical perceptions. Exhausted I then slept and woke up the next morning with a
clear head, even though still somewhat tired physically. A sensation of well-being and
renewed life flowed through me.”(22) He wrote a report about his experience to his
seniors who repeated his self-experiment, although with lower doses.
From 1949-1966, d-LSD tartrate (LSD-25) was marketed by Sandoz under the brand
name Delysid®, and was mostly used in basic psychiatric research and
psychotherapy (9, 23-26). Its subjective psychotomimetic effects were compared to
those in patients with schizophrenia and led to its use as an experimental substance
for model psychosis (2, 27-29). It was thus provided to psychiatrists and researchers
with the purpose to study these psychotic phenomena and giving them the possibility
of gaining insight into the subjective character of mental disorders.

9
Soon, a potential therapeutic use was recognized and led to first therapeutic studies
at the Psychiatric University Hospital in Zurich (30). In the following years, beneficial
effects were documented in the treatment of alcoholism (31), anxiety associated with
terminal illness (26, 32, 33) and in the treatment of cluster headache (34).

In hand with the use in a therapeutic setting, LSD was also investigated in
social/group settings. Social cognition including emotion recognition and empathy
describes the ability to infer another’s thoughts, feelings, and intentions and is thus a
highly relevant topic not only for social interactions but especially for its use in a
psychotherapeutic setting. However, various studies examining social interactions
under the influence of LSD showed inconsistent results. This was not surprising,
since experiments were carried out in small groups (3 or 4 subjects), in different
populations (healthy, alcoholics, addicts, reformatory inmates or schizophrenics) and
with variable doses (25 – 200 µg) (35-39). Further, social effects were measured
using a variety of tools and included investigations of social perception (rating of
liking others in the group or being liked by them) (39), prosocial effects like increased
solidarity, tension release, and decreased antagonism (38). Subject’s social
interactions within the group settings were mostly set up around a specific task e.g.
discussing the solution of a human relation problem. The behavior of the group and
its individuals towards problem evaluation and decisions were recorded, and
categorized with the Bales Interaction Process Analysis (35, 36, 38). Thereby, the
social interactions were found to be altered in a dose dependent manner. For small
doses of LSD (25 - 50 µg) interaction was increased, whereas it leveled out on
moderate doses (75 – 100 µg), and finally decreased on high doses up to 200 µg
where subjects were less proactive in conversations (35-39). Changes in socio-
emotional behavior were observed in all groups but with different outcomes (35-39).
Alcoholics rose in positive emotional behavior whereas schizophrenics rose in
positive as well as negative behaviors (36). Overall LSD was reported as an effective
tool for increasing social interaction and gaining insight, making it thereby a useful
therapeutic agent. Besides the psychological and socio-emotional effects,
researchers were interested in the metabolism of LSD and its dose-relation to these
effects.

10
14
For the determination of the metabolic faith, C-labeled LSD was administered to
animals, and measurements of radioactivity were used for the quantification, which
was the method with the highest sensitivity and specificity during this research era
14
(40-44). Experiments with C labeled LSD in rats, mice, guinea pigs, and cats
showed a rapid uptake in to the blood, distribution among the organs where LSD
undergoes rapid chemical alteration, followed by a steady elimination in to the bile
and the small intestine (40-44). Enterohepatic re-absorption was found to be
negligible (44, 45). There was also a difference in metabolism across the various
species. In rats, mice, guinea pigs, and cats, the biliary/faecal excretion dominated
(40-44) whereas urinary excretion was dominant in rhesus monkeys (43). In rodents,
the major metabolites in bile and urine were found to be 13- and 14-hydroxy-LSD
glucuronides (43). In faeces the deconjugated forms, 13- and 14-hydroxy-LSD were
dominating, probably cleaved by gut bacteria. In rhesus monkeys, 13- and 14-
hydroxy-LSD accounted only for a minor part of the metabolites, but the major
metabolites could not be clearly identified. However, the formation of an additional
metabolite formed out of 2-oxo-LSD was described, and named “naphthostyril
compound” (43). This compound could be the precursor of the recently identified
major human metabolite, 2-oxo-3-hydroxy-LSD (46). A further identified metabolite
was de-ethyl-LSD, or lysergic-acid-monoethylamide (LAE). In vitro studies with liver
microsomes additionally yielded nor-LSD as potential metabolite, however it could
not be confirmed in-vivo (47). Out of the various LSD metabolites, 13-hydroxy-LSD
and LAE were found to be active in animals (43).

In humans, the metabolism of LSD is largely unknown and was less investigated
compared to the one in animals. The only two studies were done in the 1960s and
1970s. Single intravenous doses of 2 μg/kg in five healthy male subjects, and single
oral doses of 160 μg in 13 healthy male subjects were administered (48, 49). The
only small pharmacokinetic study was done with the results from the study by
Aghajanian et al. following the intravenous dose of 2 μg/kg and they proposed a
three-compartmental model (48, 50-52). Plasma concentrations were 6-7 ng/ml 30
min after intravenous administration, 4 - 6 ng/ml at 30 - 120 min, and approximately 1
ng/ml at 8 h. The elimination half-life of LSD was found to be 3 h (48, 50). This was
also the first time that the effects of LSD, represented by a score of impairment in
11
solving a mathematical task, were linked to the plasma concentration (48, 50-52).
The group of Upshall et al., which orally administered 160 μg of LSD, measured
plasma concentrations in a fasted state, following a light breakfast, or a full breakfast.
They observed a difference in plasma concentrations between men in a fasted state
and men who had a full breakfast, suggesting, that the amount and composition of
food has an effect on LSD plasma levels (49). The effects of two in-vivo identified
metabolites were also investigated in humans. Intramuscular application of up to
1’200 µg LAE led to strong psychological effects, comparable to those after oral
administration of 100 µg LSD (53). The effects were described faster in onset, but
lasted only up to 2.5 hours. In contrast, oral administration of 300 µg 2-oxo-LSD did
not induce any psychological effects (44).
Both human studies used fluorimetric assays for the measurements of their plasma
samples. They made use of LSD’s fluorescence and its UV-light catalyzed hydration
to the non-fluorescent lumi-LSD (10-Hydroxy-9,10-dihydro-LSD) (48, 49). However,
this method clearly lacked specificity (48, 49). Overall, human pharmacokinetic data
is very sparse and new technologies such as LC-MS/MS allow to measure substance
concentrations more precisely and also to further characterize metabolites. Indeed,
more recent in-vitro studies using human liver microsomes and analysis of human
urine samples have confirmed the presence of LAE, 2-oxo-LSD, 13- and 14-hydroxy-
LSD, and further identified nor-LSD, lysergic-acid-ethyl-2-hydroxyethylamide (LEO),
tri-oxo-LSD and 2-oxo-3-hydroxy-LSD as potential human metabolites (54, 55).
However, systematic information about their presence after controlled intake is still
missing.

12
Contrary to the unknown pharmacokinetic parameters, the pharmacodynamics,
including subjective and autonomic effects, were widely investigated. The dose range
for a typical LSD reaction was estimated to be 50 - 200 µg. A variety of different
doses and routes of application have been used in different study populations
including healthy subjects and patients (2, 14, 29). Therefore, descriptions of the
psychological effects were varying and depended on the investigated study
population, route of administration, dose of LSD, setting of the experiment, and
expectations of subjects and investigators. Generally, symptoms could be classified
among three characteristics: Somatic, perceptual, and psychologic effects.

In humans, LSD produces changes in perception, cognition, and emotions that last
for up to 12 hours (9, 14, 23). Similar to other serotonergic drugs, mild or moderate
anticipatory anxiety is common at the onset of the drug effect (56). During the time of
full effect, mood changes are very frequent, mostly towards positive mood states (2,
14). Perceptual changes include illusions, pseudo-hallucinations, intensified color
perception, synesthesia, and alterations in time perception (2, 14, 29). Alterations of
thinking may include imaginative thoughts, broader and unusual associations, re-
experiencing biographic memories, or mystical-type experiences (2, 14).
Furthermore, LSD acutely impairs psychomotoric function including coordination and
reaction time (2, 14, 29). Under controlled and supportive conditions, these
phenomena are mostly experienced in a positive way and may have lasting positive
effects on attitude and personality including greater appreciation of music, art, and
nature, greater tolerance of others, and increased creativity and imagination (21).
However, dysphoria, anxiety, and mild transient ideas of reference or paranoid
thinking may also occur in some subjects. However, they are mostly attributable to
uncontrolled conditions and can be readily managed with reassurance in a controlled
setting (2, 14, 23, 29).

13
These numerous investigations prove that there is considerable previous experience
with the use of LSD in humans, both with regard to research and clinical application.
Psychotherapists have used LSD in thousands of patients and thus made LSD one of
the most studied pharmacological substances with more than 4000 published reports
(9, 14, 24).
These scientific activities came to a halt as a result of the political concerns in
response to the increasing abuse of LSD starting in the end of the 1960s. Since the
1970s, clinical research using scheduled hallucinogenic substances like LSD has
been prohibited in most countries, with only a few exceptions. From 1988 to 1993,
LSD was legally used in Switzerland in LSD-assisted psychotherapy in 170 patients
with a wide range of clinical disorders (57). Further uses of LSD were re-recognized
and included its use in brain research (14), treatment of cluster headache (58, 59),
alcoholism (60), and as an adjunct to psychotherapy (61). A first placebo-controlled
pilot study using LSD in patients suffering from anxiety associated with advanced-
stage life threatening diseases showed a potential therapeutic value (61).
Although some of the earlier research produced promising results, it became also
clear that the initial studies conducted with LSD do not meet todays’ research
standards. For instance, no optimal methodological procedures, e.g. double-blind,
placebo-controlled studies, were used (14, 24). In addition, many of the techniques
used today were not available or not as developed at that time. Specifically,
comprehensive validated psychological test systems, sophisticated measures of
physiological and endocrine parameters, neuroimaging or analytical techniques were
unavailable or sparse. Hence, almost no scientific clinical pharmacological data on
LSD is available.

14
2.3 The Future of LSD or Aims of the PhD Project

Despite very intensive research during the 1950s to 1970s, there are still a lot of
research questions to be answered. First, most previous investigations do not meet
our present scientific and ethical standards and have therefore to be replicated. The
results from earlier studies were primarily observational and thus very subjective.
Second, technological progress allows us to use new and more modern approaches
such as imaging techniques. Third, LSD use is still very prevalent. Among young
adults (15- to 34-year-olds), lifetime prevalence of LSD use varies from 0.1% to 5.4%
in the EU (62) and up to 7% in the US population (63). Here in Basel, we registered
over 13 cases with an acute LSD intoxication on the emergency department of the
University Hospital Basel between October 2013 and September 2015 (64-66).
Because of this renewed interest and the lack of state of the art human
pharmacological data, we decided to conduct two placebo-controlled studies in
healthy subjects.
We aimed to better characterize the pharmacology of LSD using sensitive and
validated analytical and psychometric tools. One aim of our project was to develop
and validate LC-MS/MS methods to characterize the single-dose kinetics of LSD and
establish pharmacokinetic information which is important for the evaluation of clinical
study findings such as subjective effects, autonomic effects, and functional magnetic
resonance imaging results. Additionally, our methods were used to analyze samples
of LSD emergency toxicological cases on the emergency department of the
University Hospital Basel. The detailed analytical methods and the
development/validation procedures are described in detail in the following
publications 1 and 6 (65, 67). We also investigated the subjective effects, effects on
mood, perception, emotion recognition and empathy using sensitive, validated
psychometric tools. Investigation of the LSD effect on autonomic parameters included
assessment of blood pressure, heart rate, body temperature, and pupil diameter.
Further, we aimed to define the neuronal correlates of the effects of LSD using
functional magnetic resonance imaging (fMRI) techniques. Thereby, the studies also
provided basic data for the understanding of the role of the serotonin 5-HT2A receptor
in the regulation of mood in general and on emotion recognition and empathy.

15
Our studies generated objective, high-quality scientific information on the effects of
LSD in healthy subjects, data that cannot be obtained with observational studies.
Overall, our placebo-controlled studies using LSD in healthy subjects were primarily
descriptive in nature and with a focus on the tolerability and safety which is needed
for future projects. For both clinical studies we used a double-blind placebo-controlled
cross-over design with two treatment conditions (LSD and placebo). Thus, subjects
served as their own controls omitting within-subject variability and markedly
increased study power. The treatment order was counter-balanced with washout
periods of at least 7 days between the test days. The placebo condition mainly
served as a control condition for the subjective and somatic measures. Study 1 used
a dose of 200 µg LSD and placebo in 16 subjects (8 men, 8 women), and Study 2
used 100 µg LSD and placebo in 24 subjects (12 men, 12 women). Detailed
information about the inclusion and exclusion criteria for each study is explained in
the following publications 3 and 4 (68-71). All data were obtained with the same
psychometric questionnaires which were already used with other psychoactive and
stimulant drugs in our group (72-74) and by others (12, 75-77). Detailed description
of each test is part of the respective publications 3, 4, and 5 (69, 70, 78). Both
studies were conducted in accordance with the Declaration of Helsinki and were
approved by the local ethics committee. The administration of LSD to healthy
subjects was authorized by the Swiss Federal Office for Public Health (BAG). The
studies in the 1950s to 1970s have all used d-LSD tartrate (LSD-25, molecular weight
398), whereas we used d-LSD hydrate (molecular weight 323) what corresponds to a
higher dose of LSD-25 (+23%).

16
3. Publications

3.1 Publication 1

Development and validation of a rapid turboflow LC-MS/MS method


for the quantification of LSD and 2-oxo-3-hydroxy LSD in serum and
urine samples of emergency toxicological cases

Patrick C. Dolder1,2, Matthias E. Liechti2, Katharina M. Rentsch1

1 Laboratory Medicine, University Hospital Basel and University of Basel, Switzerland


2 Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of
Clinical Research, University Hospital Basel and University of Basel, Switzerland

17
Anal Bioanal Chem (2015) 407:1577–1584
DOI 10.1007/s00216-014-8388-1

RESEARCH PAPER

Development and validation of a rapid turboflow LC-MS/MS


method for the quantification of LSD and 2-oxo-3-hydroxy LSD
in serum and urine samples of emergency toxicological cases
Patrick C. Dolder & Matthias E. Liechti &
Katharina M. Rentsch

Received: 25 September 2014 / Revised: 24 November 2014 / Accepted: 2 December 2014 / Published online: 27 December 2014
# Springer-Verlag Berlin Heidelberg 2014

Abstract Lysergic acid diethylamide (LSD) is a widely used once with a LSD concentration of 1.25 ng/ml in urine. In
recreational drug. The aim of the present study is to develop a serum of two patients, the O-H-LSD concentration was deter-
quantitative turboflow LC-MS/MS method that can be used mined to be 0.99 and 0.45 ng/ml. In the urine of a third patient,
for rapid quantification of LSD and its main metabolite 2-oxo- the O-H-LSD concentration was 9.70 ng/ml.
3-hydroxy LSD (O-H-LSD) in serum and urine in emergency
toxicological cases without time-consuming extraction steps. Keywords LSD . O-H-LSD . LC-MS . Lysergic acid
The method was developed on an ion-trap LC-MS/MS instru- diethylamide . 2-Oxo-3-hydroxy LSD . Blood . Urine
ment coupled to a turbulent-flow extraction system. The val-
idation data showed no significant matrix effects and no ion
suppression has been observed in serum and urine. Mean Introduction
intraday accuracy and precision for LSD were 101 and
6.84 %, in urine samples and 97.40 and 5.89 % in serum, Lysergic acid diethylamide (LSD) is a psychoactive substance
respectively. For O-H-LSD, the respective values were 97.50 changing the state of consciousness and perception. Its psy-
and 4.99 % in urine and 107 and 4.70 % in serum. Mean chedelic effects made it popular as a recreational drug, espe-
interday accuracy and precision for LSD were 100 and 8.26 % cially in the early 1970s, but still today LSD is widely used
in urine and 101 and 6.56 % in serum, respectively. For O-H- [1]. Additionally, LSD (200 μg) has also recently been used in
LSD, the respective values were 101 and 8.11 % in urine and a clinical study as adjunct to psychotherapy [2]. LSD is one of
99.8 and 8.35 % in serum, respectively. The lower limit of the most potent psychotropic drugs and is used in low doses.
quantification for LSD was determined to be 0.1 ng/ml. LSD Typical recreational doses of LSD range from only 25 to
concentrations in serum were expected to be up to 8 ng/ml. 2- 200 μg with long-lasting, dose-dependent psychotropic ef-
Oxo-3-hydroxy LSD concentrations in urine up to 250 ng/ml. fects [1]. Hence, low blood and urine concentrations are
The new method was accurate and precise in the range of posing a challenge to all analytical methods.
expected serum and urine concentrations in patients with a LSD can only be detected in blood up to 8 h after admin-
suspected LSD intoxication. Until now, the method has been istration due to serum concentrations in the low nanogram per
applied in five cases with suspected LSD intoxication where milliliter range. 2-Oxo-3-hydroxy LSD (O-H-LSD) is the
the intake of the drug has been verified four times with LSD main metabolite present in urine at concentrations 16–34
concentrations in serum in the range of 1.80–14.70 ng/ml and times higher than LSD [3, 4]. To our knowledge, O-H-LSD
has only been detected once in blood in a postmortem case [5].
P. C. Dolder : K. M. Rentsch (*) According to Li et al. and Klette et al. LSD and O-H-LSD
Laboratory Medicine, University Hospital, Petersgraben 4,
4031 Basel, Switzerland
were regarded as stable under storage conditions of −20 °C [6,
e-mail: [email protected] 7].
Most published methods for LSD detection use either GC-
P. C. Dolder : M. E. Liechti MS or LC-MS/MS with a single-stage quadrupole [4, 5,
Division of Clinical Pharmacology and Toxicology, Department of
Biomedicine and Department of Clinical Research, University
8–12]. The aim of the present study was to develop a
Hospital and University of Basel, Hebelstrasse 20, 4031 Basel, turboflow LC-MS/MS method with the purpose of rapid
Switzerland quantification of LSD and its main metabolite in serum and

18
1578 P.C. Dolder et al.

urine in emergency toxicological cases without time- For the instrument control, the corresponding software
consuming extraction steps. package consisting of LTQ (v.2.6) for ion detection, Xcalibur
The method was developed using an ion-trap LC-MS/MS (v.2.1.0) for method programming, and LC-Quan (v.2.6.1) for
instrument in selected reaction monitoring (SRM) mode after quantification (all Thermo Scientific, Basel, Switzerland) was
atmospheric pressure ionization (APCI) for the quantification used.
of LSD and O-H-LSD in urine and serum samples. Poch et al.
used a similar APCI LC-MS/MS ion-trap instrument, but
mainly for the detection of O-H-LSD [3]. LC method
Favretto et al. improved the method, but switched to
electrospray ionization for suitable analysis of LSD and O- The method was based on a previously published method
H-LSD in blood, urine, and vitreous humor [13]. Our method [16]. Four mobile phases were used in gradient mode. For
was established and successfully applied in five emergency extraction, loading B consisted of 10 mM ammonium carbon-
toxicological cases with a suspected LSD intoxication. Addi- ate in water; eluting A was 5 mM ammonium acetate in water
tionally, the method will be used for the analysis of both blood containing 0.10 % formic acid and eluting B 5 mM ammoni-
and urine samples from a double-blind, placebo-controlled um acetate in methanol containing 0.50 % formic acid,
clinical trial. respectively.
Loading B was used as alkaline loading buffer, eluting A
and B for chromatographic separation. Loading and Eluting C
(acetonitrile /acetone/2-propanol, 1:1:1 (V/V/V)) were used to
Materials and methods clean the extracting and the analytical columns.
The gradient system with a total run time of 12 min is
Chemicals and reagents depicted in Table 1. Under the following gradient conditions,
LSD and LSD-d3 showed a retention time of 7.63 min, while
HPLC-grade purity acetonitrile, acetone, methanol, 2- O-H-LSD had a retention time of 6.34 min.
propanol, formic acid, and acetic acid were all purchased from
Merck (Darmstadt, Germany). Ammonium acetate and am-
monium carbonate were obtained in HPLC grade from Merck MS conditions
(Darmstadt, Germany). Distilled water was obtained from an
in-house installed purifier (ELGA, Bucks, UK). For the quantification of LSD and its metabolite, APCI was
Drug-free serum lyophilisate and urine negative control as used as the ionization source in positive ion mode. Discharge
blank matrices were obtained from Bio Rad Laboratories current and discharge voltage were set to 5 μA and 4.2 kV,
(Irvine, CA, USA). LSD and LSD-d3 were obtained from respectively. The vaporizer temperature was optimized to
Lipomed (Arlesheim, Switzerland) and 2-oxo-3-hydroxy 452 °C whereas sheath and auxiliary gas provided best results
LSD (O-H-LSD) from Cerilliant (Round Rock, TX, USA). with flow rates of 40 and 20 arbitrary units (AU). The capil-
lary temperature was set to 275 °C.
LSD and O-H-LSD were quantified using single reaction
LC-MS analysis monitoring (SRM) of the corresponding mass transitions
(LSD m/z 324.6→223.23, O-H-LSD m/z 356.33→338.33,
Equipment LSD-d3 m/z 327.21→226.2). The system was tuned and
optimized for the detection of LSD.
The sample extraction system (Transcend TLX1 HPLC, Ther-
mo Scientific, Basel, Switzerland) consisted of a Thermo PAL
autosampler and two Accela 600 pumps as eluting and load- Standard solutions
ing pumps. The autosampler and the sample extraction system
were all controlled by Aria software (version 1.6.3) from LSD and LSD-d3 were bought as 1 mg/ml reference standards
Thermo Scientific (Basel, Switzerland). A cyclone and a in acetonitrile, while O-H-LSD as 0.1 mg/ml reference stan-
C18XL turboflow column (Thermo Scientific, Basel Switzer- dard in acetonitrile. Stock solutions in acetonitrile containing
land) for extraction, and a 3 μm Betasil Phenyl/Hexyl column 100,000 ng/ml LSD, LSD-d3, or 10,000 ng/ml O-H-LSD,
(Thermo Scientific, Basel, Switzerland) for chromatographic respectively, were prepared in duplicate and stored at −20 °C
separation were used. in order to have different sets for quality control (QC) and
The online extraction system was coupled to a LTQ XL calibration samples, respectively. Working solutions of each
mass spectrometer from Thermo Scientific (Basel, Switzer- analyte at 1000, 100, 10, and 1 ng/ml in water were used for
land) using atmospheric pressure ionization (APCI), due to its the preparation of QC and calibration samples as well as for
performance regarding matrix effects [14, 15]. matrix and selectivity experiments.

19
A rapid LC-MS/MS method for the quantification of LSD 1579

Table 1 Detailed extraction and


analytical separation steps of the Time (min) Extraction Analytical separation
liquid chromatography method
Flow (μl/min) %A %B %C Flow (μl/min) %A %B %C

0.00 2 – 100 – 0.30 99 1 –


0.83 0.50 – 100 – 0.30 99 1 –
0.92 0.50 – – 100 0.30 80 20 –
1.58 0.05 – – 100 0.30 55 45 –
2.03 0.50 – – 100 0.30 40 60 –
4.03 0.03 – – 100 0.30 2 98 –
9.03 0.01 – – 100 0.50 2 98 –
11.03 2. – – 100 0.50 2 98 –
11.37 2 – – 100 0.50 – – 100
11.70 2 – 100 – 0.50 – 100 –
12.20 2 – 100 – 0.50 99 1 –
12.53 2 – 100 – 0.30 99 1 –

Sample preparation Selectivity

To 100 μl of serum, 100 μl acetonitrile for protein precipita- Following the FDA validation guidelines [18], six urine and
tion and 10 μl of a LSD-d3 internal standard solution six serum samples from different patients and healthy volun-
(100 ng/ml) were added. An identical volume of urine was teers were collected and analyzed to establish selectivity and
diluted with 50 μl of an ammonium acetate buffer (50 mM, check for unwanted interferences within both matrices.
pH 4) and 10 μl of the internal standard solution. The samples
were then vigorously vortexed, centrifuged for 10 min at Matrix effects and recovery
13,200g and the supernatant afterwards transferred into
autosampler vials. Matrix effects, recovery, and process efficiency were mea-
sured and calculated according to Matuszewski et al. [19].
Matrix effects in urine and serum were calculated as ratio of
Calibration the peak area before extraction and divided by the peak area
after extraction. In contrast to Matuszewski et al., the extrac-
Calibration curve in serum was realized by spiking serum tion step consisted of simple protein precipitation as bypassing
samples with LSD and O-H-LSD to concentrations of 0.10, the extraction step on our ion-trap system was not possible.
0.25, 0.50, 0.75, 1, 2.50, 5, 7.50, and 10 ng/ml plus a blank Six serum and six urine samples were spiked once with LSD
(matrix only) and zero sample (matrix plus internal standard). and O-H-LSD before and after extraction. The peak areas of
The highest calibration point in serum was adopted from the the spiked samples were then compared with the area of the
maximum plasma concentration out of available i.v. kinetic spiked mobile phase. Urine samples were spiked to 25 ng/ml
data [17]. LSD resp. 250 ng/ml O-H-LSD, serum samples to 10 ng/ml
The calibration curve in urine was realized by spiking each. Recovery values were calculated as areas of standards
urine samples with O-H-LSD to concentrations of 1.50, 5, spiked before extraction divided by the areas of standards
10, 25, 50, 100, 125, 250, and 500 ng/ml. LSD concen- spiked after extraction. The process efficiency was also
trations were 0.10, 0.50, 1, 2, 5, 10, 12, 25, and 50 ng/ml, adopted from Matuszewsky et al. and calculated as ratio
respectively. The highest calibrator in urine was adopted between the area of the standard spiked before extraction,
from published data containing various analyzed urine and the areas of the standard in neat solution.
samples [4].
Both calibration curves were fitted linearly using a Limit of quantification
weighting factor (1/x2).
In order to demonstrate accuracy and precision of the Drug-free serum and urine samples were spiked with dif-
method, five QC’s in urine and six QC’s in serum were used ferent concentrations of LSD and O-H-LSD for the deter-
with every run. The concentrations of the QC samples can be mination of the lower limit of quantification (LLOQ). The
seen in Tables 2 and 3. parent substance and metabolite ratio was determined 1:1

20
1580 P.C. Dolder et al.

Table 2 Intraday precision and accuracy data of LSD and 2-oxo-3-hydroxy LSD measured in serum and urine at different concentrations

Weighed-in concentration [ng/ml] Measured concentration [ng/ml] Mean precision SD [%] Mean accuracy±SD [%]

Serum Urine Serum Urine Serum (n=6) Urine (n=6) Serum (n=6) Urine (n=6)

LSD 0.10 0.10 0.098±0.006 0.106±0.007 6.3 6.5 98.40±4.8 106±7.7


0.40 0.25 0.38±0.03 0.28±0.03 6.6 12.3 96.20±6.5 112±13.1
0.80 0.60 0.82±0.03 0.53±0.03 4.6 5.0 103±6.3 88.80±4.1
4 3.30 3.92±0.22 3.32±0.20 5.7 6.1 97.80±4.7 101±7.0
8 33 7.52±0.49 31.70±1.39 6.6 4.4 93.9±5.8 96.0±4.1
10 9.53±0.53 5.5 95.3±5.7
O-H-LSD 0.10 1.50 0.104±0.008 1.45±0.05 8.0 3.6 104±8.3 96.4±3.2
0.40 2.50 0.44±0.02 2.20±0.16 3.8 7.2 110±4.2 88.20±6.5
0.80 6 0.88±0.02 6.25±0.07 2.5 1.2 110±2.8 104±4.7
4 33 4.04±0.38 33.90±2.5 9.5 7.3 101±9.6 103±8.2
8 333 8.20±0.28 321±18 3.4 5.7 102±3.5 96.20±6.0
10 11.29±0.11 0.9 113±1.1

LSD lysergic acid diethylamide, O-H-LSD 2-oxo-3-hydroxy lysergic acid diethylamide

in serum and assumed 1:10 in urine samples [4]. The Reproducibility


LLOQ concentrations had to give a response at least five
times greater than the blank. Additionally, precision had to According to the FDA guidelines, a minimum of five deter-
be <20 % and the accuracy between 80 and 120 % using at minations per concentration are recommended for determina-
least five determinations per matrix and concentration. tion of precision and accuracy [18].
The reproducibility of quantification was determined by
measuring serum (n=6) and urine (n=5) quality controls (QC)
Carryover once on 1 day (intraday precision and accuracy) and on six
different days (interday precision and accuracy). All values
Carryover was determined by quantification of different had to fulfill the criteria of a variation coefficient (CV) below
blanks, running between patient samples, calibrations, and 15 %, resp. below 20 % at the LLOQ and accuracy between
quality controls. 80 and 120 %. For serum, six quality controls from LLOQ to

Table 3 Interday precision and accuracy data of LSD and 2-oxo-3-hydroxy LSD measured in serum and urine at different concentrations

Weighed-in concentration [ng/ml] Measured concentration [ng/ml] Mean precision SD [%] Mean accuracy±SD [%]

Serum Urine Serum Urine Serum (n=6) Urine (n=6) Serum (n=6) Urine (n=6)

LSD 0.10 0.10 0.11±0.01 0.10±0.02 4.60 15.00 110±5.10 104±14.60


0.40 0.25 0.39±0.02 0.26±0.02 4.20 8.80 97.5±4.10 105±9.30
0.80 0.60 0.82±0.07 0.55±0.02 8.50 4.00 103±8.70 91.1±3.60
4 3.30 3.97±0.34 3.32±0.22 8.60 6.70 99.2±8.60 101±6.50
8 33 7.41±0.59 32.8±2.3 7.90 6.90 92.7±7.30 99.3±6.30
10 10.1±0.55 5.50 101±5.50
O-H-LSD 0.10 1.50 0.10±0.08 1.58±0.19 8.10 12.50 105±8.43 105±13.10
0.40 2.50 0.39±0.03 2.64±0.35 8.40 13.40 98.3±8.20 105±14.10
0.80 6 0.79±0.08 5.56±0.16 9.80 3.00 98.5±9.70 92.6±2.70
4 33 3.79±0.35 34.8±2.2 9.20 6.60 94.8±8.70 105±6.40
8 333 8.14±0.58 327±16.8 7.20 5.10 102±7.30 98.3±5.00
10 – 10.1±0.76 7.60 – 101±7.60 –

LSD lysergic acid diethylamide, O-H-LSD 2-oxo-3-hydroxy lysergic acid diethylamide

21
A rapid LC-MS/MS method for the quantification of LSD 1581

the highest calibrator (0.10, 0.40, 0.80, 4, 8, 10 ng/ml) were Reproducibility


measured once a day. For validation in urine, five QCs from
1.5 to 333 ng/ml were used. Calibration curves in urine were linear for both substances,
LSD and O-H-LSD with R2 greater than 0.98. Mean intraday
accuracy and precision in serum were 97.40 resp. 5.89 % for
LSD and 107 resp. 4.70 % for O-H-LSD (see Table 2). Mean
interday accuracy and precision for LSD and O-H-LSD were
Results 101 resp. 6.56 % and 99.80 resp. 8.35 %, respectively (see
Table 3).
Selectivity

None of the blank urine or serum samples showed any inter- Linearity
ference within the measured mass range and time frame.
LSD and O-H-LSD calibration curves in serum were linear
over the range from 0.10 to 10 ng/ml with a mean correlation
Matrix effects and recovery coefficient (R2) of 99.86 %. The calibration curves of the
mean values are shown in Fig. 1. Error bars indicate the
The matrix effects in urine were 138 % for LSD and 122 % for standard error of the mean.
O-H-LSD. Recovery in urine was calculated to be 90.00 and Calibration curves of LSD and O-H-LSD in urine were
87.80 %, respectively. Process efficiency in urine was 124 % linear over the concentration range from 1.50 ng/ml to
for LSD and 107 % for O-H-LSD. Serum showed higher 333 ng/ml. R2 was found to be 99.93 %. The detailed calibra-
matrix effects with 128 % for LSD and 78.70 % for O-H- tion curve is shown in Fig. 2.
LSD. Recovery in serum was 64.00 % for LSD and 32.00 %,
for O-H-LSD. The process efficiencies in serum were calcu- Toxicological cases
lated to be 128 % for LSD and 79 % for O-H-LSD. No ion
suppression was found for LSD or O-H-LSD in serum and In the period from January to September 2014, five patients
urine, but as mentioned by Johansen and Jensen [10] LSD-d3 were admitted to the emergency department (ED) of the
would correct for any ion suppression. In various negative University Hospital Basel with suspected LSD intoxication.
samples, small LSD concentrations below the LLOQ could be In all five cases, LSD consumption could be confirmed.
identified which derived from the deuterated internal standard. Routinely, a LC-MS/MS method screening over 700 sub-
Following these findings, LSD-d3 was measured ten times at stances in serum was run to detect the intake of other medi-
different concentrations. The working solution of the standard cation and designer drugs.
(100 ng/ml) contained 0.12 % undeuterated LSD. This impu- As a summary, all in vivo measured concentrations in the
rity in the peak area of LSD was subtracted from all calibra- matrices available from the emergency department can be
tors, quality controls, and unknown samples. found in Table 4.

Lower limits of quantification Case 1

The lowest accurate and precisely measurable concentration A 17-year-old girl was brought to the ED with acute confusion
was 0.10 ng/ml and thereby determined as LLOQ for LSD and and loss of sense of time and orientation. She admitted con-
O-H-LSD in serum. In urine samples, the LLOQ was deter- sumption of two sugar cubes and one blot with LSD (estimat-
mined at 0.10 ng/ml for LSD and 1.50 ng/ml for O-H-LSD. ed dose, 750 μg). A plasma sample for drug screening was
taken approximately 3 h after ingestion. The patient was under
chronic treatment with trazodone for depression. An addition-
Carryover al LC-MS/MS screen in serum showed the presence of THC
and trazodone. Quantification of LSD revealed a level of
No carryover was found for LSD and O-H-LSD in serum 14.70 ng/ml and a quantifiable O-H-LSD level of 0.99 ng/ml
samples. In contrast, a slight carryover (0.10 %) was found in serum. The only other published case where O-H-LSD
for O-H-LSD in urine samples following the highest QC could be detected in blood so far, was in a reanalyzed fatal
(333 ng/ml) and the highest calibration (500 ng/ml) in urine. case 10 years after collection [5]. Figure 3 shows the chro-
As a consequence, a second consecutive blank was inserted matogram of LSD, LSD-d3 and O-H-LSD in the serum of this
between and the carryover was reduced to 0.01 %. patient.

22
1582 P.C. Dolder et al.

Fig. 1 Shows the mean 12


calibration curve of LSD in serum 11
from the validation
measurements. The determination 10
coefficient was 0.9995 9

0
0 1 2 3 4 5 6 7 8 9 10 11

Case 2 about the time-point of the LSD ingestion was available from
anamnesis. Serum analysis showed an LSD concentration of
A 17-year-old male was brought by the ambulance to the ED 6.10 ng/ml and an O-H-LSD concentration of 0.45 ng/ml. An
with thoracic pressure, restlessness, and dyspnea. He admitted additional LC-MS/MS screening revealed the presence of
the intake of one sugar cube with LSD (estimated dose THC, cocaine, and amphetamine.
250 μg) at 8 p.m. with concomitant consumption of cannabis.
He reported onset of the symptoms at 10 p.m., 2 h post-
consumption. In the emergency department, the patient was Case 4
treated with lorazepam and acetaminophen. Serum analysis
revealed a LSD concentration of 1.80 ng/ml in a blood sample A 45-year-old male presented himself to the ED with
taken at 11 p.m. agitation, disorientation, and intense visual hallucinations.
He was partying for 2 days and consumed alcohol, LSD,
Case 3 cocaine, and cannabis. The time-point of the LSD intake
was not reported. The LC-MS/MS screening confirmed the
A 21-year-old male was admitted to the ED by ambulance and intake of THC and cocaine. Quantification of the serum
the police because of aggressive and uncooperative behavior LSD level detected 4.10 ng/ml LSD, but no quantifiable O-
after consumption of an alleged LSD blot. No information H-LSD.

Fig. 2 Shows the mean 550


calibration curve of 2-oxo-3-
500
Mean of measured 2-oxo-3-hydroxy-LSD

hydroxy LSD in urine from the


validation measurements. The 450
determination coefficient was
0.9993 400
concentrations (ng/mL)

350

300

250

200

150

100

50

0
0 50 100 150 200 250 300 350 400 450 500
Concentration of calibrators (ng/ml)

23
A rapid LC-MS/MS method for the quantification of LSD 1583

Table 4 Measured concentration of LSD and 2-oxo-3-hydroxy LSD in serum and/or urine in different patients

Patient 1 Patient 2 Patient 3 Patient 4 Patient 5

Serum LSD 14.70 ng/ml 1.80 ng/ml 6.10 ng/ml 4.10 ng/ml N/A
Serum O-H-LSD 0.99 ng/ml <LLOQ 0.45 ng/ml <LLOQ N/A
Urine LSD N/A N/A N/A N/A 1.30 ng/ml
Urine O-H-LSD N/A N/A N/A N/A 9.70 ng/ml

N/A matrix was not available from the emergency department; <LLOQ value was below the lower limit of quantification

Case 5 clean peak separation and flushing the columns to minimize


carryover.
A 36-year-old male presented himself to the ED with tactile Our purpose was to establish a fast and reliable method
and visual hallucinations after consumption of an alcoholic for application in emergency toxicological cases where
beverage in a club. He suspected someone to have mixed time is crucial. With a short method run of 12 min and
some drugs in his drink. A screening for LSD in urine revealed minimum sample preparation, results will be more quickly
1.30 ng/ml LSD and 9.70 ng/ml O-H-LSD, respectively. An available so that a fast diagnosis is possible. The method
additional LC-MS/MS screening in urine confirmed the pres- was applied in five toxicology cases where consumption of
ence of THC. No time-point of the drink consumption or start LSD could be confirmed four times in serum and once in
of the LSD effect was reported. urine.
Due to the fast method and obviation of purification steps, a
slight loss in sensitivity was accepted. LLOQ and LOD in
serum were hence higher than in other comparable methods
[5, 8–10, 13]. Some showed LOQ’s as low as 0.02 ng/ml for
Discussion and conclusion LSD but needed sample preparation and a longer run time [5].
In contrast, our method was mainly developed to rapidly
The development of a sensitive method for the measurement detect levels of LSD that occur during acute intoxication.
of LSD and its metabolite is an analytical challenge due to its The range of expected LSD concentrations in serum was
low concentrations in serum and urine. difficult to determine because only few pharmacokinetic data
Purification procedures with solid-phase or liquid-liquid is available. In fact, only one pharmacokinetic study with
extraction can certainly lead to better sensitivity of the LC- controlled administration of LSD exists. In this study, peak
MS/MS method, but form a time-consuming procedure [5]. plasma concentrations of LSD were 4–6 ng/ml 1–2 h after
The short run time of 12 min was mainly given by retention administration of LSD (intravenously at 2 μg/kg) [17]. There-
times of LSD, LSD-d3, and O-H-LSD. The additional time fore, we chose 10 ng/ml as highest calibrator to cover typically
following the LSD and LSD-d3 peak was necessary to ensure used oral doses of LSD (100–400 μg) [1]. However, one case

Fig. 3 Chromatogram and the respective structural formulas of LSD, LSD-d3, and 2-oxo-3-hydroxy LSD in the serum sample of patient 1

24
1584 P.C. Dolder et al.

was found with a LSD concentration of 14 ng/ml in plasma hydroxy LSD under various storage conditions. J Anal Toxicol
26(4):193–200
among the intoxication cases presented here.
7. Li Z, McNally AJ, Wang H, Salamone SJ (1998) Stability study of
This sample had to be diluted (1:1 with distilled water) in LSD under various storage conditions. J Anal Toxicol 22(6):520–525
order to determine the correct result. Expected urine concen- 8. Berg T, Jorgenrud B, Strand DH (2013) Determination of
trations and the calibration range were established considering buprenorphine, fentanyl and LSD in whole blood by UPLC-MS-
MS. J Anal Toxicol 37(3):159–165
already published data [3, 4]. Our method fulfilled all criteria
9. Canezin J, Cailleux A, Turcant A, Le Bouil A, Harry P, Allain P
for measurement of emergency toxicological cases. All four (2001) Determination of LSD and its metabolites in human biological
cases showed concentrations of LSD in serum in the range of fluids by high-performance liquid chromatography with electrospray
1.80–14.70 ng/ml. Additionally, to our knowledge, for the tandem mass spectrometry. J Chromatogr B Biomed Sci Appl 765(1):
15–27
first time, we describe the quantification of O-H-LSD in two
10. Johansen SS, Jensen JL (2005) Liquid chromatography-tandem mass
patients in a concentration well above the LLOQ of our spectrometry determination of LSD, ISO-LSD, and the main metab-
method. olite 2-oxo-3-hydroxy-LSD in forensic samples and application in a
forensic case. J Chromatogr B Anal Technol Biomed Life Sci 825(1):
Acknowledgments The work was supported by the Swiss Center for 21–28
Applied Human Toxicology (to M.E.L.). 11. Musshoff F, Daldrup T (1997) Gas chromatographic/mass spectro-
metric determination of lysergic acid diethylamide (LSD) in serum
samples. Forensic Sci Int 88(2):133–140
12. Nelson CC, Foltz RL (1992) Determination of lysergic acid
References diethylamide (LSD), iso-LSD, and N-demethyl-LSD in body fluids
by gas chromatography/tandem mass spectrometry. Anal Chem
64(14):1578–1585
1. Passie T, Halpern JH, Stichtenoth DO, Emrich HM, Hintzen A 13. Favretto D, Frison G, Maietti S, Ferrara SD (2007) LC-ESI-MS/MS
(2008) The pharmacology of lysergic acid diethylamide: a review. on an ion trap for the determination of LSD, iso-LSD, nor-LSD and
CNS Neurosci Ther 14(4):295–314 2-oxo-3-hydroxy-LSD in blood, urine and vitreous humor. Int J Legal
2. Gasser P, Holstein D, Michel Y, Doblin R, Yazar-Klosinski B, Passie Med 121(4):259–265
T, Brenneisen R (2014) Safety and efficacy of lysergic acid 14. Caban M, Migowska N, Stepnowski P, Kwiatkowski M, Kumirska J
diethylamide-assisted psychotherapy for anxiety associated with (2012) Matrix effects and recovery calculations in analyses of phar-
life-threatening diseases. J Nerv Ment Dis 202(7):513–520 maceuticals based on the determination of β-blockers and β-agonists
3. Poch GK, Klette KL, Anderson C (2000) The quantitation of 2-oxo- in environmental samples. J Chromatogr A 1258:117–127
3-hydroxy lysergic acid diethylamide (O-H-LSD) in human urine 15. Dams R, Huestis MA, Lambert WE, Murphy CM (2003) Matrix
specimens, a metabolite of LSD: comparative analysis using liquid effect in bio-analysis of illicit drugs with LC-MS/MS: influence of
chromatography-selected ion monitoring mass spectrometry and liq- ionization type, sample preparation, and biofluid. J Am Soc Mass
uid chromatography-ion trap mass spectrometry. J Anal Toxicol Spectrom 14(11):1290–1294
24(3):170–179 16. Mueller DM, Duretz B, Espourteille FA, Rentsch KM (2011)
4. Poch GK, Klette KL, Hallare DA, Manglicmot MG, Czarny RJ, Development of a fully automated toxicological LC-MS(n) screening
McWhorter LK, Anderson CJ (1999) Detection of metabolites of system in urine using online extraction with turbulent flow chroma-
lysergic acid diethylamide (LSD) in human urine specimens: 2-oxo- tography. Anal Bioanal Chem 400(1):89–100
3-hydroxy-LSD, a prevalent metabolite of LSD. J Chromatogr B 17. Aghajanian GK, Bing OH (1964) Persistence of lysergic acid
Biomed Sci Appl 724(1):23–33 diethylamide in plasma of human subjects. Clin Pharmacol Ther 5:
5. Chung A, Hudson J, McKay G (2009) Validated ultra-performance 611–614
liquid chromatography-tandem mass spectrometry method for ana- 18. Service USDoHaH (2001) Guidance for the industry—bioanalytical
lyzing LSD, iso-LSD, nor-LSD, and O-H-LSD in blood and urine. J method validation
Anal Toxicol 33(5):253–259 19. Matuszewski BK, Constanzer ML, Chavez-Eng CM (2003)
6. Klette KL, Horn CK, Stout PR, Anderson CJ (2002) LC-mS analysis Strategies for the assessment of matrix effect in quantitative
of human urine specimens for 2-oxo-3-hydroxy LSD: method vali- bioanalytical methods based on HPLC-MS/MS. Anal Chem
dation for potential interferants and stability study of 2-oxo-3- 75(13):3019–3030

25
3.2 Publication 2

Pharmacokinetics and Concentration-Effect Relationship of Oral


LSD in Humans

Patrick C. Dolder1,2, Yasmin Schmid1, Manuel Haschke1, Katharina M. Rentsch2,


Matthias E. Liechti1

1 Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department


of Clinical Research University Hospital and University of Basel, Basel, Switzerland.
2 Laboratory Medicine, University Hospital and University of Basel, Basel, Switzerland

26
International Journal of Neuropsychopharmacology Advance Access published July 7, 2015

International Journal of Neuropsychopharmacology, 2015, 1–7

doi:10.1093/ijnp/pyv072
Research Article

research article
Pharmacokinetics and Concentration-Effect
Relationship of Oral LSD in Humans
Patrick C. Dolder, Yasmin Schmid, Manuel Haschke, Katharina M. Rentsch,
Matthias E. Liechti
Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical
Research (Mr Dolder, and Drs Schmid, Haschke, and Liechti), and Laboratory Medicine (Mr Dolder and
Dr Rentsch), University Hospital and University of Basel, Basel, Switzerland.

P.C.D. and Y.S. contributed equally to this work.


Correspondence: Matthias E. Liechti, MD, MAS, Prof., Division of Clinical Pharmacology and Toxicology, University Hospital Basel, Hebelstrasse 2, Basel,
CH-4031, Switzerland ([email protected]).

Abstract
Background: The pharmacokinetics of oral lysergic acid diethylamide are unknown despite its common recreational use and
renewed interest in its use in psychiatric research and practice.
Methods: We characterized the pharmacokinetic profile, pharmacokinetic-pharmacodynamic relationship, and urine recovery
of lysergic acid diethylamide and its main metabolite after administration of a single oral dose of lysergic acid diethylamide
(200 μg) in 8 male and 8 female healthy subjects.
Results: Plasma lysergic acid diethylamide concentrations were quantifiable (>0.1 ng/mL) in all the subjects up to 12 hours after
administration. Maximal concentrations of lysergic acid diethylamide (mean ± SD: 4.5 ± 1.4 ng/mL) were reached (median, range)
1.5 (0.5–4) hours after administration. Concentrations then decreased following first-order kinetics with a half-life of 3.6 ± 0.9
hours up to 12 hours and slower elimination thereafter with a terminal half-life of 8.9 ± 5.9 hours. One percent of the orally
administered lysergic acid diethylamide was eliminated in urine as lysergic acid diethylamide, and 13% was eliminated as
2-oxo-3-hydroxy-lysergic acid diethylamide within 24 hours. No sex differences were observed in the pharmacokinetic profiles
of lysergic acid diethylamide. The acute subjective and sympathomimetic responses to lysergic acid diethylamide lasted up to
12 hours and were closely associated with the concentrations in plasma over time and exhibited no acute tolerance.
Conclusions: These first data on the pharmacokinetics and concentration-effect relationship of oral lysergic acid diethylamide
are relevant for further clinical studies and serve as a reference for the assessment of intoxication with lysergic acid diethylamide.

Keywords:  LSD, O-H-LSD, pharmacokinetics, pharmacodynamics, plasma, urine

Trial registration: Registration identification number: NCT01878942


ClinicalTrials.gov: http://clinicaltrials.gov/ct2/show/NCT01878942.

Introduction
Lysergic acid diethylamide (LSD) is a prototypical hallucinogen 2008). However, no clinical research has been conducted with LSD
(Nichols, 2004; Passie et al., 2008). LSD became famous as a psyche- since the 1970s until recently (Gasser et al., 2014; Kupferschmidt,
delic in the 1960s, and its recreational use continues (Passie et al., 2014). Almost no scientific clinical pharmacological data on LSD

Received: April 13, 2015; Revised: June 22, 2015; Accepted: June 22, 2015
© The Author 2015. Published by Oxford University Press on behalf of CINP.
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://
creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium,
27
provided the original work is properly cited. For commercial re-use, please contact [email protected]
2 | International Journal of Neuropsychopharmacology, 2015

are available. Specifically, the pharmacokinetics (PK) of oral LSD once and 2 subjects who had used LSD twice. The subjects were
in humans are unknown. A  small PK study administered single asked to abstain from excessive alcohol consumption between
intravenous doses of 2  μg/kg in 5 healthy male human subjects test sessions and particularly limit their use to 1 drink on the
(Aghajanian and Bing, 1964). Blood samples were taken up to 8 day before the test sessions. Additionally, the participants were
hours after administration. Plasma concentrations were 6 to not allowed to drink xanthine-containing liquids after mid-
7 ng/mL 30 minutes after intravenous administration, 4–6 ng/mL night before the study day. Three subjects were light smokers
at 30–120 min, and approximately 1 ng/mL at 8 hours. The mean (<10 cigarettes/d) and were told to maintain their usual smok-
plasma elimination half-life of LSD was estimated at 175 minutes ing habits but not smoke during the sessions. We performed
in this previous study. In another study, single oral doses of 160 μg urine drug tests at screening and before each test session using
were administered to 13 male human subjects, and blood was TRIAGE 8 (Biosite, San Diego, CA). No alcohol test was performed.
sampled nonsystematically at various time points up to a maxi-
mum of 2.5 to 5 hours. Plasma levels peaked 40 to 130 minutes Study Outline
after LSD administration, and peaks ranged from 1.8 to 8.8 ng/mL
(Upshall and Wailling, 1972). The dataset and short sampling time The test sessions began at 8:15 AM. A urine sample was taken to
did not allow the calculation of PK parameters. verify abstinence from drugs of abuse, and a pregnancy test was
The aim of the present study was to characterize the single- performed in women. An indwelling intravenous catheter was
dose kinetics and PK-pharmacodynamic relationships of LSD placed in an antecubital vein for blood sampling, and the sub-
in healthy male and female subjects. For clinical and forensic jects completed baseline measurements. LSD (200 µg) or placebo
toxicologists, it is important to know the toxicokinetics of LSD was administered at 9:00 AM. A standardized lunch and dinner
and how plasma concentrations of LSD are linked to its dynamic was served at 1:30 PM and 5:30 PM, respectively. The subjects
effects and signs of intoxication. were sent home the next day at 9:30 AM after the 24-hour blood
LSD was administered in a single oral dose of 200  μg. The sample collection
same dose was used in a clinical study (Gasser et al., 2014). The
dose used was within the range of doses (50–400 μg) taken for Drugs
recreational purposes and expected to induce a full “LSD reac-
tion” (Nichols, 2004; Passie et  al., 2008). The study also evalu- Gelatin capsules that contained 100  µg LSD (D-LSD hydrate
ated the acute subjective, autonomic, and endocrine effects of with a purity (high-performance liquid chromatography) >99%;
LSD. The pharmacodynamics are reported in detail elsewhere Lipomed AG, Arlesheim, Switzerland), and corresponding pla-
(Schmid et  al., 2014), but the PK-pharmacodynamic relation- cebo capsules were prepared with authorization from the Swiss
ships are presented herein. Federal Office for Public Health. LSD was administered in a single
absolute dose of 200 µg, corresponding to a dose of 2.84 ± 0.5 µg/
kg body weight (mean ± SD; range: 2.04–3.85 μg).
Methods
Blood and Urine Sampling
Study Design
Blood was collected into lithium heparin tubes 1 hour before and
The study used a double-blind, placebo-controlled, cross-over
0.5, 1, 1.5, 2.5, 3, 4, 6, 8, 10, 12, 16, and 24 hours after LSD admin-
design with 2 experimental test sessions in balanced order. The
istration. Urine (entire volume) was collected during 3 sampling
washout periods between sessions were at least 7 days. The study
periods: 0 to 8, 8 to 16, and 16 to 24 hours after LSD administra-
was conducted in accordance with the Declaration of Helsinki
tion. Blood samples were immediately centrifuged, and plasma
and International Conference on Harmonization Guidelines in
and urine were rapidly stored at -20°C until analysis within 2
Good Clinical Practice and approved by the Ethics Committee
to 6  months. Long-term stability (6  months) has been shown
of the Canton of Basel, Switzerland and the Swiss Agency for
for LSD and 2-oxo-3-hydroxy-LSD (O-H-LSD) when kept under
Therapeutic Products (Swissmedic). The administration of LSD
refrigerated or frozen conditions (Klette et al., 2002; Martin et al.,
to healthy subjects was authorized by the Swiss Federal Office
2013). The recovery (ng) of LSD and O-H-LSD was determined by
for Public Health, Bern, Switzerland. The study was registered
multiplying the analyte urine concentrations (ng/mL) with the
at ClinicalTrials.gov (NCT01878942). All of the subjects pro-
urinary volume (mL) of the respective sampling interval.
vided written informed consent after being given written and
oral descriptions of the study, the procedures involved, and the
effects and possible risks of LSD administration. Analysis of LSD and O-H-LSD

LSD and O-H-LSD concentrations in plasma and urine were


Participants
determined using a validated liquid-chromatography-tandem
Sixteen healthy subjects (8 men and 8 women; mean age ± SD: mass-spectrometry method as reported in detail in the supple-
28.6 ± 6.2 years; range: 25–51 years) were included. The exclusion mentary Material online and elsewhere (Dolder et al., 2015). The
criteria are reported in detail elsewhere (Schmid et al., 2014) and lower limit of quantification was 0.1 ng/mL, and the upper limit
included age <25 or >65  years, pregnancy, personal or family of quantification was 10 ng/mL for LSD and O-H-LSD in both
(first-degree relative) history of psychotic or major affective dis- plasma and urine.
order, regular use of medications, chronic or acute physical ill-
ness, lifetime prevalence of illicit drug use >10 times (except for
PK
tetrahydrocannabinol), illicit drug use within the last 2 months,
and illicit drug use during the study. Nine subjects were halluci- The plasma concentration data were analyzed using noncom-
nogen-naive, and the other 7 had limited prior experience with partmental methods using Phoenix WinNonlin 6.4 (Certara,
hallucinogenic drugs, including 1 subject who had used LSD Princeton, NJ). Cmax and Tmax values were obtained directly from

28
Dolder et al.  |  3

the observed data. The area under the concentration-time concentration, Emax is the maximal effect, and h is the Hill slope
curve (AUC) from 0 to 24 hours after dosing (AUC24) was cal- using WinNonlin. Because of the hysteresis observed for most
culated using the linear-up log-down trapezoidal method. The plasma-concentration effect curves, an indirect descriptive link
terminal elimination rate constant (λz) for LSD was estimated model would be needed in which the plasma concentrations are
by log-linear regression after semilogarithmic transformation linked to the pharmacodynamic parameter by an effect com-
of the data using at least the last 3 data points of the termi- partment, providing an estimate of the equilibration half-life
nal linear phase of the concentration-time curve. The terminal between plasma and the effect compartment. However, because
half-life was calculated using λz and the equation t1/2 = ln2/λz. insufficient data pairs for the absorption phase (0-Cmax) were
The AUC to infinity was then determined by extrapolation of available, we directly linked dynamic effects to the plasma con-
the AUC24 using λz. We also determined a separate half-life for centrations using only data from Cmax up to 24 hours after drug
the Tmax to 12 hour interval, because the rate of elimination administration for this analysis. Statistical analyses were con-
changed at 12 hours in many subjects (see supplementary ducted using NCSS 2004 software (Statistical Software, Kaysville,
Figure S1 for all plots), and the decrease in plasma concentra- UT).
tions followed first-order kinetics in all subjects from Tmax to
12 hours. For this phase, we estimated the elimination rate Pharmacodynamic Measurements
constant (λ) for LSD using at least 3 data points of the con-
centration-time curve. Thus, this half-life does not describe Pharmacodynamic measures were included in this study to
the slower decrease in the concentration of LSD observed in a evaluate PK-pharmacodynamic relationships. Subjective effects
subset of subjects beyond 12 hours or 16 hours. Individual con- were assessed repeatedly over time using visual analog scales
centration-time curves show that a slower terminal decrease (VASs) (Hysek et  al., 2014), including “any drug effect,” “good
in LSD concentrations occurred only beyond 12 hours (after drug effect,” and “bad drug effect.” The VASs were presented as
eating dinner and during the night) and not concentration- 100-mm horizontal lines marked with “not at all” on the left and
dependent (ie, was not observed below a certain threshold “extremely” on the right. The VASs were administered 0, 0.5, 1,
concentration of LSD; see supplementary Figure S1). Renal 1.5, 2, 2.5, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 16, and 24 hours after drug
clearance (mL/h) was calculated as urinary recovery24 urine (ng)/ administration. Vital signs, including blood pressure, heart rate,
AUC24 (ng∙h/mL). and body (tympanic) temperature, were assessed repeatedly 0,
0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 12, and 24 hours after drug adminis-
tration using previously reported methods (Hysek et  al., 2014).
Statistical Analyses
Additionally, pupil size (dark-adapted maximal pupil diameter)
The analysis of the pharmacokinetic parameters was descrip- was measured 1, 2.5, 4, 7, and 11 hours after drug administration
tive, and geometric means and 90% CIs are shown to account for using an infrared pupillometer (PRL-200, NeurOptics, Irvine, CA)
nonnormally distributed data. The study included 8 subjects of under standardized dark-light conditions as previously reported
each sex; the data are also presented for male and female sub- (Hysek and Liechti, 2012).
jects separately. However, the study was not sufficiently pow-
ered (power: 52%) to exclude sex differences in the PK of LSD
Results
(PASS Power Analysis, Kaysville, UT).
The primary pharmacodynamic study results were reported PK
elsewhere (Schmid et  al., 2014). The a priori hypothesis relat-
ing to the PK-pharmacodynamics as defined in the study pro- Figure  1 shows the plasma-concentration-time curves for LSD
tocol was that the pharmacodynamic effects of LSD would and O-H-LSD. The PK parameters are shown in Table  1. The
show no acute pharmacological tolerance (ie, no clockwise plasma concentrations of LSD (>0.1 ng/mL) could be measured
hysteresis in the concentration-effect relationship). To assess in all of the subjects up to 12 hours, in 14 subjects up to 16
PK-pharmacodynamic relationships, the LSD-induced effect was hours, and in 11 subjects up to 24 hours after administration.
determined as a difference from placebo in the same subject at Concentrations of LSD decreased following first-order kinet-
the corresponding time point to control for circadian changes ics up to 12 hours with a half-life of 3.6 ± 0.9 hours (Figure 1b).
(Schmid et al., 2014). The pharmacodynamic changes after LSD In some subjects, a slower decrease in plasma concentrations
administration for each time point were plotted against the was observed late in time between 12 and 24 hours. This slower
respective plasma concentrations of LSD and graphed as hyster- decrease occurred after the subjective effects of LSD had mostly
esis curves for each subject. Because pupil size measurements subsided and the individual concentration-time curves showed
were unavailable at the same time points as plasma levels, pupil that the slower decrease was dependent on time >12 hours (after
size values at 7 and 11 hours were matched with concentrations eating dinner and during the night) and not on concentration
at 8 and 12 hours. No pupil size measurement was available for (ie, below a certain concentration of LSD) (see supplementary
the 24-hour time point; therefore, we used the baseline value Figure S1). The terminal half-life was 8.9 ± 5.9 hours including 4
at t  =  0 hours, assuming a return to baseline by 24 hours. The subjects (S4-S7, see supplementary Figure S1) in whom concen-
area within the hysteresis (AH) was calculated as AUCC0-Cmax – trations of LSD at 24 hours showed no further decrease com-
AUCCmax-C24 using the trapezoidal rule. AH < 0 indicates counter- pared with the 16-hour concentrations.
clockwise hysteresis (lag time between concentration and effect The O-H-LSD concentration-time profiles could be deter-
due to absorption/distribution processes). AH > 0 indicates clock- mined for only 8 subjects, because metabolite concentrations
wise hysteresis (tolerance). were not present or fell below the lower limit of quantification in
To estimate the plasma concentration of LSD at which 50% one-half of the subjects (Figure 1c-d). We could not show a differ-
of the maximal response to LSD is reached (EC50), a sigmoi- ence in the pharmacokinetic profiles of LSD between male and
dal concentration-response (variable slope) model was fitted female subjects (Table 1). The concentrations of LSD and O-H-LSD
to the plasma concentration-effect data: E = (Emax × Cph) / (Cph + in urine and the urine recovery of LSD and O-H-LSD are shown in
EC50h), in which E is the observed effect, Cp is the plasma LSD Table 2. The mean molar concentrations of O-H-LSD (molecular

29
4 | International Journal of Neuropsychopharmacology, 2015

Figure 1.  Pharmacokinetics (PK) of lysergic acid diethylamide (LSD) and 2-oxo-3-hydroxy-LSD (O-H-LSD). (a) Individual LSD plasma concentration-time curves with the
geometric mean shown in the inset. Filled circles indicate male subjects, and open circles indicate female subjects. (b) Semilogarithmic plot of the individual concen-
trations of LSD. Curves are shown separately for each individual in the supplementary Material (supplementary Figure S1). First-order kinetics were observed in all 16
subjects up to 12 hours. LSD levels fell below the lower limit of quantification (0.1 ng/mL) in 2 subjects at 16 hours and 5 subjects at 24 hours. Slower elimination was
observed between 12 and 24 hours. (c) Individual O-H-LSD plasma concentration-time curves in 8 subjects in whom metabolite concentrations could be determined,
with the geometric mean shown in the inset. (d) Semilogarithmic plot of the individual concentrations of O-H-LSD. Curves are shown separately in supplementary
Figure S2. LSD was administered at t = 0 hours.

Table 1.  Pharmacokinetic Parameters for LSD and O-H-LSD

Cmax (ng/ml) t1/2 (h) AUC24 (ng·h/ AUC∞ (ng·h/


Geometric tmax (h) Median Tmax-12 h t1/2 (h) Terminal mL) Geometric mL) Geometric CLR (mL/min)
N= Mean (95%CI) (range) Mean ± SD Mean ± SD Mean (90%CI) Mean (90%CI) Mean ± SD

LSD All 16 4.3 (3.8–4.9) 1.5 (0.5–4) 3.6 ± 0.9 8.9 ± 5.9 26 (22–30) 28 (24–33) 79 ± 36
LSD Male 8 4.4 (3.6–5.3) 1.5 (0.5–4) 3.5 ± 1.0 10.2 ± 6.7 25 (18–35) 28 (20–38) 88 ± 36
LSD Female 8 4.2 (3.4–5.3) 1.5 (0.5–3) 3.8 ± 0.8 7.6 ± 5.1 26 (23–30) 28 (25–32) 71 ± 36
a
O-H-LSD All 8 0.4 (0.3–0.5) 4 (2.5–6) 3.4 (2.6–4.3) 3.8 (2.8–5.3)

Abbreviations: AUC, area under the plasma concentration-time curv; AUC∞, AUC from time zero to infinity; AUC24, from time 0–24; CLR, renal clearance; Cmax, maxi-
mum observed plasma concentration; T1/2, plasma half-life; Tmax, time to reach Cmax; aO-H-LSD levels were above the limit of detection in only 8 subjects).

Table 2.  Urinary Elimination of LSD and O-H-LSD

LSD O-H-LSD

N= 0–8 hours 8–16 hours 16–24 hours 0–24 hours 0–8 hours 8–16 hours 16–24 hours 0–24 hours

Urinary concentrations (ng/mL)


 all 16 0.96 ± 0.8 1.1 ± 1.8 0.70 ± 0.6 8.3 ± 4.7 17.7 ± 11 14.4 ± 10
 male 8 0.78 ± 0.4 0.82 ± 0.2 0.66 ± 0.6 6.6 ± 3.4 22.7 ± 14 11.2 ± 7
 female 8 1.1 ± 1.0 1.5 ± 2.6 0.74 ± 0.7 9.9 ± 5.4 12.7 ± 5.3 17.6 ± 12
Urinary volume (L)
 all 16 1.4 ± 0.7 0.79 ± 0.4 0.47 ± 0.3
 male 8 1.8 ± 0.8 0.86 ± 0.5 0.63 ± 0.2
 female 8 1.1 ± 0.5 0.71 ± 0.4 0.30 ± 0.2
Urinary recovery (nM) Ae0-24
 all 16 3.6 ± 2.6 2.0 ± 1.7 0.82 ± 0.5 6.4 ± 2.9 28.3 ± 15 35.9 ± 27 15.3 ± 7.8 79.5 ± 41
 male 8 3.8 ± 2.4 2.0 ± 0.8 1.1 ± 0.6 6.8 ± 2.6 29.3 ± 19 49.6 ± 33 17.1 ± 8.6 96.1 ± 51
 female 8 3.5 ± 2.8 2.0 ± 2.3 0.58 ± 0.3 6.0 ± 3.3 27.3 ± 13 22.3 ± 9* 13.4 ± 6.8 62.9 ± 18

Abbreviations: Ae, amount eliminated in nM; LSD, lysergic acid diethylamide; O-H-LSD, 2-oxo-3-hydroxy-LSD.
*Significant difference from men (P < .05). Values are mean ± SD.
30
Dolder et al.  |  5

weight: 355.4) were 23.2, 49.9, and 40.6 pM/mL and 8, 14, and 19 found for heart rate (Figure 2a), blood pressure (Figure 2b), or bad
times higher than the mean molar concentrations of LSD (molec- drug effect (Figure 2g). The 95% CIs of the mean of the area within
ular weight: 323.4; 3.0, 3.5, 2.2 pM/mL) in the 0 to 8, 8 to 16, and the hysteresis loops (AH) overlapped with 0 for heart rate (4.4
16 to 24 hour sampling intervals, respectively. Of the nonmetabo- beats × ng/min × mL [-13 to +22]), blood pressure (-5 mgHg × ng/
lized LSD that was recovered from urine, 56% appeared in urine min × mL [-24 to +13]), and bad drug effect (5% × ng/min × mL
within the first 8 hours after administration and 45% of the O-H- [-29 to +38]), indicating no hysteresis. Counterclockwise hyster-
LSD appeared in urine 8 to 16 hours after LSD administration. esis (negative AH value) was observed, attributable to relatively
Of the orally administered LSD hydrate (200  μg or 618 nM), 13% higher plasma levels compared with the dynamic effects dur-
was eliminated in urine as O-H-LSD (28.3  μg or 79.5 nM) within ing the assumed drug absorption phase (0–2 hours) for body
24 hours. Only 1% (2.1 μg or 6.4 nM) of the dose of LSD was elim- temperature (Figure  2c), pupil size (Figure  2d), any drug effect
inated in urine as LSD within 24 hours. The renal clearance of (Figure 2e), and good drug effect (Figure 2f). Mean AH values (95%
LSD was 1.32 ± 0.6 mL/min or approximately 1.6% of the apparent CI) were the following: body temperature (-1°C × ng/min × mL
total clearance after oral administration (CL/F), assuming an oral [-1.5 to -0.5]), pupil size (-1.4  mm × ng/min × mL [-2.2 to -0.7]),
bioavailability of 71% (see Discussion). No significant differences any drug effect (-78% × ng/min × mL [-113 to -43]), and good drug
in LSD or O-H-LSD urine concentrations were observed between effect (-106% × ng/min × mL [-151 to -61]). The decline of the
male and female subjects (Table 2). The urine recovery of O-H-LSD response to LSD and plasma concentration over time followed a
was greater in male subjects than in female subjects during the sigmoidal Emax dose-response curve for any drug effect and good
8 to 16 hour sampling period, but no significant differences were drug effect. The EC50 mean ± SD values were 1.3 ± 0.7 ng/mL for
observed in the overall 0 to 24 hour sampling (Table 2). any drug effect and 1.0 ± 0.5 ng/mL for good drug effect. Heart
rate, blood pressure, body temperature, and bad drug effect
linearly increased with plasma concentrations of LSD and did
PK-Pharmacodynamic Relationship
not show an Emax (Figure 2a-c, g). Not enough values were avail-
Figure 2 shows the effects of LSD as a function of plasma con- able to fit changes in pupil size. No clockwise hysteresis was
centration. There was a close relationship between the LSD con- observed for any of the concentration-effect curves, meaning
centration and its dynamic effects overt time. No hysteresis was that the dynamic values were higher later in time at a given

Figure 2.  Lysergic acid diethylamide (LSD) effects plotted against LSD plasma concentrations (geometric means). The pharmacodynamic values are the mean ± SEM
differences from placebo at each time point in 16 subjects. The time of sampling is noted next to each point (in hours after LSD administration). Heart rate (a), mean
arterial pressure (b), and bad drug effect (g) showed no hysteresis. Counterclockwise hysteresis was observed for body temperature (c), pupil size (d), any drug effect (e),
and good drug effect (f), consistent with a delay between plasma concentration and effect. For most dynamic variables, maximal plasma concentrations (at approxi-
mately 2 hours) coincided with maximal dynamic effects. The dynamic changes then gradually decreased over time with decreasing plasma levels. No evidence of
acute tolerance (clockwise hysteresis) was observed for any of the dynamic effects of LSD.

31
6 | International Journal of Neuropsychopharmacology, 2015

plasma concentration and consistent with no acute tolerance to is unknown. In cats, the unbound fraction was 0.2, and LSD
the effects of LSD. LSD produced acute adverse effects, including concentrations in cerebrospinal fluid were similar to free LSD
difficulty concentrating, headache, exhaustion, and dizziness plasma concentrations (Axelrod et al., 1957). Thus, LSD concen-
lasting up to 24 hours and as reported elsewhere (Schmid et al., trations of 0.6 to 0.8 nM could be expected in cerebrospinal fluid.
2014). There were no severe adverse effects. These values are in the range of the binding affinity of LSD at
the 5-hydroxytryptamine-2A (5-HT2A) receptor (Ki  =  0.4–1.3 nM,
respectively) (Titeler et al., 1988; Egan et al., 1998) and also close
Discussion
to the EC50 for the functional stimulant activity of LSD at the
The present study determined the single-dose PK of oral LSD in receptor in vitro (EC50 = 7.2 nM) (Egan et al., 1998). Pupil size was
humans. The concentrations of LSD were maximal after 1.5 hours also strongly increased at low concentrations of LSD. We pre-
(median) and gradually declined to very low levels by 12 hours. We viously showed that pupil diameters were significantly larger
observed first-order kinetics of LSD up to 12 hours in all subjects compared with placebo until the last pupil measurement at 11
and an inconsistent slower decrease in concentrations thereafter hours after LSD administration. In contrast, elevations in blood
in some subjects. This could be attributable to redistribution from pressure, heart rate, and body temperature were only significant
tissue or due to less precise quantification of the very low plasma up to 5 hours after LSD administration compared with placebo,
levels of LSD at 12 to 24 hours (ie, close to the lower limit of quan- as reported elsewhere (Schmid et  al., 2014). Additionally, the
tification). The half-life of 3.6 hours during the first 12 hours after increases in heart rate, blood pressure, body temperature, and
drug administration is close to the 3 hours previously observed bad drug effects showed no ceiling effect in the concentration-
in a small study that used intravenous LSD administration effect curves, in contrast to the other dynamic effects of LSD.
(Aghajanian and Bing, 1964). Only 1% of the orally administered Heart rate, body temperature, blood pressure, and bad drug
LSD was eliminated renally. LSD is almost completely metabo- effects would likely increase further with higher doses of LSD,
lized in rats, guinea pigs, and monkeys (Axelrod et al., 1957; Siddik whereas the pupillary or good subjective effects can be expected
et al., 1979). In humans, the major metabolite of LSD detectable in to be similar to those seen in the present study. The hyperten-
urine is O-H-LSD (Klette et al., 2000; Poch et al., 2000; Canezin et al., sive effects of LSD may result from 5-HT2A and/or α1-adrenergic
2001). In the present study, O-H-LSD was detected in blood plasma receptor-mediated vasoconstrictive effects at higher doses (Dyer
at very low concentrations and in only one-half of the subjects. and Gant, 1973; Blessing and Seaman, 2003).
The urine concentrations of O-H-LSD in the present study were No evidence of acute tolerance was observed, which would
approximately 10, 15, and 20 times higher than those of LSD at 0 become apparent as clockwise hysteresis in the concentration-
to 8, 8 to 16, and 16 to 24 hours after LSD administration. Similarly, response curve and has been shown for 3,4-methylenediox-
in LSD-positive forensic urine samples, O-H-LSD concentrations ymethamphetamine (MDMA) (Hysek et  al., 2011). In contrast
are higher than those of LSD, and O-H-LSD can be detected for and as typically expected for most drugs, counterclockwise
a longer time than LSD after LSD administration (Reuschel et al., hysteresis was observed early in time until the end of the
1999; Klette et  al., 2000; Poch et  al., 2000). In the present study, assumed drug absorption phase. No similar studies on the
13% of the orally administered LSD was recovered from urine PK-pharmacodynamic relationship of LSD have been performed.
as O-H-LSD within 24 hours. LSD is metabolized to O-H-LSD by Only one other small study measured plasma LSD concentra-
cytochrome P450 enzymes, but the specific enzymes and mech- tions and concomitant pharmacodynamic effects (Aghajanian
anisms are unknown (Klette et al., 2000). To our knowledge, it is and Bing, 1964). LSD was administered intravenously in 5 male
unknown whether O-H-LSD is pharmacologically active. subjects. To obtain a crude index of performance, subjects
The oral bioavailability of LSD can be crudely estimated were given one of a series of equivalent tests, consisting of
using the previous data on intravenous LSD administration simple addition problems, after each blood sample was drawn
(Aghajanian and Bing, 1964) and our data on oral LSD. After intra- (Aghajanian and Bing, 1964). After the distribution phase (30
venous LSD administration (2  μg/kg of the free base in 5 male minutes after intravenous LSD administration), the impair-
subjects), a mean total plasma exposure (AUC∞) of 31.4 ng∙mL/h ments in performance declined in parallel with the plasma
was obtained (15.7 ng∙mL/h per μg/kg free base), calculated based levels of LSD, also suggesting a close temporal relationship
on the published plasma concentration profile (Aghajanian and between the PK and pharmacodynamics of LSD (Aghajanian
Bing, 1964). After oral LSD administration in the present study and Bing, 1964). In contrast to the single-dose administration in
(2.5  μg/kg free base in 8 male subjects), the mean AUC∞ was the present study, tolerance to the subjective effects of LSD with
28 ng∙mL/h (11.2 ng∙mL/h per μg/kg free base). Based on these repeated daily LSD administration has been reported (Abramson
data, the oral bioavailability of LSD is approximately 71%. In the et  al., 1956; Belleville et  al., 1956). However, a gradual increase
present study, LSD was administered after a light meal. When in head twitches and catatonic postures and no tolerance was
ingested with a “full breakfast,” oral LSD was reported to result observed up to 3 to 4  days after continuous LSD administra-
in lower plasma concentrations compared with administration tion in rats (Ellison et al., 1980). Also in contrast to our findings
on an empty stomach (Upshall and Wailling, 1972). However, with LSD, we observed pronounced acute tolerance to the psy-
these observations were made in only 2 to 3 subjects (Upshall chotropic and cardiostimulant effects of MDMA using the same
and Wailling, 1972) and would need confirmation. Remaining to methodology (Hysek et al., 2011). As a result, the pharmacody-
be tested is whether food reduces or delays the absorption of oral namic effects of MDMA last significantly shorter than would
LSD. Additionally, the PK profiles were similar in male and female be expected based on plasma levels. The subjective and cardi-
subjects. However, the study was too underpowered to statisti- ostimulant effects of MDMA last only 5 hours despite its long
cally exclude sex differences in the PK of LSD. half-life of 10 hours (Hysek et al., 2011). In contrast, the subjec-
We found a close relationship between the plasma con- tive drug effects of LSD lasted for 12 hours in most subjects and
centrations of LSD and physiologic response or psychotropic up to 16 hours in some subjects in the present study despite
effects of LSD over time. Estimated EC50 values for the psycho- LSD’s shorter half-life. Thus, subjects with MDMA in blood may
tropic effects were in the range of 1.0 to 1.3 ng/mL (approxi- no longer be clinically intoxicated, whereas subjects with quan-
mately 3–4 nM). The unbound fraction of LSD in human plasma tifiable LSD concentrations in plasma are clinically intoxicated.

32
Dolder et al.  |  7

A mechanistic explanation for this acute tolerance in the case Ellison G, Ring M, Ross D, Axelrood B (1980) Cumulative altera-
of MDMA is that it mainly produces its acute effects through tions in rat behavior during continuous administration of LSD
the release of endogenous serotonin and norepinephrine (ie, or mescaline: absence of tolerance? Biol Psychiatry 15:95–102.
as an indirect serotonergic and noradrenergic agonist). In con- Gasser P, Holstein D, Michel Y, Doblin R, Yazar-Klosinski B, Pas-
trast, LSD is thought to produce its psychotropic hallucinogenic sie T, Brenneisen R (2014) Safety and efficacy of lysergic acid
effects through a direct interaction with the 5-HT2A receptor (ie, diethylamide-assisted psychotherapy for anxiety associated
as a direct serotonergic agonist), resulting in pharmacodynamic with life-threatening diseases. J Nerv Ment Dis 202:513–520.
effects to which no acute tolerance was observed in our study. Hysek CM, Liechti ME (2012) Effects of MDMA alone and after
In summary, we show first data on the PK and pretreatement with reboxetine, duloxetine, clonidine, carve-
PK-pharmacodynamic relationship of oral LSD in human sub- dilol, and doxazosin on pupillary light reflex. Psychopharma-
jects. The PK profiles exhibit first-order kinetics of LSD up to cology 224:363–376.
12 hours. LSD produces physiological and psychotropic effects Hysek CM, Simmler LD, Ineichen M, Grouzmann E, Hoener MC,
lasting up to 12 hours, closely related to the plasma concentra- Brenneisen R, Huwyler J, Liechti ME (2011) The norepineph-
tions of LSD and inhibiting no acute tolerance. The findings are rine transporter inhibitor reboxetine reduces stimulant
important for further clinical studies and serve as a reference effects of MDMA (“ecstasy”) in humans. Clin Pharmacol Ther
for the assessment of intoxication with LSD. 90:246–255.
Hysek CM, Simmler LD, Schillinger N, Meyer N, Schmid Y,
Donzelli M, Grouzmann E, Liechti ME (2014) Pharmacoki-
Acknowledgments netic and pharmacodynamic effects of methylphenidate and
The authors thank Stefan Borgwardt, Felix Müller, and Florian MDMA administered alone and in combination. Int J Neu-
Enzler for their assistance with conducting the clinical study; ropsychopharmacol 17:371–381.
Stephan Krähenbühl for comments on the manuscript; and Klette KL, Anderson CJ, Poch GK, Nimrod AC, ElSohly MA (2000)
Michael Arends for editorial assistance. Supported by the Metabolism of lysergic acid diethylamide (LSD) to 2-oxo-
University Hospital Basel, Switzerland, and Swiss National 3-hydroxy LSD (O-H-LSD) in human liver microsomes and
Science Foundation (grant no. 320030_1449493). cryopreserved human hepatocytes. J Anal Toxicol 24:550–556.
Klette KL, Horn CK, Stout PR, Anderson CJ (2002) LC-MS analysis
of human urine specimens for 2-oxo-3-hydroxy LSD: method
Statement of Interest validation for potential interferants and stability study of
The authors declare no competing financial interests. 2-oxo-3-hydroxy LSD under various storage conditions. J Anal
Toxicol 26:193–200.
Kupferschmidt K (2014) High hopes. Science 345:18–23.
References Martin R, Schurenkamp J, Gasse A, Pfeiffer H, Kohler H (2013)
Abramson HA, Jarvik ME, Gorin MH, Hirsch MW (1956) Lysergic Determination of psilocin, bufotenine, LSD and its metabo-
acid diethylamide (LSD-25): XVII tolerance development and lites in serum, plasma and urine by SPE-LC-MS/MS. Int J Legal
its relationship to a theory of psychosis. J Psychol 41:81–105. Med 127:593–601.
Aghajanian GK, Bing OH (1964) Persistence of lysergic acid dieth- Nichols DE (2004) Hallucinogens. Pharmacol Ther 101:131–181.
ylamide in the plasma of human subjects. Clin Pharmacol Passie T, Halpern JH, Stichtenoth DO, Emrich HM, Hintzen A
Ther 5:611–614. (2008) The pharmacology of lysergic acid diethylamide: a
Axelrod J, Brady RO, Witkop B, Evarts EV (1957) The distribution review. CNS Neurosci Ther 14:295–314.
and metabolism of lysergic acid diethylamide. Ann N Y Acad Poch GK, Klette KL, Anderson C (2000) The quantitation of 2-oxo-
Sci 66:435–444. 3-hydroxy lysergic acid diethylamide (O-H-LSD) in human
Belleville RE, Fraser HF, Isbell H, Wikler A, Logan CR (1956) Stud- urine specimens, a metabolite of LSD: comparative analysis
ies on lysergic acid diethylamide (LSD-25): I.  Effects in for- using liquid chromatography-selected ion monitoring mass
mer morphine addicts and development of tolerance during spectrometry and liquid chromatography-ion trap mass
chronic intoxication. AMA Arch Neurol Psychiatry 76:468–478. spectrometry. J Anal Toxicol 24:170–179.
Blessing WW, Seaman B (2003) 5-hydroxytryptamine2A receptors Reuschel SA, Eades D, Foltz RL (1999) Recent advances in chro-
regulate sympathetic nerves constricting the cutaneous vas- matographic and mass spectrometric methods for determi-
cular bed in rabbits and rats. Neuroscience 117:939–948. nation of LSD and its metabolites in physiological specimens.
Canezin J, Cailleux A, Turcant A, Le Bouil A, Harry P, Allain P J Chromatogr B Biomed Sci Appl 733:145–159.
(2001) Determination of LSD and its metabolites in human Schmid Y, Enzler F, Gasser P, Grouzmann E, Preller KH, Vollen-
biological fluids by high-performance liquid chromatography weider FX, Brenneisen R, Müller F, Borgwardt S, Liechti ME
with electrospray tandem mass spectrometry. J Chromatogr (2014) Acute effects of lysergic acid diethylamide in healthy
B Biomed Sci Appl 765:15–27. subjects. Biol Psychiatry doi: 10.1016/j.biopsych.2014.11.015.
Dolder PC, Liechti ME, Rentsch KM (2015) Development and Siddik ZH, Barnes RD, Dring LG, Smith RL, Williams RT (1979) The
validation of a rapid turboflow LC-MS/MS method for the fate of lysergic acid DI[14C]ethylamide ([14C]LSD) in the rat,
quantification of LSD and 2-oxo-3-hydroxy LSD in serum and guinea pig and rhesus monkey and of [14C]iso-LSD in rat. Bio-
urine samples of emergency toxicological cases. Anal Bioanal chem Pharmacol 28:3093–3101.
Chem 407:1577–84. Titeler M, Lyon RA, Glennon RA (1988) Radioligand binding evi-
Dyer DC, Gant DW (1973) Vasoconstriction produced by halluci- dence implicates the brain 5-HT2 receptor as a site of action
nogens on isolated human and sheep umbilical vasculature. for LSD and phenylisopropylamine hallucinogens. Psychop-
J Pharmacol Exp Ther 184:366–375. harmacology 94:213–216.
Egan CT, Herrick-Davis K, Miller K, Glennon RA, Teitler M (1998) Upshall DG, Wailling DG (1972) The determination of LSD in
Agonist activity of LSD and lisuride at cloned 5HT2A and 5HT2C human plasma following oral administration. Clin Chim Acta
receptors. Psychopharmacology 136:409–414. 36:67–73.

33
3.3 Publication 3

Alterations of consciousness and mystical-type experiences after


acute LSD in humans

Matthias E. Liechti1, Patrick C. Dolder1, Yasmin Schmid1

1 Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of


Clinical Research, University Hospital Basel and University of Basel, Switzerland

34
Psychopharmacology
DOI 10.1007/s00213-016-4453-0

ORIGINAL INVESTIGATION

Alterations of consciousness and mystical-type experiences


after acute LSD in humans
Matthias E. Liechti 1 & Patrick C. Dolder 1 & Yasmin Schmid 1

Received: 25 August 2016 / Accepted: 27 September 2016


# The Author(s) 2016. This article is published with open access at Springerlink.com

Abstract On the 5D-ASC scale, LSD produced higher ratings of blissful


Rationale Lysergic acid diethylamide (LSD) is used state, insightfulness, and changed meaning of percepts after
recreationally and in clinical research. Acute mystical-type 200 μg compared with 100 μg. Plasma levels of LSD were not
experiences that are acutely induced by hallucinogens are positively correlated with its effects, with the exception of ego
thought to contribute to their potential therapeutic effects. dissolution at 100 μg.
However, no data have been reported on LSD-induced mys- Conclusions Mystical-type experiences were infrequent after
tical experiences and their relationship to alterations of con- LSD, possibly because of the set and setting used in the pres-
sciousness. Additionally, LSD dose- and concentration- ent study. LSD may produce greater or different alterations of
response functions with regard to alterations of consciousness consciousness at 200 μg (i.e., a dose that is currently used in
are lacking. psychotherapy in Switzerland) compared with 100 μg (i.e., a
Methods We conducted two placebo-controlled, double- dose used in imaging studies). Ego dissolution may reflect
blind, cross-over studies using oral administration of 100 plasma levels of LSD, whereas more robustly induced effects
and 200 μg LSD in 24 and 16 subjects, respectively. Acute of LSD may not result in such associations.
effects of LSD were assessed using the 5 Dimensions of
Altered States of Consciousness (5D-ASC) scale after both Keywords LSD . Altered states of consciousness . Mystical
doses and the Mystical Experience Questionnaire (MEQ) after experiences
200 μg.
Results On the MEQ, 200 μg LSD induced mystical experi-
ences that were comparable to those in patients who Introduction
underwent LSD-assisted psychotherapy but were fewer than
those reported for psilocybin in healthy subjects or patients. Lysergic acid diethylamide (LSD) is the prototypical halluci-
nogen (Nichols 2016; Passie et al. 2008). LSD became fa-
mous, with a high cultural influence, in the 1960s. LSD con-
The studies were registered at ClinicalTrials.gov (NCT02308969,
tinues to be used for recreational and personal purposes
NCT01878942).
(Krebs and Johansen 2013). Additionally, there is much inter-
Electronic supplementary material The online version of this article est in its therapeutic potential (Baumeister et al. 2014;
(doi:10.1007/s00213-016-4453-0) contains supplementary material,
which is available to authorized users.
Davenport 2016; Gasser et al. 2014; Gasser et al. 2015;
Krebs and Johansen 2012; Kupferschmidt 2014). Only one
* Matthias E. Liechti modern study has tested the therapeutic effects of LSD in
[email protected] patients (Gasser et al. 2014), whereas several clinical trials
have recently evaluated the therapeutic potential of psilocybin
1
(Bogenschutz et al. 2015; Carhart-Harris et al. 2016a; Garcia-
Psychopharmacology Research, Division of Clinical Pharmacology
and Toxicology, Department of Biomedicine and Department of
Romeu et al. 2015; Griffiths 2016; Grob et al. 2011; Guss
Clinical Research, University Hospital Basel, University of Basel, 2016), a similar serotonergic hallucinogen (Rickli et al.
Hebelstrasse 2, CH-4031 Basel, Switzerland 2016). A series of studies showed that psilocybin acutely

35
Psychopharmacology

induced mystical experiences in healthy subjects and patients with many other psychedelics, providing an opportunity to com-
(Garcia-Romeu et al. 2015; Griffiths et al. 2008; Griffiths et al. pare findings between studies and across substances and research
2011; Griffiths et al. 2006; MacLean et al. 2011). groups. Thus, the present study assessed LSD dose- and plasma
Additionally, greater acute effects of psilocybin on the concentration-response functions using the 5D-ASC scale in 40
Mystical Experience Questionnaire (MEQ; Barrett et al. subjects (Dolder et al. 2015b; Dolder et al. 2016; Schmid et al.
2015; Griffiths et al. 2006; MacLean et al. 2012) were asso- 2015), thus allowing comparisons with other studies that used
ciated with positive long-term effects on mood and personality the 5D-ASC scale but did not determine plasma LSD concentra-
in healthy subjects (Griffiths et al. 2008; Griffiths et al. 2011; tions (Carhart-Harris et al. 2016b; Carhart-Harris et al. 2016c;
Griffiths et al. 2006; MacLean et al. 2011) and better thera- Kaelen et al. 2016; Lebedev et al. 2016; Roseman et al. 2016;
peutic outcomes in patients with anxiety, depression, and sub- Speth et al. 2016; Tagliazucchi et al. 2016; Terhune et al. 2016).
stance use disorder (Garcia-Romeu et al. 2015; Griffiths 2016; A third goal of the present study was to assess associations across
Griffiths et al. 2008; Griffiths et al. 2011; Griffiths et al. 2006; subjects between the peak and total plasma exposure to LSD and
MacLean et al. 2011). Early studies reported on mystical its effects on 5D-ASC scale scores (Studerus et al. 2010). The
experiences after experimental administration of LSD, but effects of 100 μg LSD on 5D-ASC scale scores are reported for
methodological details are missing (Turek et al. 1974). the first time in the present study, whereas the effects of 200 μg
Whether and the extent to which LSD produces mystical- have been previously published (Schmid et al. 2015). However,
type effects in the MEQ are currently unknown. Therefore, the latter study did not evaluate dose- or concentration-response
we characterized the effects of 200 μg LSD on the MEQ functions. Other data that were generated in the present study
and evaluated the way in which mystical experiences are have been previously reported including acute and subacute ad-
related to LSD-induced increases in 5 Dimensions of verse effects (Dolder et al. 2015b; Dolder et al. 2016; Schmid
Altered States of Consciousness (5D-ASC) scale scores et al. 2015; Strajhar et al. 2016).
and plasma levels of LSD.
Clinical experimental research with LSD has recently seen a
resurgence (Carhart-Harris et al. 2016b; Carhart-Harris et al. Material and methods
2015; Carhart-Harris et al. 2016c; Dolder et al. 2015b; Dolder
et al. 2016; Kaelen et al. 2015; Kaelen et al. 2016; Lebedev et al. Study design
2016; Roseman et al. 2016; Schmid et al. 2015; Speth et al.
2016; Strajhar et al. 2016; Tagliazucchi et al. 2016; Terhune We performed two similar studies using double-blind, place-
et al. 2016). An increasing amount of data has been generated bo-controlled, cross-over designs with two experimental test
on the effects of LSD (75 μg) on various neuronal correlates of sessions (LSD and placebo) in a balanced order. Study 1 used
brain activation (Carhart-Harris et al. 2016c; Kaelen et al. 2016; a dose of 100 μg LSD and placebo in 24 subjects. Study 2
Lebedev et al. 2016; Roseman et al. 2016). Researchers have used 200 μg LSD and placebo in 16 subjects. The washout
correlated subjective drug effects with brain functional magnetic periods between sessions were at least 7 days. The studies
resonance imaging (fMRI) data (Carhart-Harris et al. 2016c; were conducted in accordance with the Declaration of
Kaelen et al. 2016; Lebedev et al. 2016; Roseman et al. 2016). Helsinki and approved by the local ethics committee. The
This approach likely produces significant findings for subjective administration of LSD to healthy subjects was authorized by
effects that show large between-subject variance but not for sub- the Swiss Federal Office for Public Health, Bern, Switzerland.
jective effects of the substance that are consistently present in all All of the subjects provided written consent before participat-
subjects. Lower doses of LSD may also result in more variable ing in either of the studies, and they were paid for their par-
responses across subjects compared with higher doses. ticipation. The studies were registered at ClinicalTrials.gov
Furthermore, higher doses of LSD (e.g., 200 μg) that are cur- (NCT02308969, NCT01878942).
rently used therapeutically (Gasser et al. 2014) may produce
more pronounced but also qualitatively different subjective ef- Participants
fects (Dolder et al. 2016). Importantly, plasma concentrations of
LSD have not been determined in any of the published LSD Forty healthy participants were recruited from the University
fMRI studies to date; therefore, unclear is the way in which of Basel campus via online advertisement. Twenty-four sub-
LSD exposure in the body is linked to subjective effects in these jects (12 men, 12 women; 33 ± 11 years old [mean ± SD];
studies. Therefore, a second goal of the present study was to range, 25–60 years) participated in study 1, and 16 subjects (8
describe the subjective peak effects of two doses of LSD (100 men, 8 women; 29 ± 6 years old; range, 25–51 years) partic-
and 200 μg) using the 5D-ASC scale (Studerus et al. 2010). The ipated in study 2. The inclusion and exclusion criteria were
5D-ASC scale has been used in all of the recent experimental identical for both studies. Subjects younger than 25 years of
studies with LSD (Carhart-Harris et al. 2016b; Carhart-Harris age were excluded from participating in the study. Additional
et al. 2016c; Schmid et al. 2015; Tagliazucchi et al. 2016) and exclusion criteria were age >65 years, pregnancy (urine

36
Psychopharmacology

pregnancy test at screening and before each test session), per- purposes (Passie et al. 2008). Corresponding placebo capsules
sonal or family (first-degree relative) history of major psychi- were used.
atric disorders (assessed by the semi-structured clinical inter-
view for Diagnostic and Statistical Manual of Mental Measures
Disorders, 4th edition, Axis I disorders by the study physician
and an additional interview by a trained psychiatrist), use of Mystical-type experiences In study 2, mystical experiences
medications that may interfere with the study medication, were assessed using a German version (Supplementary
chronic or acute physical illness (abnormal physical exam, Appendix 1) of the 43-item MEQ (Griffiths et al. 2006;
electrocardiogram, or hematological and chemical blood anal- MacLean et al. 2012; Pahnke 1969) embedded in the 100-
yses), tobacco smoking (>10 cigarettes/day), lifetime preva- item States of Consciousness Questionnaire (SOCQ; (Griffiths
lence of illicit drug use >10 times (except for tetrahydrocan- et al. 2006). The original English questionnaire was indepen-
nabinol), illicit drug use within the last 2 months, and illicit dently forward-translated into German by two translators with
drug use during the study (determined by urine drug tests). German as their mother tongue. Discrepancies between the two
The subjects were asked to abstain from excessive alcohol forward-translated versions and a previous German version
consumption between test sessions and particularly limit their were then discussed and selected items backtranslated. The
use to one standard drink on the day before the test sessions. version was then pretested for comprehension by persons with
Additionally, the participants were not allowed to drink previous LSD or MDMA use.
xanthine-containing liquids after midnight before the study The MEQ has been used in numerous experimental and
day. Eleven subjects had used a hallucinogen, including therapeutic trials with psilocybin (Garcia-Romeu et al. 2015;
LSD (six participants), one to three times, and most of the Griffiths et al. 2008; Griffiths et al. 2011; Griffiths et al. 2006;
subjects (29) were hallucinogen-naive. We performed urine MacLean et al. 2011). The MEQ items provide scale scores for
drug tests at screening and before each test session, and no each of seven domains of mystical experiences: internal unity,
substances were detected during the study. external unity, sacredness, noetic quality (as real as or more real
than everyday reality), deeply felt positive mood, transcendence
Study procedures of time and space, and ineffability/paradoxicality (difficulty de-
scribing the experience in words). The total of all scale scores
Each study included a screening visit, a psychiatric interview, was used as an overall measure of the mystical-type experience.
two 25-h experimental sessions, and an end-of-study visit. The We also derived the four scale scores of the newly validated
experimental sessions were conducted in a quiet standard hos- revised 30-item MEQ: mystical, positive mood, transcendence
pital patient room. The participants were resting in hospital beds of time and space, and ineffability (Barrett et al. 2015). A com-
except when going to the restroom. Only one research subject plete mystical experience was defined as scores ≥60 % on all
and one or two investigators were present during the experi- MEQ30 factors (Barrett et al. 2015). The MEQ was administered
mental sessions. The participants could interact with the inves- 24 h after drug administration, and the participants were asked to
tigator, rest quietly, and/or listen to music via headphones, but retrospectively rate drug effects during peak drug effects. For
no other entertainment was provided. LSD or placebo was ad- comparison, we included MEQ ratings that were obtained 6 h
ministered at 9:00 AM. The subjects were never alone during after administration of 3,4-methylenedioxymethamphetamine
the first 12 h after drug administration, and the investigator was (MDMA) and methylphenidate in another study using a similar
in a room next to the subject for up to 24 h while the subjects research setting (Schmid et al. 2014). Additionally, we included
were asleep (mostly from 1:00 AM to 8:00 AM). MEQ ratings from patients who were treated with 200 μg LSD
for anxiety related to life-threatening illness in another study
Study drug (Diesch 2015; Gasser et al. 2014; Gasser et al. 2015). All of these
additional MEQ findings have not been previously published in
LSD (d-LSD hydrate, HPLC purity >99 %, Lipomed AG, scientific journals and were obtained in studies that were previ-
Arlesheim, Switzerland) was administered in single oral doses ously described in detail (Diesch 2015; Gasser et al. 2014; Gasser
of 100 or 200 μg as gelatin capsules. Note that these LSD et al. 2015; Schmid et al. 2014).
hydrate doses correspond to LSD tartrate doses of 123 and
246 μg, respectively. In the 1960–1970s, small doses of Alterations of consciousness The 5D-ASC scale was used in
LSD tartrate of 25–150 μg were typically used in “psycholytic both studies to assess the overall peak alterations of conscious-
therapy” and higher doses of >200 μg in “psychedelic” ther- ness. The 5D-ASC scale measures altered states of conscious-
apy (Pahnke et al. 1970). The dose used in a recent LSD- ness and contains 94 items (visual analog scales). The instru-
assisted psychotherapy study was 200 μg LSD hydrate ment consists of five subscales/dimensions (Dittrich 1998)
(Gasser et al. 2014). Both doses used in the present study were and 11 lower-order scales (Studerus et al. 2010). The 5D-
within the range of doses that are taken for recreational ASC dimension “Oceanic Boundlessness” (27 items)

37
Psychopharmacology

measures derealization and depersonalization associated with Results


positive emotional states, ranging from heightened mood to
euphoric exaltation. The corresponding lower-order scales in- Mystical-type experiences
clude “experience of unity,” “spiritual experience,” “blissful
state,” and “insightfulness.” The dimension “Anxious Ego LSD (200 μg) significantly increased all MEQ scores com-
Dissolution” (21 items) summarizes ego disintegration and pared with placebo (Fig. 1a, Table 1). The effects of MDMA
loss of self-control phenomena associated with anxiety. The and methylphenidate on MEQ scores are included for com-
corresponding lower-order scales include “disembodiment,” parison (Fig. 1a). The effects of LSD (200 μg) and placebo on
“impaired control of cognition,” and “anxiety.” The dimen- MEQ scores in 11 patients during LSD-assisted psychothera-
sion “Visionary Restructuralization” (18 items) consists of the py (Gasser et al. 2014) are also shown in Fig. 1b. LSD-
lower-order scales “complex imagery,” “elementary imagery,” induced mystical experiences were comparable in healthy
“audio-visual synesthesia,” and “changed meaning of per- subjects in the laboratory setting in the present study and in
cepts.” Two additional dimensions describe “Auditory patients in the therapeutic setting (Fig. 1b). Only two subjects
Alterations” (15 items) and “Reduction of Vigilance” (12 in each of the studies had a complete mystical experience. The
items). The scale is well-validated and widely used to charac- MEQ30 total scores were <5 % in both settings after placebo
terize the subjective effects of various psychedelic drugs administration (Fig. 1b).
(Carhart-Harris et al. 2016b; Hasler et al. 2004; Hysek et al.
2011; Schmid et al. 2015; Vollenweider et al. 2007;
Vollenweider and Kometer 2010). In addition to the subscale Alterations of consciousness
analyses, we also analyzed the effects on ego dissolution item
71 (the boundaries between myself and my surroundings LSD induced pronounced peak alterations of waking con-
seemed to blur) because the concept of ego dissolution was sciousness, with significant increases in all dimensions and
often used in recent imaging studies (Tagliazucchi et al. 2016). subscales of the 5D-ASC scale (Fig. 2). The 200 μg dose of
The 5D-ASC scale was administered 24 h after drug admin- LSD produced significantly greater scores on the overall ASC
istration, and the participants were asked to retrospectively scale, the dimension of visionary restructuralization, and the
rate the drug effects. 5D-ASC ratings were also performed at blissful state, insightfulness, and changed meaning of percepts
3 and 10 h in study 1. subscales compared with the 100 μg dose (Fig. 2, Table 1). The
mean ± SEM ego dissolution (item 71) scores were 49 ± 6 and
Analysis of plasma LSD concentrations 53 ± 10 after the 100 and 200 μg doses, respectively (Table 1).
There were only minimal differences between the 5D-ASC
Blood was collected into lithium heparin tubes before and 0.5, ratings at 3, 10, and 24 h (supplementary Fig. S1 online).
1, 1.5, 2.5, 3, 4, 6, 8, 10, 12, 16, and 24 h after LSD admin-
istration. The 0.5, 1.5, and 2.5 h samples were not collected in Plasma LSD concentrations
study 1. Blood samples were immediately centrifuged, and the
plasma was rapidly stored at −20 °C and later analyzed using Plasma concentrations varied between subjects, especially at
liquid-chromatography-tandem mass-spectrometry as previ- the lower 100 μg dose. The median (range) Cmax values were
ously reported (Dolder et al. 2015a; Steuer et al. 2016). 1.4 ng/ml (0.32–3.7) and 3.2 ng/ml (1.9–7.1) for the 100 and
Maximal plasma concentrations (Cmax) and total exposure (ar- 200 μg doses, respectively. The corresponding AUC values
ea under the plasma concentration-time curve [AUC]) were were 8.5 ng × h/ml (1–19) and 20.7 ng × h/ml (11–39).
estimated using compartmental modeling in Phoenix
WinNonlin 6.4 (Certara, Princeton, NJ, USA). A one-
compartment model was used with first-order input, first- Associations between alterations of consciousness
order elimination, and no lag time. and mystical-type experiences

Table 2 shows the cross-tabulation of all correlations between


Statistical analyses the 5D-ASC scale and MEQ30 subscale ratings. LSD-induced
alterations of consciousness (ASC total score) were significant-
The data analysis was performed using Statistica 12 software ly correlated with ratings of mystical experience (MEQ30 total
(StatSoft, Tulsa, OK, USA). Differences between LSD and score) on the MEQ (Rp = 0.87, p < 0.001, n = 16; Fig. 3).
placebo or between the 100 and 200 μg doses of LSD were Scores on the MEQ positive mood scale were strongly associ-
compared using dependent or independent t tests, respectively. ated with scores on the ASC experience of unity and blissful
Associations between outcome measures were assessed using state scales (Rp = 0.85 and 0.80, respectively; both p < 0.001,
Pearson correlations. Significance was assumed at p < 0.05. n = 16; Table 2).

38
Psychopharmacology

Fig. 1 Effects of LSD on the Mystical Experience Questionnaire (MEQ). Schmid et al. 2014). Similar to the present study, the MEQ was
a In the present study in healthy subjects, LSD (200 μg) significantly administered on the day after LSD (200 μg) or active placebo (25 μg
increased scores on all scales of the MEQ43 and MEQ30 compared with LSD) administration and was embedded into the larger 100-item States of
placebo (Table 1). The data are expressed as the mean ± SEM in 16 Consciousness Questionnaire (SOCQ; Griffiths et al. 2006). The patient
subjects. For comparison, 3,4-methylenedioxymethamphetamine data are expressed as the mean ± SEM in 11 subjects for LSD (200 μg,
(MDMA; 75 mg) and methylphenidate (40 mg) produced small same formulation as in the present study) and four subjects for placebo.
increases in MEQ ratings in 30 different participants in another study in On the 43- and 30-item versions of the MEQ, LSD (200 μg) increased
the same research setting (Schmid et al. 2014). b Effects of LSD on the MEQ rating scores in the patients in the therapeutic setting (b) to a similar
MEQ in patients with anxiety in the context of life-threatening illness. extent as in the healthy subjects in the present study (a). Notably, the
The data were analyzed identically to the data that were obtained in the placebo response (a very low dose of LSD of 25 μg was used as the
present study. The study and patient characteristics have been previously active placebo) in the patients was small (b), which was also similar to
published in detail (Diesch 2015; Gasser et al. 2014; Gasser et al. 2015; the response in healthy subjects in the present study (a)

Correlations between plasma LSD concentrations groups (Table 3). For example, LSD induced consistently
and LSD-induced alterations of consciousness high ratings of audio-visual synesthesia in almost all of
and mystical-type experiences the subjects at the high dose (200 μg), resulting in little
within-subject variance and no association with plasma
The Cmax and AUC values for LSD were not positively exposure to LSD (Table 3, Fig. 4a). One exception was
correlated with ratings of peak subjective effects on the ego dissolution (item 71) at the lower dose of LSD
5D-ASC scale or MEQ across subjects or within dose (100 μg; Table 3, Fig. 4b). The ratings showed high

39
Psychopharmacology

Table 1 Statistics for the effects


of LSD in the 5D-ASC and MEQ LSD 100 μg LSD 200 μg LSD 100 vs. 200 μg
T test vs. placebo T test vs. placebo T test

T= P= T= P= T= P=

5 Dimensions Altered States of Consciousness (ASC) scale


Total ASC score 9.72 <0.001 10.02 <0.001 2.23 <0.05
Oceanic boundlessness 8.44 <0.001 9.61 <0.001 1.89 NS
Anxious ego dissolution 6.43 <0.001 4.01 <0.001 1.50 NS
Visionary restructuralization 9.79 <0.001 15.32 <0.001 2.34 <0.05
Auditory alterations 3.72 <0.01 5.87 <0.001 0.42 NS
Reductions of vigilance 7.44 <0.001 5.93 <0.001 0.79 NS
Experience of unity 6.85 <0.001 7.77 <0.001 0.68 NS
Spiritual experience 4.31 <0.001 3.91 <0.001 1.10 NS
Blissful state 6.56 <0.001 8.27 <0.001 3.00 <0.01
Insightfulness 4.11 <0.001 5.81 <0.001 2.28 <0.05
Disembodiment 6.93 <0.001 5.87 <0.001 0.13 NS
Impaired control and cognition 7.01 <0.001 5.04 <0.001 0.86 NS
Anxiety 3.02 <0.001 2.04 NS 1.37 NS
Complex imagery 7.10 <0.001 7.48 <0.001 0.31 NS
Elementary imagery 9.96 <0.001 11.12 <0.001 0.57 NS
Audio-visual synsthesia 9.19 <0.001 12.52 <0.001 1.96 NS
Changed meaning of percepts 6.25 <0.001 9.66 <0.001 3.39 <0.01
Ego dissolution (item 71) 7.63 <0.001 5.32 <0.001 0.36 NS
Mystical Effects Questionnaire (MEC43)
Internal unity NA NA 6.22 <0.001 NA NA
External unity NA NA 6.08 <0.001 NA NA
Sacredness NA NA 6.80 <0.001 NA NA
Noetic quality NA NA 5.71 <0.001 NA NA
Deeply felt positive mood NA NA 11.43 <0.001 NA NA
Transcendence of time/space NA NA 10.63 <0.001 NA NA
Ineffability NA NA 16.22 <0.001 NA NA
Mystical Effects Questionnaire (MEQ30)
Mystical NA NA 5.99 <0.001 NA NA
Positive mood NA NA 13.13 <0.001 NA NA
Transcendence of time/space NA NA 11.12 <0.001 NA NA
Ineffability NA NA 25.14 <0.001 NA NA
MEC30 total score NA NA 14.91 <0.001 NA NA

Sixteen subjects participated in the high-dose study (200 μg) and 24 subjects in the moderate-dose study (100 μg).
Dependent T tests were performed to assess differences from placebo, and independent T tests were performed to
assess differences between doses of LSD
NA not assessed

interindividual variance, and there was a significant pos- Discussion


itive correlation with the LSD AUC value in the 100 μg
dose group (R p = 0.51, p < 0.05, n = 16; Table 3, The present study characterized LSD-induced mystical
Fig. 4b). At the 200 μg dose, there were significant neg- experiences using the MEQ after a dose of 200 μg
ative correlations between Cmax values for LSD and sub- and alterations of consciousness on the 5D-ASC scale
jective effects on the 5D-ASC scale including visionary after a dose of 100 μg. The study also evaluated asso-
restructuralization, elementary imagery, and changed ciations between plasma LSD concentrations and these
meaning of percepts. subjective effects.

40
Psychopharmacology

Fig. 2 Effects of LSD on the 5 Dimensions of Altered States of the 5D-ASC scale were significant compared with placebo for both doses
Consciousness (5D-ASC) scale. LSD mainly increased ratings of oceanic and all of the scales, with the exception of the effects of the 200 μg dose on
boundlessness (OB) and visionary restructuralization (VR), with anxiety (Table 1). At 200 μg, LSD produced significant and relevantly
significantly higher ratings for the ASC total score and VR dimension at higher ratings of blissful state, insightfulness, and changed meaning of
200 μg compared with 100 μg. LSD-induced increases in anxious ego percepts compared with 100 μg (one asterisk p < 0.05, two asterisks
dissolution (AED) and auditory alterations (AA) were relatively small. p < 0.01, t tests). The data are expressed as the mean ± SEM in 24
LSD also produced vigilance reduction (VIR). LSD-induced changes on subjects and 16 subjects for the 100 and 200 μg doses of LSD, respectively

LSD produced mean MEQ30 total score ratings of 61 % this therapeutic setting, LSD produced similar mystical expe-
(range 40–98 %) and a complete mystical experience in only riences as in the present study and complete mystical experi-
two participants (12.5 %). The MEQ has typically been used ences in only two of 11 patients. MEQ scores were only within
with psilocybin, and data on MEQ30 scores are available for the range of 3–9 % after active placebo administration (25 μg
various doses of psilocybin, placebo, and methylphenidate LSD) on the MEQ subscales. Altogether, these findings indi-
(active placebo; Barrett et al. 2015). Psilocybin (at the highest cate that mainly the placebo response and/or the expectancy of
studied dose of 30 mg/70 kg) produced a high mean MEQ30 a mystical experience were greater in the study setting in some
total score rating of 77 % and complete mystical experiences psilocybin studies compared with the LSD studies.
in as many as 67 % of healthy subjects (Barrett et al. 2015). Additionally, the participants in the psilocybin studies may
However, in this psilocybin study setting, inactive and active have been more spiritually inclined (Griffiths et al. 2006) than
placebo (methylphenidate) also produced high mean MEQ30 our study participants leading to more mystical experiences
ratings of 23 and 33 %, respectively (Barrett et al. 2015). In (Studerus et al. 2012). Furthermore, others may have provided
contrast, in the present study, placebo increased MEQ30 more extensive preparation of the subjects and interpersonal
scores only to 1 %. Similarly, MDMA and methylphenidate support, contributing to mystical experiences.
produced only small increases in MEQ scores in a similar The present findings do not support the view that LSD
laboratory setting (Schmid et al. 2014). Another study evalu- produces lower overall effects than psilocybin at the doses
ated psilocybin-assisted psychotherapy in tobacco smokers tested. In contrast, the high dose of LSD (200 μg) produced
and also found complete mystical experiences in only 10 of greater placebo-adjusted positive mood ratings than psilocy-
26 sessions (38 %) that were conducted in 14 patients with bin on the MEQ30 (Barrett et al. 2015) and very pronounced
high-dose psilocybin (30 mg/70 kg; Garcia-Romeu et al. increases in 5D-ASC blissful state ratings and produced far
2015; Johnson et al. 2014). Accounting for the higher placebo greater effects than the highest doses of psilocybin or dimeth-
ratings in some of the psilocybin studies compared with our yltryptamine (DMT) that were tested so far on this scale
study, LSD increased MEQ30 score differences from placebo (Gouzoulis-Mayfrank et al. 2005; Hasler et al. 2004).
overall more than psilocybin and produced greater ineffability Additionally, LSD-induced MEQ scores were highly correlat-
and positive mood but lower effects on the mystical subscale ed with 5D-ASC scores in the present study.
than psilocybin (Barrett et al. 2015). One could argue that mystical and spiritual experiences are
Additionally, the MEQ has been used in patients with anx- not the most prominent feature of the LSD response. Mean
iety associated with life-threatening illness who were treated ratings on the spiritual experience scale of the 5D-ASC were
with 200 μg LSD (Gasser et al. 2014; Gasser et al. 2015). In 22 and 33 % at the 100 and 200 μg doses, respectively, in the

41
Psychopharmacology

Table 2 Associations between


LSD-induced alterations in Mystical Effects Questionnaire (MEQ30)
consciousness (5D-ASC) and
mystical experiences (MEQ30) MEQ30 total Mystical Positive Transcendence of Ineffability
score mood time/space

5D-ASC scale
Total ASC score 0.87 0.73 0.65 0.82 0.57
Oceanic boundlessness 0.93 0.88 0.83 0.74 0.45
Anxious ego 0.60 0.39 0.35 0.68 0.55
dissolution
Visionary 0.65 0.54 0.38 0.68 0.45
restructuralization
Auditory alterations 0.30 0.14 0.02 0.49 0.38
Reductions of 0.61 0.41 0.47 0.64 0.47
vigilance
Experience of unity 0.82 0.86 0.85 0.56 0.25
Spiritual experience 0.79 0.76 0.76 0.60 0.33
Blissful State 0.80 0.77 0.80 0.72 0.16
Insightfulness 0.77 0.79 0.68 0.52 0.42
Disembodiment 0.71 0.53 0.62 0.71 0.41
Impaired control and 0.63 0.37 0.45 0.79 0.46
cognition
Anxiety 0.45 0.32 0.19 0.47 0.51
Complex imagery 0.48 0.31 0.32 0.69 0.19
Elementary imagery 0.36 0.37 0.08 0.29 0.42
Audio-visual 0.23 0.07 0.22 0.45 −0.01
synesthesia
Changed meaning of 0.80 0.67 0.59 0.70 0.63
percepts
Ego dissolution (item 0.74 0.73 0.74 0.65 0.12
71)

Values are Pearson correlation coefficients in 16 subjects describing correlations between %5D-ASC and
%MEQ30 scores. Bold values for P < 0.05, italic values for P < 0.001

present study and approximately 23 % after 75 μg LSD in


another study (Carhart-Harris et al. 2016c). Mean ratings of
“the experience had a spiritual or mystical quality” were also
only approximately 28 % in an imaging study that evaluated
the effects of LSD (Tagliazucchi et al. 2016). However, a
direct within-subjects comparison of LSD and psilocybin
in the same research setting is needed to determine possible
differences in mystical-type responses between these sub-
stances. Whether mystical-type experiences (Barrett et al.
2015; Garcia-Romeu et al. 2015; MacLean et al. 2011) are
critical for the therapeutic potential of substance-assisted
psychotherapy requires further study. At least in the case
of LSD, the mystical experiences (MEQ scores) were high-
ly associated with other alterations of consciousness on the
5D-ASC scale, and LSD produced additional effects on
Fig. 3 LSD-induced alterations of consciousness are significantly emotion processing that could facilitate psychotherapeutic
associated with the LSD-induced mystical experience. The data are interventions (Dolder et al. 2016).
expressed as a percentage of ASC total scores on the 5D-ASC scale and
a percentage of total scores on the MEQ30 for each of 16 participants after
Recent experimental studies associated the subjective ef-
administration of 200 μg LSD. The lines indicate the regression and 95 % fects of LSD (75 μg, intravenous) on the 5D-ASC scale with
confidence intervals (Rp = 0.87, p < 0.001) fMRI data but in the absence of data on plasma LSD levels

42
Psychopharmacology

Table 3 Associations between predicted maximal LSD plasma


concentrations (Cmax) and LSD exposure (AUC) and alterations in
consciousness (SD-ASC) and mystical experiences (MEQ30)

N = 24 N = 16

100 μg 200 μg

Cmax AUC Cmax AUC

5D-ASC scale
ASC total score 0.19 0.21 −0.35 0.15
Oceanic boundlessness 0.24 0.26 −0.35 0.10
Anxious ego dissolution 0.04 0.07 −0.10 0.32
Visionary restructuralization 0.12 0.15 −0.59 −0.16
Auditory alterations 0.02 0.12 −0.18 0.08
Reductions of vigilance −0.01 0.13 −0.10 0.38
Experience of unity 0.34 0.33 −0.03 0.33
Spiritual experience −0.02 0.06 −0.32 −0.03
Blissful state 0.25 0.14 −0.23 0.03
Insightfulness 0.24 0.20 −0.37 0.12
Disembodiment −0.04 0.08 −0.23 0.08
Impaired control and cognition −0.01 0.01 −0.20 0.18
Anxiety 0.22 0.30 0.01 0.38
Complex imagery 0.06 0.14 −0.28 −0.04
Elementary imagery −0.13 −0.03 −0.53 −0.15 Fig. 4 Correlations between plasma LSD concentrations and subjective
peak effects. a At 200 μg, LSD induced high ratings of audio-visual
Audio-visual synesthesia 0.23 0.26 −0.01 0.00 synesthesia in all but two of the 16 participants. There was little
Changed meaning of percepts −0.03 −0.06 −0.62 −0.10 variance in the response and no correlation between total plasma
Ego dissolution (item 71) 0.40 0.51 −0.27 −0.14 exposure to LSD (area under the concentration-time curve [AUC]) and
MEQ30 audio-visual synesthesia (Rp = 0.0, p > 0.05, n = 16). b In contrast, ego
dissolution was present to highly variable degrees across subjects after
MEC30 total score NA −0.30 0.17 administration of 100 μg LSD. Total exposure to LSD (AUC) positively
Mystical NA −0.25 0.13 correlated with LSD-induced ego dissolution (Rp = 0.51, p < 0.05,
Positive mood NA −0.08 0.21 n = 24). The lines indicate the regression and 95 % confidence intervals
Transcendence of time/space NA −0.23 0.10
Ineffability NA −0.49 0.13
changed meaning of percepts. As previously reported, the
Values are Pearson correlation coefficients describing correlations, the 200 μg dose of LSD also produced greater feelings of close-
peak concentrations of LSD predicted by the one-compartment model, ness to others, happiness, openness, and trust than the 100 μg
and LSD-induced %5D-ASC and %MEQ30 scores. Bold values for
P < 0.05Cmax maximal LSD plasma concentration predicted by the one- dose (Dolder et al. 2016). Altogether, the data indicate that the
compartment pharmacokinetic model, AUC area under the LSD 200 μg dose produces overall greater effects and particularly
concentration-time curve predicted by the model more positive and MDMA-like effects than lower doses
(Dolder et al. 2016). This is relevant because the higher dose
is currently being used in LSD-assisted psychotherapy
(Carhart-Harris et al. 2016c; Kaelen et al. 2016; Lebedev et al. (Gasser et al. 2014; Gasser et al. 2015), and the lower dose
2016; Roseman et al. 2016). Assuming high oral bioavailabil- is being tested in experimental fMRI studies (Carhart-Harris
ity of LSD of 70–100 % (Dolder et al. 2015b), similar plasma et al. 2016c). The 200 μg dose of LSD also produced greater
exposure (AUC) can be assumed after oral administration of ASC scores than high doses of the serotonergic hallucinogens
100 μg LSD (present study I) or intravenous administration of DMT and psilocybin (Gouzoulis-Mayfrank et al. 2005; Hasler
75 μg LSD (all studies by Carhart-Harris and colleagues). et al. 2004; Vollenweider and Kometer 2010), ketamine
Supporting this assumption, the intravenous 75 μg dose of (Gouzoulis-Mayfrank et al. 2005; Studerus et al. 2010), and
LSD produced very similar mean ratings on the 5D-ASC scale MDMA (Hysek et al. 2011), although direct comparisons
(Carhart-Harris et al. 2016b) to the present study that used an within the same studies and subjects are missing.
oral dose of 100 μg. In contrast, the 200 μg dose produced The present analyses showed no positive correlations be-
significantly greater ASC total scores and particularly greater tween LSD levels and effects across subjects, possibly be-
5D-ASC subscale scores of blissful state, insightfulness, and cause of the relatively high levels of LSD and generally

43
Psychopharmacology

consistently high subjective response ratings in most subjects. mystical experiences. LSD produced significantly greater
Thus, if relatively high and similar doses of LSD are used that bliss, insightfulness, and changes in meaning of percepts at
result in plasma levels clearly above the EC50 of a particular 200 μg compared with 100 μg, in addition to the previously
response measure, then it is unlikely that the response varies reported greater empathogenic effects. This could be relevant
relevantly across subjects because responses are close to max- for LSD-assisted psychotherapy (200 μg) and the interpreta-
imal. This would typically also be the case with measures with tion of fMRI data (75–100 μg). Generally, no association was
a maximal effect limit such as VAS ratings and some physio- found between plasma LSD levels and its robust effects when
logical effects like pupil size (Hysek and Liechti 2012). analyzed across different subjects and within a dose group.
In fact, responses to MDMA or LSD or other drugs in a This may have implications for studies that interrelate differ-
standardized experimental setting may vary only if the re- ent effects of LSD, namely fMRI studies.
sponse is not induced consistently in all subjects (e.g., at the
beginning of the response) and are mostly attributable to indi- Acknowledgments The authors thank Dr. Peter Gasser for providing
MEQ data in patients and Michael Arends for text editing. MEL dedicat-
vidual differences in drug absorption/distribution (Hysek and
ed this work to Dr. Athina Markou, and thanks Drs. Athina Markou and
Liechti 2012) or when a response is evaluated that is not ro- Mark A. Geyer for their sincere support and mentorship.
bustly induced or when a lower dose is used. Specifically,
correlations of plasma levels with the subjective and cardio- Compliance with ethical standards The studies were conducted in
vascular effects of MDMA across subjects are only weak dur- accordance with the Declaration of Helsinki and approved by the local
ethics committee. All of the subjects provided written consent before
ing the peak response but stronger at onset (Hysek and Liechti
participating in either of the studies.
2012). This is an important consideration. For example, LSD-
induced subjective ego dissolution was recently shown to be Conflict of interest None.
associated with specific brain activation patterns in a study
that administered a relatively low dose of LSD of 75 μg intra- Funding This work was supported by the Swiss National Science
venously (Tagliazucchi et al. 2016). Interestingly, LSD- Foundation (grant no. 320030_170249 to MEL).
induced ego dissolution correlated with plasma LSD levels
after administration of an equivalent oral dose of 100 μg in
the present study, and this was the only pharmacodynamic Open Access This article is distributed under the terms of the Creative
effect of LSD for which a positive association with plasma Commons Attribution 4.0 International License (http://
levels could be demonstrated across subjects. This finding creativecommons.org/licenses/by/4.0/), which permits unrestricted use,
distribution, and reproduction in any medium, provided you give
needs to be kept in mind when interpreting associations be- appropriate credit to the original author(s) and the source, provide a link
tween ego dissolution and fMRI parameters because the fMRI to the Creative Commons license, and indicate if changes were made.
findings may also reflect other processes that are related to the
plasma levels of LSD. Furthermore, the likelihood of detect-
ing correlations within a dose group increases for effects that References
are not robustly induced in all subjects and thus for effects that
are not typically present in all subjects after LSD administra- Barrett FS, Johnson MW, Griffiths RR (2015) Validation of the revised
tion. Finally, unclear is the extent to which a full LSD re- mystical experience questionnaire in experimental sessions with psi-
sponse was induced in the imaging studies that have been locybin. J Psychopharmacol 29:1182–1190
Baumeister D, Barnes G, Giaroli G, Tracy D (2014) Classical hallucino-
conducted to date because all of these studies used relatively
gens as antidepressants? A review of pharmacodynamics and puta-
low 75 or 100 μg doses. In the present study, the 200 μg dose tive clinical roles. Ther Adv Psychopharmacol 4:156–169
of LSD produced particularly marked increases in visionary Bogenschutz MP, Forcehimes AA, Pommy JA, Wilcox CE, Barbosa PC,
restructuralization including changed meaning of percepts Strassman RJ (2015) Psilocybin-assisted treatment for alcohol depen-
dence: a proof-of-concept study. J Psychopharmacol 29:289–299
which were significantly greater after the 200 compared with
Carhart-Harris RL, Kaelen M, Whalley MG, Bolstridge M, Feilding A,
the 100 μg dose. Contrary to expectations, these perceptual Nutt DJ (2015) LSD enhances suggestibility in healthy volunteers.
alterations were greater in participants with relatively lower Psychopharmacology 232:785–794
Cmax levels of LSD within the 200 μg dose group further Carhart-Harris RL, Bolstridge M, Rucker J, Day CM, Erritzoe D, Kaelen
supporting the view that higher plasma levels of LSD may M, Bloomfield M, Rickard JA, Forbes B, Feilding A, Taylor D,
Pilling S, Curran VH, Nutt DJ (2016a) Psilocybin with psycholog-
not produce greater subjective alterations above a certain ical support for treatment-resistant depression: an open-label feasi-
threshold level and if high doses of LSD are used. bility study. Lancet Psychiatry 3:619–627
In conclusion, LSD (200 μg) rarely produced full mystical Carhart-Harris RL, Kaelen M, Bolstridge M, Williams TM, Williams LT,
experiences in the present study and in patients during LSD- Underwood R, Feilding A, Nutt DJ (2016b) The paradoxical psy-
chological effects of lysergic acid diethylamide (LSD). Psychol Med
assisted psychotherapy compared with psilocybin in another 46:1379–1390
set and setting. This raises questions regarding expectancy Carhart-Harris RL, Muthukumaraswamy S, Roseman L, Kaelen M,
effects and placebo responses and the therapeutic role of Droog W, Murphy K, Tagliazucchi E, Schenberg EE, Nest T,

44
Psychopharmacology

Orban C, Leech R, Williams LT, Williams TM, Bolstridge M, Sessa Hysek CM, Simmler LD, Ineichen M, Grouzmann E, Hoener MC,
B, McGonigle J, Sereno MI, Nichols D, Hellyer PJ, Hobden P, Brenneisen R, Huwyler J, Liechti ME (2011) The norepinephrine
Evans J, Singh KD, Wise RG, Curran HV, Feilding A, Nutt DJ transporter inhibitor reboxetine reduces stimulant effects of MDMA
(2016c) Neural correlates of the LSD experience revealed by multi- ("ecstasy") in humans. Clin Pharmacol Ther 90:246–255
modal neuroimaging. Proc Natl Acad Sci U S A 113:4853–4858 Johnson MW, Garcia-Romeu A, Cosimano MP, Griffiths RR (2014) Pilot
Davenport WJ (2016) Psychedelic and nonpsychedelic LSD and psilocy- study of the 5-HT2AR agonist psilocybin in the treatment of tobacco
bin for cluster headache. CMAJ 188:217 addiction. J Psychopharmacol 28:983–992
Diesch MK (2015) LSD: Rückkehr in the Klinische Forschung. Kaelen M, Barrett FS, Roseman L, Lorenz R, Family N, Bolstridge M,
Nachtschatten Verlag, Solothurn Curran HV, Feilding A, Nutt DJ, Carhart-Harris RL (2015) LSD
Dittrich A (1998) The standardized psychometric assessment of altered enhances the emotional response to music. Psychopharmacology
states of consciousness (ASCs) in humans. Pharmacopsychiatry 232:3607–3614
31(Suppl 2):80–84 Kaelen M, Roseman L, Kahan J, Santos-Ribeiro A, Orban C, Lorenz R,
Dolder PC, Liechti ME, Rentsch KM (2015a) Development and valida- Barrett FS, Bolstridge M, Williams T, Williams L, Wall MB,
tion of a rapid turboflow LC-MS/MS method for the quantification Feilding A, Muthukumaraswamy S, Nutt DJ, Carhart-Harris R
of LSD and 2-oxo-3-hydroxy LSD in serum and urine samples of (2016) LSD modulates music-induced imagery via changes in
emergency toxicological cases. Anal Bioanal Chem 407:1577–1584 parahippocampal connectivity. Eur Neuropsychopharmacol 26:
Dolder PC, Schmid Y, Haschke M, Rentsch KM, Liechti ME (2015b) 1099–1109
Pharmacokinetics and concentration-effect relationship of oral LSD Krebs TS, Johansen PO (2012) Lysergic acid diethylamide (LSD) for
in humans. Int J Neuropsychopharmacol 19. doi:10.1093 alcoholism: meta-analysis of randomized controlled trials. J
/ijnp/pyv072 Psychopharmacol 26:994–1002
Dolder PC, Schmid Y, Mueller F, Borgwardt S, Liechti ME (2016) LSD Krebs TS, Johansen PO (2013) Over 30 million psychedelic users in the
acutely impairs fear recognition and enhances emotional empathy United States. F1000Res 2:98
and sociality. Neuropsychopharmacology 41:2638–2646 Kupferschmidt K (2014) High hopes. Science 345:18–23
Garcia-Romeu A, Griffiths RR, Johnson MW (2015) Psilocybin- Lebedev AV, Kaelen M, Lovden M, Nilsson J, Feilding A, Nutt DJ,
occasioned mystical experiences in the treatment of tobacco addic- Carhart-Harris RL (2016) LSD-induced entropic brain activity pre-
tion. Curr Drug Abuse Rev 7:157–164 dicts subsequent personality change. Hum Brain Mapp 37:3203–
Gasser P, Holstein D, Michel Y, Doblin R, Yazar-Klosinski B, Passie T, 3213
Brenneisen R (2014) Safety and efficacy of lysergic acid MacLean KA, Johnson MW, Griffiths RR (2011) Mystical experiences
diethylamide-assisted psychotherapy for anxiety associated with occasioned by the hallucinogen psilocybin lead to increases in the
life-threatening diseases. J Nerv Ment Dis 202:513–520 personality domain of openness. J Psychopharmacol 25:1453–1461
Gasser P, Kirchner K, Passie T (2015) LSD-assisted psychotherapy for anx-
MacLean KA, Leoutsakos JM, Johnson MW, Griffiths RR (2012) Factor
iety associated with a life-threatening disease: a qualitative study of
analysis of the Mystical Experience Questionnaire: a study of expe-
acute and sustained subjective effects. J Psychopharmacol 29:57–68
riences occasioned by the hallucinogen psilocybin. J Sci Study Relig
Gouzoulis-Mayfrank E, Heekeren K, Neukirch A, Stoll M, Stock C,
51:721–737
Obradovic M, Kovar KA (2005) Psychological effects of (S)-keta-
Nichols DE (2016) Psychedelics. Pharmacol Rev 68:264–355
mine and N,N-dimethyltryptamine (DMT): a double-blind, cross-
over study in healthy volunteers. Pharmacopsychiatry 38:301–311 Pahnke WN (1969) Psychedelic drugs and mystical experience. Int
Griffiths R (2016) Overview of the Johns Hopkins psilocybin research Psychiatry Clin 5:149–162
project. Interdisciplinary Conference on Psychedelics Research, Pahnke WN, Kurland AA, Unger S, Savage C, Grof S (1970) The exper-
Amsterdam, June 3–5, 2016 imental use of psychedelic (LSD) psychotherapy. JAMA 212:1856–
Griffiths RR, Richards WA, McCann U, Jesse R (2006) Psilocybin can 1863
occasion mystical-type experiences having substantial and sustained Passie T, Halpern JH, Stichtenoth DO, Emrich HM, Hintzen A (2008)
personal meaning and spiritual significance. Psychopharmacology The pharmacology of lysergic acid diethylamide: a review. CNS
187:268–283 discussion 284-292 Neurosci Ther 14:295–314
Griffiths R, Richards W, Johnson M, McCann U, Jesse R (2008) Rickli A, Moning OD, Hoener MC, Liechti ME (2016) Receptor inter-
Mystical-type experiences occasioned by psilocybin mediate the action profiles of novel psychoactive tryptamines compared with
attribution of personal meaning and spiritual significance 14 months classic hallucinogens. Eur Neuropsychopharmacol 26:1327–1337
later. J Psychopharmacol 22:621–632 Roseman L, Sereno MI, Leech R, Kaelen M, Orban C, McGonigle J,
Griffiths RR, Johnson MW, Richards WA, Richards BD, McCann U, Jesse Feilding A, Nutt DJ, Carhart-Harris RL (2016) LSD alters eyes-
R (2011) Psilocybin occasioned mystical-type experiences: immediate closed functional connectivity within the early visual cortex in a
and persisting dose-related effects. Psychopharmacology 218:649–665 retinotopic fashion. Hum Brain Mapp 37:3031–3040
Grob CS, Danforth AL, Chopra GS, Hagerty M, McKay CR, Halberstadt Schmid Y, Hysek CM, Simmler LD, Crockett MJ, Quednow BB, Liechti
AL, Greer GR (2011) Pilot study of psilocybin treatment for anxiety ME (2014) Differential effects of MDMA and methylphenidate on
in patients with advanced-stage cancer. Arch Gen Psych 68:71–78 social cognition. J Psychopharmacol 28:847–856
Guss J (2016) The NYU School of Medicine study on psilocybin-assisted Schmid Y, Enzler F, Gasser P, Grouzmann E, Preller KH, Vollenweider
therapy for treatment of existential distress in cancer patients: histo- FX, Brenneisen R, Muller F, Borgwardt S, Liechti ME (2015) Acute
ry, study structure, therapist training, outcome data. Interdisciplinary effects of lysergic acid diethylamide in healthy subjects. Biol
Conference on Psychedelic Research Amsterdam 3–5 June, 2016 Psychiatry 78:544–553
Hasler F, Grimberg U, Benz MA, Huber T, Vollenweider FX (2004) Speth J, Speth C, Kaelen M, Schloerscheidt AM, Feilding A, Nutt DJ,
Acute psychological and physiological effects of psilocybin in Carhart-Harris RL (2016) Decreased mental time travel to the past
healthy humans: a double-blind, placebo-controlled dose-effect correlates with default-mode network disintegration under lysergic
study. Psychopharmacology 172:145–156 acid diethylamide. J Psychopharmacol 30:344–353
Hysek CM, Liechti ME (2012) Effects of MDMA alone and after Steuer AE, Poetzsch M, Stock L, Eisenbeiss L, Schmid Y, Liechti ME,
pretreatement with reboxetine, duloxetine, clonidine, carvedilol, Kraemer T (2016) Development and validation of an ultra-fast and
and doxazosin on pupillary light reflex. Psychopharmacology 224: sensitive microflow liquid chromatography-tandem mass spectrom-
363–376 etry (MFLC-MS/MS) method for quantification of LSD and its

45
Psychopharmacology

metabolites in plasma and application to a controlled LSD adminis- correlates with LSD-induced ego dissolution. Curr Biol 26:1043–
tration study in humans. Drug Test Anal. doi:10.1002/dta.2042 1050
Strajhar P, Schmid Y, Liakoni E, Dolder PC, Rentsch KM, Kratschmar Terhune DB, Luke DP, Kaelen M, Bolstridge M, Feilding A, Nutt D,
DV, Odermatt A, Liechti ME (2016) Acute effects of lysergic acid Carhart-Harris R, Ward J (2016) A placebo-controlled investigation
diethylamide on circulating steroid levels in healthy subjects. J of synaesthesia-like experiences under LSD. Neuropsychologia 88:
Neuroendocrinol 28:12374 28–34
Studerus E, Gamma A, Vollenweider FX (2010) Psychometric evaluation Turek IS, Soskin RA, Kurland AA (1974) Methylenedioxyamphetamine
of the altered states of consciousness rating scale (OAV). PLoS One (MDA)-subjective effects. J Psychoactive Drugs 6:7–14
5:e12412 Vollenweider FX, Kometer M (2010) The neurobiology of psychedelic
Studerus E, Gamma A, Kometer M, Vollenweider FX (2012) Prediction drugs: implications for the treatment of mood disorders. Nat Rev
of psilocybin response in healthy volunteers. PLoS One 7:e30800 Neurosci 11:642–651
Tagliazucchi E, Roseman L, Kaelen M, Orban C, Muthukumaraswamy Vollenweider FX, Csomor PA, Knappe B, Geyer MA, Quednow BB
SD, Murphy K, Laufs H, Leech R, McGonigle J, Crossley N, (2007) The effects of the preferential 5-HT2A agonist psilocybin
Bullmore E, Williams T, Bolstridge M, Feilding A, Nutt DJ, on prepulse inhibition of startle in healthy human volunteers depend
Carhart-Harris R (2016) Increased global functional connectivity on interstimulus interval. Neuropsychopharmacology 32:1876–
1887

46
Online Supplement:

ASC OB AED VR

100

P la c e b o
80 LSD 3h

LSD 10h
% s c a le m a x im u m

+
+ LSD 24h
#
+
60
*
* * * *
* * * *
* +
** + +
+ +
40
* *
** *
*

20

0
ASC AB AED VR A A V IR

ry
ity

ty

ts
ry
ss

e
ce

n
te

a
o

p
e

e
e
ta
n

si
iti
n

xi

e
g

g
im
ln
U

lS

e
n

rc
a

a
n
ri

h
g
d
f

Im

Im
A

e
e
o

tf
u

st
o
o
xp

P
sf

C
b
ce

yn
x

ry
g

f
lis

m
lE

le
si

o
d
n

ta

lS
e

p
B
e

In

g
is
a

n
la
ri

in
a
itu

e
D
e

u
o

n
o

m
xp

is
ir

C
tr

a
le
p

e
-V
n
E

E
o

M
io
C

d
d
d

e
u
e

g
A
ir

n
a

a
p

h
Im

C
Figure S1. Effects of LSD (100 µg) on the 5 Dimensions of Altered States of Consciousness
(5D-ASC) scale repeatedly used at 3, 10, and 24h to retrospectively rate the LSD effects in 24
subjects in Study 1. The aim of the repeated administration was to test whether self-ratings shortly
after the peak response (3h), at the end of the response (10h) or on the next day (at 24h) differ from
each other. We hypothesized that there would not be any relevant differences. ANOVA with time as
between-subject factor (3, 10, and 24 h) on the total ASC score showed a significant effect of time
(F2,46=5.50, P>0.01). Tukey post hoc tests showed higher ratings at 3h compared with 10 and 24h
(both P<0.01) but no differences between the 10 and 24h ratings. ANOVA with time and dimension (5
main dimensions) as factors showed a significant main effect of time (F 2,46=6.17, P<0.01) and scale
F4,92=19.87, P<0.001 and a significant time and dimension interaction (F 8,184=3.5, P<0.001). Tukey
post hoc test showed greater ratings at 3h compared with ratings at 10 h on all dimensions (all
P<0.01, Figure S1) and compared with ratings at 24 h for AED, AA, and VIR (all P<0.01). Ratings at
10 h did not differ from ratings at 24h with the exception of ratings for VR which were greater at 24 h
compared with 10 h (P<0.05). ANOVA with time and scale (all 11 scales of the 5D-ASC) showed no
significant main effect of time (F2,46=2.37, P=0.10), a significant main effect of scale (F 10,230=18.90,
P<0.001) and a significant time and scale interaction (F 20,460=1.81, P<0.05). Post hoc tests showed
that only ratings of “impaired control and cognition” were higher at 3h compared with 24h. There were
no other differences between the ratings at 3, 10, and 24 h. Together the data indicates higher ratings
of the overall effect when assessed during the response at 3h compared to ratings taken immediately
after the response or on the next day. However, the differences were minimal and not present
between ratings at 10 h and 24 h. OB, oceanic boundlessness; AED, anxious ego-dissolution; VR,
visionary restructuralization; AA, auditory alterations; VIR, vigilance reduction. **P<0.01 and
++ +++ #
***P<0.001 for 3h vs. 10h; P<0.01 and P<0.001 for 3h vs. 24h; P<0.05 for 10h vs. 24h (Tukey
post hoc tests based on significant time and scale interactions in the ANOVA). The data are expressed
as the mean ± SEM in 24 subjects.

47
3.4 Publication 4

LSD Acutely Impairs Fear Recognition and Enhances Emotional


Empathy and Sociality

Patrick C Dolder1, Yasmin Schmid1, Felix Müller2, Stefan J. Borgwardt2, Matthias E


Liechti1

1 Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department


of Clinical Research University Hospital and University of Basel, Basel, Switzerland.
2 Department of Psychiatry (UPK), University of Basel, Basel, Switzerland

48
Neuropsychopharmacology (2016) 41, 2638–2646 OPEN
Official journal of the American College of Neuropsychopharmacology

www.neuropsychopharmacology.org

LSD Acutely Impairs Fear Recognition and Enhances Emotional


Empathy and Sociality



Patrick C Dolder1, Yasmin Schmid1, Felix Müller2, Stefan Borgwardt2 and Matthias E Liechti*,1


1
Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of Clinical Research, University Hospital Basel,

Basel, Switzerland; 2Department of Psychiatry (UPK), University of Basel, Basel, Switzerland







Lysergic acid diethylamide (LSD) is used recreationally and has been evaluated as an adjunct to psychotherapy to treat anxiety in patients

with life-threatening illness. LSD is well-known to induce perceptual alterations, but unknown is whether LSD alters emotional processing

in ways that can support psychotherapy. We investigated the acute effects of LSD on emotional processing using the Face Emotion

Recognition Task (FERT) and Multifaceted Empathy Test (MET). The effects of LSD on social behavior were tested using the Social Value

Orientation (SVO) test. Two similar placebo-controlled, double-blind, random-order, crossover studies were conducted using 100 μg LSD

in 24 subjects and 200 μg LSD in 16 subjects. All of the subjects were healthy and mostly hallucinogen-naive 25- to 65-year-old volunteers


(20 men, 20 women). LSD produced feelings of happiness, trust, closeness to others, enhanced explicit and implicit emotional empathy on

the MET, and impaired the recognition of sad and fearful faces on the FERT. LSD enhanced the participants’ desire to be with other people

and increased their prosocial behavior on the SVO test. These effects of LSD on emotion processing and sociality may be useful for

LSD-assisted psychotherapy.

Neuropsychopharmacology (2016) 41, 2638–2646; doi:10.1038/npp.2016.82; published online 22 June 2016

INTRODUCTION and additionally improved mood at 6 months after treatment


in patients with advanced-stage cancer (Grob et al, 2011).
The classic serotonergic psychedelic/hallucinogen lysergic
Additionally, psilocybin was recently studied as a treatment
acid diethylamide (LSD) was widely studied in humans in
for tobacco (Johnson et al, 2014) and alcohol (Bogenschutz
the 1950s to 1970s. However, little to no clinical research on
et al, 2015) dependence. MDMA-assisted psychotherapy
LSD has been conducted since then (Nichols, 2016; Passie
reduced symptoms of post-traumatic stress disorder at
et al, 2008). Today, LSD is again the focus of clinical
2 months (Mithoefer et al, 2010), and the benefits of MDMA
investigations, including experimental studies in healthy
were reportedly sustained for several years (Mithoefer et al,
subjects (Carhart-Harris et al, 2016,2015; Dolder et al, 2015b;
2013). These first findings from modern clinical studies with
Schmid et al, 2015; Strajhar et al, 2016), and clinical trials that psychedelics and MDMA should be confirmed in larger
evaluate LSD-assisted psychotherapy (Gasser et al, 2014). trials. Exploring the mechanisms that may contribute to
LSD that was administered only a few times decreased these beneficial and lasting effects after only a few
anxiety and increased quality of life over a period of administrations of the substances is also important.
12 months in patients with anxiety associated with terminal Studies that use psychedelics and MDMA in healthy
illness (Gasser et al, 2015). The acute LSD experiences were subjects are well suited to assess the mechanism of action of
hypothesized to lead to a restructuring of the person's these substances. Both LSD and psilocybin appear to produce
emotional trust and situational understanding (Gasser effects that last beyond the acute drug response in both
et al, 2015). Similar to LSD, the serotonergic hallucinogen patients and healthy subjects. Specifically, LSD increased
psilocybin and serotonin (5-hydroxytryptamine (5-HT)) optimism and trait openness at 2 weeks (Carhart-Harris et al,
releaser 3,4-methylenedioxymethamphetamine (MDMA; 2016), and psilocybin produced positive changes in attitudes,
ecstasy) have been used to facilitate psychotherapy in mood, and behavior at 2 (Griffiths et al, 2006) and 14 months
clinical trials (Grob et al, 2011; Mithoefer et al, 2010; (Griffiths et al, 2011) after administration. Psilocybin
Oehen et al, 2013). Psilocybin reduced anxiety at 3 months increased personality trait openness in participants who
had ‘mystical experiences’ during their psilocybin session
*Correspondence: Professor ME Liechti, Department of Biomedicine (MacLean et al, 2011). Therefore, some of the lasting
and Department of Internal Medicine, Division of Clinical Pharmacology
and Toxicology, University Hospital Basel, Hebelstrasse 2, Basel CH-4031,
beneficial effects appear to be associated with an acute
Switzerland, Tel: +41 61 328 68 68, Fax: +41 61 265 45 60, psychedelic response, including a ‘peak’ or ‘mystical’
E-mail: [email protected] experience (Carhart-Harris et al, 2016; MacLean et al, 2011).
Received 4 March 2016; revised 26 April 2016; accepted 18 May 2016; Both LSD and psilocybin are 5-HT2A receptor agonists,
accepted article preview online 1 June 2016 and their psychedelic effects are mediated by 5-HT2A
49
LSD and emotion processing
PC Dolder et al
2639
receptor stimulation (Vollenweider et al, 1998). The long- were identical for both studies. Subjects younger than 25
term effects of LSD and psilocybin may be related to their years of age were excluded from participating in the study.
psychedelic and 5-HT2A receptor activation properties. In Additional exclusion criteria were age 465 years, pregnancy
contrast to the psychedelics LSD and psilocybin, MDMA (urine pregnancy test at screening and before each test
is considered an empathogen (entactogen) that mainly session), personal or family (first-degree relative) history of
enhances positive feelings, empathy, and prosociality major psychiatric disorders (assessed by the semistructured
(Hysek et al, 2014a; Kirkpatrick et al, 2014) while having clinical interview for Diagnostic and Statistical Manual of
few hallucinogen-like effects. Additionally, MDMA has been Mental Disorders, 4th edition, Axis I disorders by the study
shown to positively alter emotion processing (Bedi et al, physician and an additional interview by a trained psychia-
2010; Hysek et al, 2012,2014a; Kirkpatrick et al, 2012,2014; trist), use of medications that may interfere with the study
Schmid et al, 2014). These acute effects of MDMA on medication, chronic or acute physical illness (abnormal
emotion processing and social behavior may be beneficial physical exam, electrocardiogram, or hematological and
during psychotherapy in the absence of a full psychedelic chemical blood analyses), tobacco smoking (410 cigar-
peak experience. LSD also produced acute MDMA-like ettes/day), lifetime prevalence of illicit drug use 410 times
subjective effects, including greater well-being, happiness, (except for tetrahydrocannabinol), illicit drug use within the
closeness to others, openness, and trust (Schmid et al, 2015). last 2 months, and illicit drug use during the study
Thus, LSD and MDMA may have common effects on the (determined by urine drug tests). The subjects were asked
processing of emotional information with relevance to their to abstain from excessive alcohol consumption between test
positive acute and possibly long-term effects during sessions and particularly limit their use to one standard drink
psychotherapy. However, the effects of LSD in tests of on the day before the test sessions. Additionally, the
emotion processing are unknown. Therefore, the present participants were not allowed to drink xanthine-containing
study investigated the acute effects of LSD using the Face liquids after midnight before the study day. Eleven subjects
Emotion Recognition Task (FERT) and Multifaceted Em- had used a hallucinogen including LSD (6 participants) one
pathy Test (MET). The effects of LSD on social behavior to three times, and most of the subjects (29) were
were also evaluated using the Social Value Orientation (SVO) hallucinogen-naïve (Supplementary Table S1). We per-
test. Additionally, we assessed the subjective mood effects of formed urine drug tests at screening and before each test
LSD using Visual Analog Scales (VASs) and the Adjective session, and no substances were detected during the study.
Mood Rating Scale (AMRS), vital signs, and adverse effects.
We hypothesized that LSD would impair the recognition of Study Procedures
negative emotions on the FERT and enhance emotional
empathy on the MET and prosociality on the SVO test. Each study included a screening visit, a psychiatric interview,
two 25-h experimental sessions, and an end-of-study visit.
The experimental sessions were conducted in a quiet
MATERIALS AND METHODS standard hospital patient room. The participants were resting
Study Design in hospital beds except when going to the restroom. Only one
research subject and one investigator were present during
We pooled data from two similar studies using double-blind, the experimental sessions. Participants could interact with
placebo-controlled, crossover designs with two experimental the investigator, rest quietly and/or listen to music via
test sessions (LSD and placebo) in a balanced order. Study 1 headphones, but no other entertainment was provided. LSD
used a dose of 100 μg LSD and placebo in 24 subjects. Study 2 or placebo was administered at 0900 hours. The subjects
used 200 μg LSD or placebo in 16 subjects. The washout were never alone during the first 12 h after drug adminis-
periods between sessions were at least 7 days. The studies tration, and the investigator was in a room next to the subject
were conducted in accordance with the Declaration of for up to 24 h while subjects were asleep (mostly from 0100
Helsinki and approved by the local ethics committee. The to 0800 hours). Because subjective responses to LSD are
administration of LSD to healthy subjects was authorized by pronounced and peak at 2–3 h and last up to 12 h (Passie
the Swiss Federal Office for Public Health, Bern, Switzerland. et al, 2008; Schmid et al, 2015), effects on emotion processing
All of the subjects provided written consent before and prosociality were assessed 5 and 7 h after the 100 and
participating in either of the studies, and they were paid 200 μg doses, respectively, when the subjective effects of LSD
for their participation. The studies were registered at amounted to approximately 50% of the peak responses
ClinicalTrials.gov (NCT02308969, NCT01878942). The sub- (Dolder et al, 2015b; Schmid et al, 2015).
jective, endocrine, and pharmacokinetic effects of LSD in
Study 2 were previously reported (Dolder et al, 2015b;
Schmid et al, 2015; Strajhar et al, 2016). Study Drug
LSD (D-LSD hydrate; Lipomed AG, Arlesheim, Switzerland)
was administered in single oral doses of 100 or 200 μg. Both
Participants
doses are within the range of doses that are taken for
Forty healthy participants were recruited from the University recreational purposes (Passie et al, 2008).
of Basel campus via online advertisement. Twenty-four
subjects (12 men, 12 women; 33 ± 11 years old (mean ± SD);
Measures
range, 25–60 years) participated in Study 1, and 16 subjects
(8 men, 8 women; 29 ± 6 years old; range, 25–51 years) Facial Emotion Recognition Task. We used the FERT,
participated in Study 2. The inclusion and exclusion criteria which is sensitive to the effects of other psychoactive
50
Neuropsychopharmacology
LSD and emotion processing
PC Dolder et al
2640
substances, including serotonin and norepinephrine uptake altruistic behavior. The SVO was performed 6 and 8 h after
inhibitors (Harmer et al, 2004), MDMA (Bedi et al, 2010; the 100 and 200 μg doses of LSD, respectively.
Hysek et al, 2014b; Kirkpatrick et al, 2014; Schmid et al,
2014), and methylphenidate (Hysek et al, 2014b; Schmid Subjective mood. The VASs and the AMRS (Janke and
et al, 2014). The task included 10 neutral faces and 160 faces Debus, 1978) were repeatedly used to assess subjective effects
that expressed one of four basic emotions (ie, happiness, including aspects of empathy and sociality (Hysek et al,
sadness, anger, and fear), with pictures morphed between 0% 2014a; Schmid et al, 2015) (Supplementary Material and
(neutral) and 100% in 10% steps. Two female and two male Methods).
pictures were used for each of the four emotions. The stimuli
were presented in random order for 500 ms and then were Vital signs and adverse effects. Blood pressure, heart rate,
replaced by the rating screen where participants had to body temperature, pupil diameter, and adverse effects were
indicate the correct emotion. The outcome measure was measured as described in the Supplementary Material and
accuracy (proportion correct). The FERT was performed 5 Methods.
and 7 h after the 100 and 200 μg doses of LSD, respectively.
Drug concentrations. Blood samples for the analysis of
plasma LSD levels were collected in lithium heparin tubes
Multifaceted Empathy Test. The MET is a reliable and after completing the social cognitive tests 6 and 8 h after
valid task that assesses the cognitive and emotional aspects of administration of the 100 and 200 μg doses of LSD or
empathy (Dziobek et al, 2008). The MET has been shown to placebo, respectively. Plasma LSD concentrations were
be sensitive to oxytocin (Hurlemann et al, 2010), MDMA determined using liquid-chromatography tandem mass
(Hysek et al, 2014a; Kuypers et al, 2014; Schmid et al, 2014), spectrometry (Dolder et al, 2015a).
and psilocybin (Preller et al, 2015). The computer-assisted
test consisted of 40 photographs that showed people in
emotionally charged situations. To assess cognitive empathy, Statistical Analyses
the participants were required to infer the mental state of the All of the data were analyzed using repeated measures
subject in each scene and indicate the correct mental state analysis of variance (ANOVA), with drug (LSD vs placebo)
from a list of four responses. Cognitive empathy was defined as the within-subjects factor and dose (100 vs 200 μg) as the
as the percentage of correct responses relative to total between-subjects factor, followed by the Tukey’s post hoc test
responses. To measure emotional empathy, the subjects were based on significant main effects or interactions. Repeated
asked to rate how much they were feeling for an individual in subjective measures were expressed as peak effects prior to
each scene (ie, explicit emotional empathy) and how much the ANOVAs. Additionally, differences at individual time
they were aroused by each scene (ie, implicit emotional points were also compared using paired t-tests. Modulatory
empathy) on a 1–9 point scale. The latter rating provides an effects by sex or previous hallucinogen use were excluded by
inherent additional assessment of emotional empathy, which adding sex or substance use as an additional factor to the
is considered to reduce the likelihood of socially desirable ANOVAs. Sex or previous substance use did not moderate
answers. The three aspects of empathy were each tested with outcome measures.
20 stimuli with positive valence and 20 stimuli with negative
valence, resulting in a total of 120 trials. The MET was
performed 5 h and 30 min after the 100 μg LSD dose and 7 h RESULTS
and 30 min after the 200 μg LSD, respectively.
Facial Emotion Recognition
The effects of LSD on the FERT are shown in Figure 1. Data
SVO test. We used the paper version of the validated SVO were missing from 2 of the 24 subjects in the 100 μg LSD
test to assess social behavior (Murphy et al, 2011). The SVO dose group because of technical problems. LSD impaired the
measure was previously shown to be sensitive to MDMA recognition of fearful faces (main effect of drug: F1,36 = 20.71,
(Hysek et al, 2014a). In this economic resource allocation po0.001), with no drug × dose interaction. Impairments
task, prosociality is defined as behavior that maximizes the were found in both the 100 and 200 μg dose groups
sum of resources for the self and others and minimizes the compared with placebo (po0.01 and po0.05, respectively).
difference between the two. The test consists of six primary A significant main effect of drug (F1,36 = 7.36, p = 0.01)
and nine secondary SVO slider items with a resource indicated that LSD also impaired the recognition of sad faces,
allocation choice over a defined continuum of joint payoffs but post hoc comparisons of the two dose groups with
(Murphy et al, 2011). The participants were instructed to placebo did not reach significance. No significant effects of
choose a resource allocation that defined their most LSD on the decoding of neutral, happy, or angry facial
preferred joint distribution between themselves and another expressions were found.
person. The allocated funds had real value, and four
randomly selected subjects received the funds they earned. Empathy
Mean allocations for the self and the other were calculated
(Hysek et al, 2014a; Murphy et al, 2011), and the inverse The effects of LSD on explicit emotional and cognitive
tangent of the ratio of these two means produced an angle empathy are shown in Figure 2. Data were missing from 2 of
that indicated the participants’ SVO index. A smaller SVO the 24 subjects in the 100 μg LSD dose group because of
angle indicates more individualistic or competitive behavior, technical problems. There were significant main effects of
and a larger SVO angle indicates more prosocial or even drug on explicit and implicit emotional empathy ratings
51
Neuropsychopharmacology
LSD and emotion processing
PC Dolder et al
2641

Figure 1 Lysergic acid diethylamide (LSD) impaired fear recognition on Figure 2 Lysergic acid diethylamide (LSD) increased emotional empathy
the Face Emotion Recognition Task . LSD also impaired the decoding of sad and decreased cognitive empathy on the Multifaceted Empathy Test. The
faces (significant main effect of drug), but the effects did not reach statistical data are expressed as mean ± SEM in 22 and 16 subjects in the 100 and
significance in the individual dose groups. The data are expressed as 200 μg LSD dose groups, respectively. *po0.05, **po0.01, significant
mean ± SEM in 22 and 16 subjects in the 100 and 200 μg LSD dose groups, difference from placebo.
respectively. *po0.05, **po0.01, significant difference from placebo.

(F1,36 = 14.05, po0.001 and F1,36 = 6.71, p = 0.01, respec- peak effects at the higher compared with the lower dose.
tively), indicating that LSD increased both aspects of Ratings of ‘happy’ were similarly increased by both doses.
emotional empathy. The post hoc tests showed that the LSD produced small dose-dependent increases in ‘bad drug
200 μg dose but not the 100 μg dose of LSD produced a effect’ and ‘fear’ (Figure 3, Table 1). On the AMRS, LSD
significant effect on explicit (po0.01) and implicit (p = 0.01) significantly increased ratings of ‘well-being’, ‘emotional
empathy scores compared with placebo. The valence-specific excitation’, ‘inactivity’, ‘introversion’, and ‘dreaminess’
analysis showed that LSD significantly increased explicit and compared with placebo (Figure 4 and Table 1). There was
implicit emotional empathy scores for positive emotional a significant main effect of LSD on ‘fear’ but no significant
stimuli ( F1,36 = 24.32, po0.001 and F1,36 = 10.47, po0.01, effects in the individual studies.
respectively) but there were only trend effects for negative
emotional stimuli (F1,36 = 3.29, p = 0.08 and F1,36 = 2.82, Vital Signs and Adverse Effects
p = 0.1, respectively). LSD decreased cognitive empathy,
reflected by a significant main effect of drug (F1,36 = 16.87, Peak values and statistics are shown in Table 1. Compared
po0.001). The post hoc tests showed that this effect was with placebo, LSD increased blood pressure, heart rate, and
significant for both the 100 and 200 μg doses compared with body temperature as well as pupil size in the dark and after a
the respective placebo conditions (both po0.05). light stimulus (Table 1). These effects were similar for both
doses (no drug × dose interaction). Compared with placebo,
Social Value Orientation both doses of LSD increased the total acute (0–10 h) adverse
effects. Only the high dose increased the total subacute
A significant effect of drug was found on the SVO angle (10–24 h) adverse effects. Adverse effects 24–72 h were
(F1,38 = 4.31, po0.05), indicating that LSD increased proso- slightly increased in the total sample but not in the individual
ciality. The post hoc tests showed that this effect did not studies (Table 1). The frequently reported adverse effects are
reach significance in the individual LSD dose groups and was presented in Supplementary Table S2. There were no severe
only evident in the larger total study sample. adverse events.

Subjective Mood Effects Plasma Drug Levels and Correlations Between Effects
Subjective effects on the VASs are shown in Figure 3, and Plasma concentrations of LSD were 0.7 ± 0.3 ng/ml (mean ± SD)
maximal values are presented in Table 1. LSD increased 6 h after administration of the 100 μg dose and 1.3 ± 0.6 ng/ml
maximal VAS rating scores, including those reflecting 8 h after administration of the 200 μg dose. These time points of
empathy and prosociality such as ‘feeling close to others’, blood sample collection were immediately after the social
‘open’, ‘trust’, and ‘I want to be with others’, with greater cognitive tests performed in the respective dose groups. Plasma
52
Neuropsychopharmacology
LSD and emotion processing
PC Dolder et al
2642

Figure 3 Subjective effects of lysergic acid diethylamide (LSD) over time on the Visual Analog Scales (VASs). LSD or placebo was administered at t = 0. The
data are expressed as mean ± SEM in 24 and 16 subjects in the 100 and 200 μg LSD dose groups, respectively. LSD significantly increased ratings on all VASs
with significant dose–response effects, except for ratings of ‘happy’. The corresponding maximal effects and statistics are shown in Table 1. Emotion
recognition (Face Emotion Recognition Task), empathy (Multifaceted Empathy Test), and social value orientation (SVO) tests were conducted 5–6 and 7–8 h
after the administration of the 100 and 200 μg LSD dose, respectively. +/*po0.05, ++/**po0.01, +++/***po0.001 for the 100/200 μg LSD dose,
respectively, compared with placebo (T-tests).

LSD levels correlated with explicit emotional empathy scores on Schmid et al, 2014), and increased prosociality on the SVO
the MET for positive (Spearman Rs = 0.37, po0.05, n = 38) but test (Hysek et al, 2014a). LSD did not facilitate perception of
not for negative emotional situations. Plasma levels of LSD happiness in the FERT similar to MDMA (Bedi et al, 2010;
were not associated with FERT or SVO test measures. Hysek et al, 2014b), possibly because detection of positive
Plasma levels of LSD were associated with LSD-induced ratings basic emotions is very accurate in healthy subjects and
of trust (Spearman Rs = 0.32, po0.05, n = 40). LSD-induced difficult to enhance. Thus, the 5-HT2A receptor agonist LSD
VAS ratings for feelings of ‘closeness’ and ‘trust’ were associated and 5-HT releaser MDMA may produce overall similar
with greater explicit empathy for positive emotional stimuli effects on the processing of emotional information. However,
(Spearman Rs = 0.35, po0.05 and Rs = 0.47, po0.01, respec- in contrast to MDMA, LSD also impaired cognitive empathy
tively, n = 38). on the MET, and the higher dose also decreased the
recognition of neutral faces on the FERT, indicating
nonspecific performance effects. Similar to LSD, the
DISCUSSION 5-HT2A receptor agonist psilocybin decreased the recogni-
tion of negative facial expressions (Kometer et al, 2012) and
LSD positively altered the processing of emotional informa- increased emotional empathy on the MET (Preller et al,
tion by decreasing the recognition of fearful and sad faces 2015). Altogether, these findings indicate that LSD affects
and enhancing emotional empathy and prosociality. We are emotion processing similarly to MDMA and psilocybin.
aware of no other published data on the acute effects of LSD The marked acute psychedelic/hallucinogenic ‘peak
on emotion processing. However, MDMA produced very response’ to LSD and psilocybin has been considered
similar effects to those of LSD in the present study. MDMA relevant to their lasting effects (Carhart-Harris et al, 2016;
reduced the recognition of sad and fearful faces but not Griffiths et al, 2011). The present study showed that LSD has
happy faces on the FERT (Bedi et al, 2010; Hysek et al, dose-dependent subjective effects on empathogenic mood,
2014b), increased explicit and implicit emotional empathy on including ‘feelings of closeness to others’, ‘wanting to be
the MET (Hysek et al, 2014a; Kuypers et al, 2014) (mainly for with others’, ‘happiness,’ ‘openness,’ and ‘trust’ (Schmid
positive emotionally charged situations) (Hysek et al, 2014a; et al, 2015), in addition to more hallucinogen-specific
53
Neuropsychopharmacology
LSD and emotion processing
PC Dolder et al
2643
Table 1 Values and Statistics for the Subjective and Cardiovascular Peak Effects
Placebo 100 μg LSD 100 μg Placebo 200 μg LSD 200 μg Drug Drug × Dose
(mean ± SE) (mean ± SE) (mean ± SE) (mean ± SE)
F1,38 = p= F1,38 = p=

Subjective effects
Visual Analog Scales (VAS, %)
Any drug effect 0.9 ± 0.6 87.5 ± 3.3*** 0.1 ± 0.1 97.2 ± 1.7***# 1939 *** 6.21 *
Good drug effect 0.9 ± 0.6 85.2 ± 3.4*** 0.1 ± 0.1 96.8 ± 1.5***## 1661 *** 7.66 **
Bad drug effect 0.0 ± 0.0 17.3 ± 3.6** 0.1 ± 0.06 40.0 ± 8.2***### 51.17 *** 8.01 **
Fear 0.0 ± 0.0 8.4 ± 2.3 0.06 ± 0.1 31.3 ± 8.6***### 27.74 *** 9.24 **
Happy 1.2 ± 0.6 30 ± 3.4*** 5.0 ± 2.0 39.1 ± 4.2*** 141.5 *** 1 NS
Closeness to others 0.0 ± 0.0 15.2 ± 3.2*** 4.3 ± 1.8 32.3 ± 4.7***### 61.68 *** 5.38 *
Open 0.2 ± 0.2 17.0 ± 2.8*** 3.9 ± 1.5 41.0 ± 3.6***### 128.9 *** 18.4 ***
Trust 0.0 ± 0.0 22.0 ± 4.1*** 4.8 ± 2.1 39.8 ± 4.0***### 81.37 *** 4.2 *
I want to be hugged 0.0 ± 0.0 8.8 ± 2.7 3.4 ± 1.9 27.8 ± 6.8***### 23.52 *** 5.13 *
I want to hug someone 0.0 ± 0.0 10.4 ± 2.7* − 1.4 ± 3.3 27.6 ± 6.1***## 41.21 *** 9.13 **
I want to be alone 0.6 ± 0.6 7.7 ± 2.5 5.1 ± 1.9 17.6 ± 5.6 9.93 ** 0.76 NS
I want to be with other people 0.8 ± 0.8 12.8 ± 2.5** 10.8 ± 4.2 42.8 ± 5.5***### 79.87 *** 16.25 ***

Adjective Mood Rating Scale (AMRS, Δ score)


Well-being 0.0 ± 0.6 2.5 ± 1.0 1.8 ± 0.7 6.6 ± 1.6* 11.49 ** 1.11 NS
Emotional excitation − 0.3 ± 0.2 2.3 ± 0.5** − 0.3 ± 0.3 4.7 ± 1.0***## 53.5 *** 4.77 *
Inactivity 2.6 ± 0.7 9.0 ± 1.1** 1.3 ± 1.1 10.6 ± 2.7*** 30.82 *** 1.05 NS
Extroversion − 0.5 ± 0.3 − 0.1 ± 0.6 0.1 ± 0.5 1.5 ± 0.7 2.67 NS 0.77 NS
Introversion 0.4 ± 0.1 4.1 ± 0.6*** 0.5 ± 0.4 4.3 ± 0.8*** 51.92 *** 0.01 NS
Fear − 0.1 ± 0.1 0.9 ± 0.3 − 0.4 ± 0.3 1.3 ± 1.0 9.51 ** 0.72 NS
Dreaminess 0.2 ± 0.3 6.9 ± 0.7*** 0.8 ± 0.5 7.9 ± 0.6*** 160.2 *** 0.11 NS

Vital signs
Systolic blood pressure (mm Hg) 129 ± 2.0 142 ± 2.1*** 133 ± 3.8 148 ± 2.9*** 63.8 *** 0.13 NS
Diastolic blood pressure (mm Hg) 76.9 ± 1.5 85.7 ± 1.7*** 78.2 ± 2.0 87.6 ± 1.9*** 68.8 *** 0.08 NS
Heart rate (beats/min) 70.6 ± 1.8 79.1 ± 2.7** 72.8 ± 2.6 86.9 ± 4.29*** 33.7 *** 2.05 NS
Body temperature (Δ°C) 0.5 ± 0.1 0.8 ± 0.1** 0.3 ± 0.1 0.7 ± 0.1** 23.74 *** 0.22 NS
Pupil size (mm) 6.1 ± 0.2 6.9 ± 0.1*** 6.5 ± 0.2 7.2 ± 0.1*** 61.08 *** 0.81 NS
Pupil size after light (mm) 4.3 ± 0.2 5.2 ± 0.2*** 4.6 ± 0.2 5.6 ± 0.2*** 89.61 *** 0.02 NS

List of complaints (Δ LC total score)


Acute adverse effects (0–10 h) 0.5 ± 0.3 9.8 ± 1.8*** 0.1 ± 0.6 10.4 ± 3.0*** 38.37 *** 0 NS
Subacute adverse effects (10–24 h) − 0.2 ± 0.3 0.4 ± 0.2 − 0.4 ± 0.4 3.7 ± 1.4** 12.06 ** 6.76 *
Subacute adverse effects (24–72 h) − 0.5 ± 0.3 − 0.1 ± 0.2 − 0.8 ± 0.4 0.6 ± 0.9 6.03 * 1.83 NS

Values are mean ± SEM of the peak or peak changes (Δ) from baseline in 40 subjects. Sixteen subjects participated in the high dose study (200 μg) and 24 subjects in the
moderate dose study (100 μg).
*for po0.05, **for po0.01, ***for po0.001 compared with placebo. # for po0.05, ## for po0.01, ### for po0.001 compared with LSD 100 μg.

psychedelic peak effects. These acute subjective effects of also observed late in time at 6–8 h after LSD administration
LSD and its effects on the emotion processing and behavioral and after the peak response when a ‘plateau phase’ was
tests in the present study are very similar to those of the reached. At that time, the subjects were also less over-
prototypic empathogen MDMA. However, LSD induced whelmed by initially strong and mostly novel psychedelic
higher AMRS intro- than extroversion while MDMA experiences, which may open a window for psychother-
produced more extro- than introversion (Hysek et al, apeutic interventions. The emotional effects during the
2014a). Importantly, the subjective feelings of ‘happiness’, later phase of the acute LSD response (6–10 h) are likely
‘trust’, ‘closeness to others’, and ‘desire to be with others’ at beneficial to acutely facilitating the therapeutic alliance.
the high dose of LSD were maintained up to 6–12 h, and the Future research should address the relative contribu-
effects of LSD on emotion processing and prosociality were tions of the psychedelic peak experience vs empathogenic
54
Neuropsychopharmacology
LSD and emotion processing
PC Dolder et al
2644

Figure 4 Subjective effects on the Adjective Mood Rating Scale. Lysergic acid diethylamide (LSD) or placebo was administered at t = 0. The data are
expressed as mean ± SEM changes from baseline (−1 h) in 24 and 16 subjects in the 100 and 200 μg LSD dose groups, respectively. Emotion recognition
(Face Emotion Recognition Task), empathy (Multifaceted Empathy Test), and social value orientation (SVO) tests were conducted 5–6 and 7–8 h after the
administration of the 100 and 200 μg LSD dose, respectively. The corresponding maximal effects and statistics are shown in Table 1. *po0.05, **po0.01,
***po0.001 compared with placebo (T-tests).

emotional effects of LSD to its potential therapeutic effects. strong alterations in wake consciousness and impairments in
Additionally, it seems that only the higher 200 μg dose of concentration (Schmid et al, 2015). The participants needed
LSD produced robust empathogenic effects. Furthermore, to adjust to the altered state of consciousness; therefore,
the relevance of deficits in cognitive empathy for the testing occurred after a ‘plateau phase’ was reached. Never-
therapeutic process is unclear. theless, at the time of testing, the subjective effects and plasma
The present study also showed that LSD was well tolerated concentrations of LSD were still at approximately 50% of the
in a controlled setting in healthy subjects. Adverse effects of peak responses and clearly effective in producing typical LSD
LSD mainly included acute dizziness, headache, and fatigue/ effects, providing a good time interval for conducting the
exhaustion lasting up to 72 h. Both doses of LSD produced neurocognitive tasks (Carhart-Harris et al, 2016; Schmid et al,
comparable moderate sympathomimetic effects including 2015). Additionally, the tests were performed later after the
elevated blood pressure, heart rate, body temperature, and high dose than after the low dose of LSD. However, at the
mydriasis. times of testing, plasma LSD concentrations were twice as
The present study used two doses of LSD within a high after the 200 μg dose compared with the 100 μg dose, and
clinically relevant dose range. In fact, the higher dose was
generating a dose/concentration–response effect was possible.
identical to both the amount and pharmaceutical formula-
The study has limitations. First, the dose effects of LSD
tion that were used in a clinical study in patients with anxiety
were studied in different participants and not within-subject.
(Gasser et al, 2014) and continue to be used in patients in
Second, we assessed only emotion recognition and no other
Switzerland. Additionally, LSD was administered to subjects
across a relatively wide age range (25–60 years). Importantly, measures such as face muscle responses to emotions (Wardle
the subjects typically had no or very limited hallucinogen et al, 2014) and the stimuli were artificial (pictures) rather
experience, which is possibly similar to cases in which LSD is than real people. With regard to the use of LSD in
used therapeutically in patients. In contrast, other contem- psychotherapy, we only assessed ‘empathic concern for
porary studies used lower doses of LSD in subjects with others’ but not whether the participants ‘felt cared for or
extensive prior substance use (Carhart-Harris et al, understood by someone else’ (Wardle and de Wit, 2014). It is
2016,2015). However, in the present study, previous possible that LSD affected attention and motivation and
hallucinogen use (1–3 times including LSD in six subjects) thereby task performance. Thus, it will be important to
did not alter the responses to LSD. replicate and expand our findings using additional emotion
In the present study, the tests were performed approxi- recognition tests (Wardle and de Wit, 2014), tests of
mately 3 h after the peak effects (Dolder et al, 2015b; Schmid responses to emotions (Wardle and de Wit, 2014; Wardle
et al, 2015). At the time of the peak response of LSD, test et al, 2014), and other measures of social interaction (Frye
administration would not have been feasible because of the et al, 2014).
55
Neuropsychopharmacology
LSD and emotion processing
PC Dolder et al
2645
In conclusion, LSD impaired emotion recognition of experiences: immediate and persisting dose-related effects.
negative emotions and enhanced emotional empathy, Psychopharmacology 218: 649–665.
particularly for positive emotional situations, and had Griffiths RR, Richards WA, McCann U, Jesse R (2006). Psilocybin
subjective and behaviorally tested prosocial effects. These can occasion mystical-type experiences having substantial and
effects of LSD in healthy participants likely have translational sustained personal meaning and spiritual significance. Psycho-
pharmacology 187: 268–283 discussion 284-292.
relevance to LSD-assisted psychotherapy in patients and can
Grob CS, Danforth AL, Chopra GS, Hagerty M, McKay CR,
be expected to reduce the perception of negative emotions Halberstadt AL et al (2011). Pilot study of psilocybin treatment
and facilitate the therapeutic alliance. for anxiety in patients with advanced-stage cancer. Arch Gen
Psychiatry 68: 71–78.
Harmer CJ, Shelley NC, Cowen PJ, Goodwin GM (2004). Increased
FUNDING AND DISCLOSURE positive versus negative affective perception and memory in
healthy volunteers following selective serotonin and norepinephr-
This work was supported by the Swiss National Science ine reuptake inhibition. Am J Psychiatry 161: 1256–1263.
Foundation (grant no. 320030_1449493 to MEL) and the Hurlemann R, Patin A, Onur OA, Cohen MX, Baumgartner T,
University of Basel (to FM). The authors declare no conflict Metzler S et al (2010). Oxytocin enhances amygdala-dependent,
of interest. socially reinforced learning and emotional empathy in humans.
J Neurosci 30: 4999–5007.
Hysek CM, Domes G, Liechti ME (2012). MDMA enhances ‘mind
ACKNOWLEDGMENTS reading’ of positive emotions and impairs ‘mind reading’ of
negative emotions. Psychopharmacology 222: 293–302.
We acknowledge the assistance of M Arends in text Hysek CM, Schmid Y, Simmler LD, Domes G, Heinrichs M,
editing. The studies were registered at ClinicalTrials.gov Eisenegger C et al (2014a). MDMA enhances emotional empathy
(NCT02308969, NCT01878942). and prosocial behavior. Soc Cogn Affect Neurosci 9: 1645–1652.
Hysek CM, Simmler LD, Schillinger N, Meyer N, Schmid Y,
Donzelli M et al (2014b). Pharmacokinetic and pharmacody-
namic effects of methylphenidate and MDMA administered alone
REFERENCES and in combination. Int J Neuropsychopharmacol 17: 371–381.
Bedi G, Hyman D, de Wit H (2010). Is ecstasy an ‘empathogen’? Janke W, Debus G (1978). Die Eigenschaftswörterliste. Hogrefe:
Effects of ± 3,4-methylenedioxymethamphetamine on prosocial Göttingen.
feelings and identification of emotional states in others. Biol Johnson MW, Garcia-Romeu A, Cosimano MP, Griffiths RR (2014).
Psychiatry 68: 1134–1140. Pilot study of the 5-HT2AR agonist psilocybin in the treatment of
Bogenschutz MP, Forcehimes AA, Pommy JA, Wilcox CE, tobacco addiction. J Psychopharmacol 28: 983–992.
Barbosa PC, Strassman RJ (2015). Psilocybin-assisted treatment Kirkpatrick MG, Gunderson EW, Perez AY, Haney M, Foltin RW,
for alcohol dependence: a proof-of-concept study. J Psychophar- Hart CL (2012). A direct comparison of the behavioral and
macol 29: 289–299. physiological effects of methamphetamine and 3,4-methylene-
Carhart-Harris RL, Kaelen M, Bolstridge M, Williams TM, dioxymethamphetamine (MDMA) in humans. Psychopharmacol-
Williams LT, Underwood R et al (2016). The paradoxical ogy 219: 109–122.
psychological effects of lysergic acid diethylamide (LSD). Psychol Kirkpatrick MG, Lee R, Wardle MC, Jacob S, de Wit H (2014).
Med 46: 1379–1390. Effects of MDMA and intranasal oxytocin on social and
Carhart-Harris RL, Kaelen M, Whalley MG, Bolstridge M, emotional processing. Neuropsychopharmacology 39: 1654–1663.
Feilding A, Nutt DJ (2015). LSD enhances suggestibility in Kometer M, Schmidt A, Bachmann R, Studerus E, Seifritz E,
healthy volunteers. Psychopharmacology 232: 785–794. Vollenweider FX (2012). Psilocybin biases facial recognition,
Dolder PC, Liechti ME, Rentsch KM (2015a). Development and goal-directed behavior, and mood state toward positive relative to
validation of a rapid turboflow LC-MS/MS method for the negative emotions through different serotonergic subreceptors.
quantification of LSD and 2-oxo-3-hydroxy LSD in serum and Biol Psychiatry 72: 898–906.
urine samples of emergency toxicological cases. Anal Bioanal Kuypers KP, de la Torre R, Farre M, Yubero-Lahoz S, Dziobek I,
Chem 407: 1577–1584. Van den Bos W et al (2014). No evidence that MDMA-induced
Dolder PC, Schmid Y, Haschke M, Rentsch KM, Liechti ME (2015b). enhancement of emotional empathy is related to peripheral
Pharmacokinetics and concentration-effect relationship of oral LSD oxytocin levels or 5-HT1a receptor activation. PLoS One 9:
in humans. Int J Neuropsychopharmacol (doi:10.1093/ijnp/pyv072). e100719.
Dziobek I, Rogers K, Fleck S, Bahnemann M, Heekeren HR, Wolf OT MacLean KA, Johnson MW, Griffiths RR (2011). Mystical
et al (2008). Dissociation of cognitive and emotional empathy in experiences occasioned by the hallucinogen psilocybin lead to
adults with Asperger syndrome using the Multifaceted Empathy increases in the personality domain of openness. J Psychophar-
Test (MET). J Autism Dev Disord 38: 464–473. macol 25: 1453–1461.
Frye CG, Wardle MC, Norman GJ, de Wit H (2014). MDMA Mithoefer MC, Wagner MT, Mithoefer AT, Jerome I, Doblin R
decreases the effects of simulated social rejection. Pharmacol (2010). The safety and efficacy of ± 3,4-methylenedioxymetham-
Biochem Behav 117: 1–6. phetamine-assisted psychotherapy in subjects with chronic,
Gasser P, Holstein D, Michel Y, Doblin R, Yazar-Klosinski B, treatment-resistant posttraumatic stress disorder: the first rando-
Passie T et al (2014). Safety and efficacy of lysergic acid mized controlled pilot study. J Psychopharmacol 25: 439–452.
diethylamide-assisted psychotherapy for anxiety associated with Mithoefer MC, Wagner MT, Mithoefer AT, Jerome L, Martin SF,
life-threatening diseases. J Nerv Ment Dis 202: 513–520. Yazar-Klosinski B et al (2013). Durability of improvement in
Gasser P, Kirchner K, Passie T (2015). LSD-assisted psychotherapy post-traumatic stress disorder symptoms and absence of harmful
for anxiety associated with a life-threatening disease: a qualitative effects or drug dependency after 3,4-methylenedioxymethamphe-
study of acute and sustained subjective effects. J Psychopharmacol tamine-assisted psychotherapy: a prospective long-term follow-
29: 57–68. up study. J Psychopharmacol 27: 28–39.
Griffiths RR, Johnson MW, Richards WA, Richards BD, Murphy RO, Ackermann KA, Handgraaf MJJ (2011). Measuring
McCann U, Jesse R (2011). Psilocybin occasioned mystical-type social value orientation. Judgm Decis Mak 6: 771–781.
56
Neuropsychopharmacology
LSD and emotion processing
PC Dolder et al
2646
Nichols DE (2016). Psychedelics. Pharmacol Rev 68: 264–355. Vollenweider FX, Vollenweider-Scherpenhuyzen MF, Babler A,
Oehen P, Traber R, Widmer V, Schnyder U (2013). A randomized, Vogel H, Hell D (1998). Psilocybin induces schizophrenia-like
controlled pilot study of MDMA (±3,4-methylenedioxymetham- psychosis in humans via a serotonin-2 agonist action. Neuroreport
phetamine)-assisted psychotherapy for treatment of resistant, 9: 3897–3902.
chronic post-traumatic stress disorder (PTSD). J Psychopharma- Wardle MC, de Wit H (2014). MDMA alters emotional processing
col 27: 40–52. and facilitates positive social interaction. Psychopharmacology
Passie T, Halpern JH, Stichtenoth DO, Emrich HM, Hintzen A 231: 4219–4229.
(2008). The pharmacology of lysergic acid diethylamide: a review. Wardle MC, Kirkpatrick MG, de Wit H (2014). 'Ecstasy' as a social
CNS Neurosci Ther 14: 295–314. drug: MDMA preferentially affects responses to emotional stimuli
Preller KH, Pokorny T, Krähenmann R, Dziobek I, Stämpfli P, with social content. Soc Cogn Affect Neurosci 9: 1076–1081.
Vollenweider FX (2015). The effect of 5-HT2A/1a agonist
treatment on social cognition, empathy, and social decision-
making. Eur Psychiatry 30(Suppl. 1): 22. This work is licensed under a Creative Commons
Schmid Y, Enzler F, Gasser P, Grouzmann E, Preller KH, Attribution-NonCommercial-NoDerivs 4.0
Vollenweider FX et al (2015). Acute effects of lysergic acid International License. The images or other third party
diethylamide in healthy subjects. Biol Psychiatry 78(8): 544–553. material in this article are included in the article’s Creative
Schmid Y, Hysek CM, Simmler LD, Crockett MJ, Quednow BB, Commons license, unless indicated otherwise in the credit line;
Liechti ME (2014). Differential effects of MDMA and if the material is not included under the Creative Commons
methylphenidate on social cognition. J Psychopharmacol 28:
license, users will need to obtain permission from the license
847–856.
Strajhar P, Schmid Y, Liakoni E, Dolder PC, Rentsch KM, holder to reproduce the material. To view a copy of this license,
Kratschmar DV et al (2016). Acute effects of LSD on visit http://creativecommons.org/licenses/by-nc-nd/4.0/
circulating steroid levels in healthy subjects. J Neuroendocrinol
(doi:10.1111/jne.12374). © The Author(s) 2016

Supplementary Information accompanies the paper on the Neuropsychopharmacology website (http://www.nature.com/npp)

57

Neuropsychopharmacology
Supplementary Information

Material and methods

Subjective effects

The Visual Analog Scales (VASs) were repeatedly used to assess subjective effects over time.
The VASs included “any drug effect,” “good drug effect,” “drug high,” “bad drug effect,” “fear,” “happy,”
“closeness to others,” “open,” “trust”, “I want to be hugged”, “I want to hug someone”, “I want to be
alone”, and “I want to be with others” and have previously been used (Hysek et al, 2014a; Schmid et
al, 2015). The VASs were presented as 100-mm horizontal lines (0-+100%) marked from “not at all” on
the left and “extremely” on the right. The VASs for “happy,” “closeness to others,” “open,” “trust”, “I
want to be hugged”, “I want to hug someone”, “I want to be alone”, and “I want to be with others” were
bidirectional ( 50%), marked from “not at all” on the left (-50), to “normal” in the middle (0), to
“extremely” on the right (+50). The VASs were administered 1 h before and 0, 0.5, 1, 1.5, 2, 2.5, 3, 4,
5, 6, 7, 8, 9, 10, 11, 12, 16, and 24 h after drug administration.
The 60-item Likert-scale short version of the Adjective Mood Rating Scale (AMRS) (Janke and
Debus, 1978) was administered 1 h before and 3, 10, and 24 h after placebo or LSD. The AMRS
contains subscales for well-being, emotional excitation, activity, inactivity, extro- and introversion, fear,
and dreaminess. The AMRS has previously been shown to be sensitive to the effects of
psychostimulants, empathogens, and hallucinogens (Hasler et al, 2004; Hysek et al, 2014b; Schmid et
al, 2015).

Vital signs, pupillary function, and adverse effects

Blood pressure, heart rate and body temperature were assessed repeatedly 1 h before and 0,
0.5, 1, 1.5, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, and 24 h after drug administration. Blood pressure (systolic
and diastolic) and heart rate were measured using an automatic oscillometric device (OMRON
Healthcare Europe NA, Hoofddorp, Netherlands). The measurements were performed in duplicate at
an interval of 1 min and after a resting time of at least 10 min. The averages were calculated for
analysis. Core (tympanic) temperature was measured using an GENIUSTM 2 ear thermometer (Tyco
Healthcare Group LP, Watertown, NY, USA).
Pupillometry was performed 1 h before and 1, 2.5, 4, 7, and 11 h after drug administration using
an infrared pupillometer (PRL-200, NeurOptics, Irvine, CA, USA) under standardized dark-light
conditions as previously described (Hysek and Liechti, 2012). The dark-adapted maximal pupil
diameter, minimal pupil diameter after a light stimulus, and constriction amplitude (difference between
maximal and minimal pupil size) were recorded.
Adverse effects were assessed using the 66-item List of Complaints (Zerssen, 1976) before and
10, 24, and 72 h after drug administration for the 0-10, 10-24, and 24-72 h time intervals, respectively.
Complaints are assessed as present or not present and there is no grading of the complaint. However,
the scale yields a total adverse effects score, reliably measuring general discomfort.

References

Hasler F, Grimberg U, Benz MA, Huber T, Vollenweider FX (2004). Acute psychological and
physiological effects of psilocybin in healthy humans: a double-blind, placebo-controlled dose-effect
study. Psychopharmacology 172: 145-156.
Hysek CM, Liechti ME (2012). Effects of MDMA alone and after pretreatement with reboxetine,
duloxetine, clonidine, carvedilol, and doxazosin on pupillary light reflex. Psychopharmacology 224:
363-376.
Hysek CM, Schmid Y, Simmler LD, Domes G, Heinrichs M, Eisenegger C, et al (2014a). MDMA
enhances emotional empathy and prosocial behavior. Soc Cogn Affect Neurosci 9: 1645-1652.
Hysek CM, Simmler LD, Schillinger N, Meyer N, Schmid Y, Donzelli M, et al (2014b). Pharmacokinetic
and pharmacodynamic effects of methylphenidate and MDMA administered alone and in
combination. Int J Neuropsychopharmacol 17: 371-381.
Janke W, Debus G (1978). Die Eigenschaftswörterliste. Hogrefe: Göttingen.
Schmid Y, Enzler F, Gasser P, Grouzmann E, Preller KH, Vollenweider FX, et al (2015). Acute effects
of lysergic acid diethylamide in healthy subjects. Biol Psychiatry 78: 544-553.
Zerssen DV (1976). Die Beschwerden-Liste. Münchener Informationssystem Psychis, München.

58
Table S1. Life-time prevalence of substance use

Subject MDMA amphetamine cocaine LSD psilocybin methylphenidate mescaline THC

Study 1

1 2 0 0 0 0 0 0 0
2 1 0 0 0 0 0 0 0
3 0 0 0 0 0 0 0 5-10
4 3 0 1 0 0 0 0 10-20
5 0 0 0 0 0 0 0 0
6 0 0 0 0 0 0 0 0
7 5 1 1 1 0 0 0 0
8 1 0 0 0 0 2 0 0
9 0 0 0 0 0 0 0 0
10 0 0 0 0 0 0 0 5-10
11 0 0 0 0 0 0 0 0
12 5 0 0 0 0 0 0 5-10
13 0 0 0 0 0 0 0 0
14 0 0 0 0 0 0 0 5-10
15 0 0 0 0 0 0 0 10-20
16 0 0 0 0 1 0 0 5-10
17 0 0 0 1 0 0 0 0
18 0 0 0 1 1 0 0 0
19 2 0 0 0 0 0 0 0
20 0 0 0 0 0 0 0 2
21 0 0 0 0 0 0 0 0
22 0 0 0 0 0 0 0 0
23 3 1 0 0 0 1 0 0
24 0 0 0 0 0 0 0 0
Study 2

1 4 0 0 0 0 2 0 1
2 2 0 0 0 1 0 1 5-10
3 4 0 0 1 0 2 0 10-20
4 0 0 0 0 3 0 0 5-10
5 2 0 0 0 0 0 0 50-100
6 0 0 0 0 0 0 0 100-200
7 3 0 2 2 0 0 0 100-200
8 2 0 0 0 0 0 0 0
9 1 0 0 0 2 0 0 5-10
10 0 0 0 0 0 0 0 100-200
11 0 0 0 0 0 0 0 20-50
12 2 1 2 0 1 0 0 50-100
13 8 0 0 2 0 0 0 50-100
14 0 0 0 0 0 1 0 2
15 3 1 1 0 0 1 0 1
16 0 0 0 0 0 0 0 5-10

Values are times used in life

59
Table S2. Percent of acute and sub-acute adverse drug effects up to 72 hours.

Study 1 Placebo 100 µg LSD 100 µg

0h 0-10h 10-24h 24-72h 0h 0-10h 10-24h 24-72h

Difficulty concentrating 0% 0% 0% 0% 0% 54% 0% 0%


Headache 0% 29% 13% 0% 4% 58% 17% 21%
Exhaustion 4% 13% 4% 0% 4% 25% 4% 4%
Dizziness 0% 0% 0% 0% 0% 25% 0% 0%
Lack of appetite 0% 0% 0% 0% 0% 46% 0% 4%
Dry mouth 4% 4% 0% 0% 0% 29% 0% 0%
Imbalance 0% 0% 0% 0% 0% 42% 0% 0%
Nausea 0% 0% 0% 0% 0% 21% 0% 0%
Fatigue 29% 29% 13% 4% 17% 58% 29% 33%

Study 2 Placebo 200 µg LSD 200 µg

0h 0-10h 10-24h 24-72h 0h 0-10h 10-24h 24-72h

Difficulty concentrating 0% 4% 4% 0% 0% 42% 13% 8%


Headache 4% 13% 8% 0% 8% 38% 33% 8%
Exhaustion 4% 4% 4% 0% 4% 29% 42% 13%
Dizziness 4% 0% 0% 0% 0% 29% 8% 0%
Lack of appetite 0% 0% 0% 0% 4% 21% 13% 0%
Dry mouth 0% 0% 0% 0% 0% 21% 13% 4%
Imbalance 0% 0% 0% 0% 0% 21% 4% 0%
Nausea 4% 4% 4% 0% 8% 17% 0% 0%
Fatigue 25% 17% 17% 0% 38% 38% 46% 17%

Data percent in 16 and 24 subjects for the high and low dose group, respectively.

60
STUDY 1 Enrollment
Assessed for
LSD = 100 µg eligibility (n=26)

Excluded (n=2)
•Not meeting inclusion criteria(n=2)

Randomized (n=24)

Allocation
Placebo- LSD-
LSD (n=12) Placebo (n=12)

Drop outs (n=0)

Participants completed the study (n=24)

Analysis
Analysed (n=24)
• Excluded from analysis (n=0)
61
STUDY 2 Enrollment
Assessed for
LSD = 200 µg eligibility (n=20)

Excluded (n=4)
•Not meeting inclusion criteria(n=4)

Randomized (n=16)

Allocation
Placebo- LSD-
LSD (n=8) Placebo (n=8)

Drop outs (n=0)

Participants completed the study (n=16)

Analysis
Analysed (n=16)
• Excluded from analysis (n=0)
62
3.5 Publication 5

Acute effects of LSD on amygdala activity during processing of


fearful stimuli in healthy subjects

Felix Müller1, Claudia Lenz1, Patrick C. Dolder2, Samuel Harder2, Yasmin Schmid2,
Undine E. Lang1, Matthias E. Liechti2, Stefan J. Borgwardt1

1 Department of Psychiatry (UPK), University of Basel, Switzerland


2 Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of
Clinical Research, University Hospital Basel and University of Basel, Switzerland

63
OPEN
Citation: Transl Psychiatry (2017) 7, e1084; doi:10.1038/tp.2017.54
www.nature.com/tp

ORIGINAL ARTICLE
Acute effects of LSD on amygdala activity during processing
of fearful stimuli in healthy subjects
F Mueller1,3, C Lenz1,3, PC Dolder2, S Harder2, Y Schmid2, UE Lang1, ME Liechti2 and S Borgwardt1

Lysergic acid diethylamide (LSD) induces profound changes in various mental domains, including perception, self-awareness and
emotions. We used functional magnetic resonance imaging (fMRI) to investigate the acute effects of LSD on the neural substrate of
emotional processing in humans. Using a double-blind, randomised, cross-over study design, placebo or 100 μg LSD were orally
administered to 20 healthy subjects before the fMRI scan, taking into account the subjective and pharmacological peak effects of
LSD. The plasma levels of LSD were determined immediately before and after the scan. The study (including the a priori-defined
study end point) was registered at ClinicalTrials.gov before study start (NCT02308969). The administration of LSD reduced reactivity
of the left amygdala and the right medial prefrontal cortex relative to placebo during the presentation of fearful faces (Po 0.05,
family-wise error). Notably, there was a significant negative correlation between LSD-induced amygdala response to fearful stimuli
and the LSD-induced subjective drug effects (Po 0.05). These data suggest that acute administration of LSD modulates the
engagement of brain regions that mediate emotional processing.

Translational Psychiatry (2017) 7, e1084; doi:10.1038/tp.2017.54; published online 4 April 2017

INTRODUCTION between placebo and LSD during processing of emotional stimuli,


Lysergic acid diethylamide (LSD), a potent psychoactive trials for fearful faces were contrasted against trials for neutral
substance,1 induces profound changes in various mental domains, faces. We thereby focused on the amygdala as one central part of
including perception, self-awareness and emotions.2,3 As with the neural emotion processing, in particular, of anxiety13,14 and
other psychedelics (for example, psilocybin and mescaline), these additionally included two other regions (the fusiform gyrus and
effects are mainly mediated through agonism at the serotonin the medial frontal gyrus) known to be responsive to fearful
5-HT2A receptor.1,4 Currently, there are renewed efforts to use faces.14 Differences between placebo and LSD conditions were
substances like LSD and psilocybin in basic research and clinical evaluated by second-level paired t-test analysis. In addition, the
practice.2,3,5,6,7 Psilocybin has been studied as a treatment option amygdala response to fearful faces after LSD was correlated with
for addiction, depression and for anxiety in patients with advanced the subjective drug effect, as assessed by a visual analogue scale
stage cancer.8–11 LSD has been shown to reduce anxiety in patients directly before the scan. The primary and a priori-defined study
with life-threatening diseases.12 With the investigation of its basic hypothesis was that LSD would decrease the amygdala response
pharmacological and psychological effects, there is also rising to fearful stimuli and that this decrease would be associated with
interest in the neuronal correlates of the LSD-induced altered state the subjective psychedelic effects.
of consciousness. Although several modern studies on psilocybin
have been conducted, recent data on LSD in humans are still very
limited.1 MATERIALS AND METHODS
Functional neuroimaging provides a sensitive means of We used a randomised, placebo-controlled, double-blind, cross-over design.
examining how LSD acts on the brain. No data investigating LSD Each participant completed two study sessions, with a washout period of at
least 7 days between the sessions. The study was approved by the Ethics
effects on emotion processing have yet been published. The aim
Committee for Northwest/Central Switzerland (EKNZ) and by the Federal
of the present study was therefore to investigate these acute Office of Public Health. Written informed consent was obtained from all the
effects of LSD using functional magnetic resonance imaging participants. The study (including the a priori-defined study end point) was
(fMRI). Using a double-blind, randomised, cross-over study design, registered at clinicaltrials.gov before study start (NCT02308969).
placebo or 100 μg LSD were orally administered to 20 physically
and mentally healthy participants 2.5 h before the fMRI scan,
Subjects
taking into account the subjective and pharmacological peak
The subjects were recruited by advertisement and word of mouth. The
effects of LSD.2,6 Subjects only had minimal lifetime exposure to
sample size was determined by power analysis based on previous data.15,16
illicit drugs; notably, only two subjects had had prior experience The exclusion criteria were age o25 or 465 years, pregnancy (as
with a psychedelic, both on one occasion only. During the fMRI determined by urine test), nursing, hypertension (4140/90 mm Hg) or
scan, human fearful and neutral faces of a well-validated paradigm hypotension (systolic blood pressureo85 mm Hg), cardiac or neurological
were presented. To test our hypothesis that there were differences disorders, use of any regular medication, as determined by medical history

1
Department of Psychiatry, Universitäre Psychiatrische Kliniken, University of Basel, Basel, Switzerland and 2Division of Clinical Pharmacology and Toxicology, Department of
Biomedicine and Clinical Research, University Hospital Basel, Basel, Switzerland. Correspondence: Professor S Borgwardt, Department of Psychiatry, Universitäre Psychiatrische
Kliniken, University of Basel, Wilhelm Klein-Strasse 27, 4012 Basel, Switzerland.
E-mail: [email protected]
3
These two authors contributed equally to this work.
Received 22 November 2016; revised 30 January 2017; accepted 14 February 2017

64
Acute effects of LSD on amygdala activity in healthy subjects
F Mueller et al
2
and general medical examination including electrocardiography, blood participants were presented with 10 different facial identities (pictures of
chemistry and haematology, use of illicit drugs (except cannabis) 410 human faces from the Ekman & Friesen series of Pictures of Facial Affect),
times or any time within the previous 2 months (as assessed by the history each expressing 50 or 100% intensities of fear or a neutral expression.
and urine test for benzodiazepines, cocaine, amphetamines, methadone, There were thus 30 different facial stimuli in total. Each face was shown
opiates and barbiturates), smoking of 410 cigarettes per day, history of twice for 2 s, resulting in a total of 60 stimuli during the paradigm. The
drug dependence, personal or first-degree relative with a history of order of facial identities and expression type was pseudo-randomised to
seizures, personal or first-degree relative with an axis I major psychiatric prevent successive presentation of the same identity or facial expression
disorder (as determined by general medical history and a semi-structured type. The length of the interstimulus interval, during which subjects
interview for Diagnostic and Statistical Manual of Mental Disorders, fourth viewed a fixation cross, was varied from 3 to 8 s according to a Poisson
edition). The subjects provided written informed consent and received distribution, with an average interval of 5.9 s. To ensure maximal attention
monetary compensation for their participation. to the presented faces, subjects were requested to decide on the gender of
face stimuli by pressing a left or a right button. Accuracy and reaction
times were monitored and recorded.
Study procedure
The study included a screening visit, two 25 h test sessions and an end of
study visit. The experimental sessions took place in a quiet room in the Data analysis
University Hospital of Basel, Switzerland. The study dates were between The data analysis was performed using SPM12 (http://www.fil.ion.ucl.ac.uk/
December 2014 (first subject screened) and September 2015 (last end of spm/). All the volumes were slice time corrected, realigned to the first volume,
study session). The participants were monitored for adverse reactions and co-registered to the pre-processed T1-weighted structural volume, normalized
events during the study dates and at the end of study visit. All the adverse into a standard stereotactic space (Montreal Neurological Institute, MNI) and
events were recorded. The participants were instructed to abstain from any smoothed with a 6 mm full width at half maximum Gaussian kernel. The
illicit drugs during the whole study period and, additionally, to abstain from dummy scans were excluded from any further processing and the remaining
caffeine, chocolate and alcohol for at least 8 h before the sessions. The urine volumes were quality checked for severe head motion and image artefacts.
drug tests (for tetrahydrocannabinol, benzodiazepines, cocaine, ampheta- the subjects with head motion of 42 mm translation or 42° rotation were
mines, methadone, opiates and barbiturates) were taken randomly on one excluded. During model specification, the onset times for each trial of neutral,
of the two sessions. In women, pregnancy tests were performed before 50% and 100% fearful faces were convolved with a canonical haemodynamic
every session. Except for tetrahydrocannabinol, which can be detected for response function. The serial correlations were removed with a first-order
several weeks, detection of any drug of abuse resulted in study exclusion. A autoregressive model and a high-pass filter (128 s) was applied to remove low
light standardised breakfast was served at both the sessions. Placebo and frequency noise. The six motion parameters for translation and rotation were
LSD were administered orally, 2.5 h before the MRI scan at 0900 h, taking entered as nuisance covariates. In addition, time and dispersion derivatives
into account the subjective and pharmacological peak effects of LSD.2,6 were included in the individual design matrix during the first-level analysis.
Each trial for 50 and 100% fearful faces was then contrasted against neutral
faces, and then produced a subject-specific contrast image propagated to the
Drugs and randomization second-level analysis. One-sample t-tests were used to assess the activity
Gelatin capsules containing 100 μg D-lysergic acid diethylamide hydrate induced by the main effect task over all included subjects. The threshold over
(Lipomed, Arlesheim, Switzerland) and identical capsules containing the whole brain was set at P = 0.05, corrected for multiple comparisons (family-
mannitol were prepared. Each subject received either placebo or LSD on wise error, FWE). Differences between the LSD and placebo treatment were
two study sessions in a counterbalanced manner. Only the person evaluated by a second-level paired t-test. Whole-brain threshold was set at
dispensing the substance (who was not further involved in conducting the P = 0.001, uncorrected for multiple comparisons, with an extent threshold of
study) was aware of the treatment assignment. Subjects and study k = 10 voxels. We restricted our analysis to three meta-analytically identified14
personnel were blind to the treatment order. regions of interest, namely the amygdala, the fusiform gyrus and the medial
frontal gyrus. Those regions were specifically described to be involved in the
processing of fearful faces compared with neutral faces.14 Based on the
Image acquisition Harvard-Oxford Atlas for cortical and subcortical structures, a mask comprising
Scanning was conducted on a 3 Tesla MRI system (Magnetom Prisma, those regions was created. Small volume correction was used for clusters
Siemens Healthcare, Erlangen, Germany), using a 20-channel phased array observed within this hypothesized region of interest. The statistical threshold
radio frequency head coil. Functional MRI acquisition was based on an was adjusted to provide a FWE of Po0.05, corrected for small volumes. The
interleaved T2*-weighted echo planar imaging sequence, with 39 axial small volume correction was performed in the global maximum, with a sphere
slices with a slice thickness of 3 mm, a 0.5 mm inter-slice gap, a field-of- of 5 mm, in accordance with previous fMRI studies on amygdala activity.17,18
view of 228 × 228 cm2 and an in-plane image matrix size of 76 × 76— The correlation with the subjective effect of LSD in the visual analogue
resulting in 3 × 3 × 3 mm3 resolution. The corresponding repetition time scale was performed using the extracted beta values of the amygdala cluster
was 2.5 s, echo time 30 ms and bandwidth = 2350 Hz per pixel. In total, 152 under the LSD condition. We thereby used the ‘100% fearful versus neutral
volumes were acquired (including three dummy scan volumes to ensure contrast’ to obtain the distinct effect of the fearful stimuli. The calculations
signal stabilization). were performed using SPSS version 23.00 (IBM, Zurich, Switzerland).

Subjective effect measurements RESULTS


The visual analogue scale ‘Any subjective drug effects’ was used to assess
We included data sets from 20 healthy subjects—9 men, 11 women;
the overall subjective response to LSD before the scan. The visual analogue
scale was presented as a 100 mm horizontal line (0–100%) marked ‘not at mean age 32 ± 10.2 years; range: 25–58 years, all right-handed and
all’ on the left and ‘extreme’ on the right. The scale was rated by the all but one with an academic background, originally with 24 study
volunteers 2 h after the administration of LSD or placebo. participants. The data sets from four subjects were excluded
because of artefacts due to head movements. The lifetime drug
use of the 20 included subjects is shown in Table 1. None of the
Plasma levels
participants tested positive for any drug (including tetrahydrocan-
The blood was collected into lithium heparin tubes 2 and 3 h after the nabinol) in the screening or test session. No serious adverse
administration of LSD and placebo, respectively. The blood samples were
immediately centrifuged and rapidly stored at − 20 °C until analysis. LSD
reactions or events occurred during the whole period of the study in
concentrations in plasma were determined using a validated liquid any of the participants. The plasma levels of LSD were determined
chromatography-tandem mass spectrometry method.6 immediately before and after the scan and were 1.3 ± 0.6 ng ml − 1
(mean ± s.d.) and 1.1 ± 0.5 ng ml − 1 (mean ± s.d.), respectively.
fMRI paradigm
During the fMRI acquisition, the study subjects participated in a 6 min Task performance
experiment based on event-related design implemented with E-Prime 2.0 The differences between the LSD and placebo conditions in task
(Psychology Software Tools, Pittsburgh, PA, USA). During the task, performance were assessed using paired t-tests. The mean subject
65
Translational Psychiatry (2017), 1 – 5
Acute effects of LSD on amygdala activity in healthy subjects
F Mueller et al
3
Table 1. Cumulative lifetime use of legal and illicit drugs of the included subjects

Nicotine Stimulants
No. of subjects with regular use 6/20 No. of subjects who have ever used 4/20
Cigarettes per day (mean/s.d./range) 1.40/4.03/0–10 Lifetime occasions (mean/s.d./range) 0.35/0.5/0–2

Caffeine Sedatives
No. of subjects with regular use 20/20 No. of subjects who have ever used 0/20
Units per day (mean/s.d./range) 3.05/1.96/1–8 Lifetime occasions (mean/s.d./range) 0/0/0

Alcohol Psychedelics
No. of subjects with regular use 20/20 No. of subjects who have ever used 2/20
Units per week (mean/s.d./range) 4.50/2.89/1–10 Lifetime occasions (mean/s.d./range) 0.10/0/0–1

Cannabis Opioids
No. of subjects who have ever used 15/20 No. of subjects who have ever used 1/20
Lifetime occasions (mean/s.d./range) 7.85/13.39/1–50 Lifetime occasions (mean/s.d./range) 0.05/0/0–1

MDMA Others
No. of subjects who have ever used 6/20 No. subjects who have ever used 0/20
Lifetime occasions (mean/s.d./range) 0.60/0.89/0–3 Lifetime occasions (mean/s.d./range) 0/0/0
Abbreviation: MDMA, 3,4-methylenedioxymethamphetamine.

response times did not differ significantly between the two


conditions (LSD: 964 ± 128 ms (mean ± s.d.); placebo: 910 ± 289 ms
(mean ± s.d.); t21 = 2.0, P = 0.06). Furthermore, no significant differ-
ences were found between the conditions in correctness of response
(LSD: 93.1 ± 10.8% (mean ± s.d.); placebo: 97.3 ± 3.3% (mean ± s.d.);
t21 = − 1.8, P = 0.08) or absence of button presses (LSD: 4.5 ± 9.3%
(mean ± s.d.); placebo: 1.3 ± 1.8% (mean ± s.d.); t21 = 1.5, P = 0.16).

Effect of task
With both treatments (n = 40), viewing neutral faces versus
baseline was associated with bilateral activation in a network
comprising the cerebellum, fusiform gyrus, occipital gyrus and the
middle cingulate gyrus and lateral activation in the left frontal and
lingual gyrus (FWE-corrected at P o 0.05).
Viewing 100 and 50% fearful faces versus baseline was
associated with bilateral activation in the cerebellum, fusiform
gyrus, occipital gyrus, middle superior parietal lobule and lateral
activation in the left cingulate and frontal gyrus (FWE-corrected at
P o0.05). Under the placebo condition, presentation of fearful
faces induced a significant (small volume correction, P o0.05 FWE
cluster level) activation of the left amygdala (MNImax x = − 20,
y = − 12, z = − 12; cluster size 22; Z-score 3.59) compared with
presentation of neutral faces.

Effect of LSD on neural response to fearful versus neutral faces


Compared with placebo, administration of LSD reduced neural
response to fearful versus neutral faces in the left and right
amygdala and the medial frontal gyrus (P o 0.001, k = 10; see
Figure 1). No increased activity was observed. After correction for Figure 1. Neural response to fearful versus neutral faces after
multiple comparisons (small volume correction, P o 0.05 FWE), placebo compared with LSD treatment. LSD decreased reactivity
significantly reduced activity was observed in the left amygdala (shown in red-yellow) to fearful faces in the amygdala (a) and the
(MNImax x = − 15, y = 9, z = − 14; cluster size 24; Z-score 3.12) and right medial frontal gyrus (b). Regions of interest (amygdala,
fusiform gyrus, medial frontal gyrus) are shown in blue. Threshold
the right medial frontal gyrus (MNImax x = 15, y = 42, z = 16; cluster Po0.001, k = 10. Right is right side of the brain. LSD, lysergic acid
size 12; Z-score 3.78). In addition, there was a significant negative diethylamide.
correlation between amygdala blood oxygen-level dependent
response to fearful stimuli under the LSD condition and the LSD-
induced subjective drug effects (r = − 0.46, P o 0.05; see Figure 2). reactivity to fearful stimuli in healthy subjects. In addition,
amygdala deactivation by LSD was associated with its acute
subjective psychedelic effects. We administered 100 μg LSD, a
DISCUSSION representative dose that produces typical and robust psychedelic
In summary, the present study used fMRI for we believe the first effects.19 In addition, subjects had only had a minimal exposure
time to investigate the effects of LSD on the neural substrate of to recreational drugs and were mostly psychedelic-naive, as is
emotional processing. We found that LSD decreased amygdala probably the case in patients receiving LSD-assisted
66
Translational Psychiatry (2017), 1 – 5
Acute effects of LSD on amygdala activity in healthy subjects
F Mueller et al
4
involved in emotional functions.40 Within the mPFC–amygdala
circuit, the more ventral parts of the mPFC have been implicated in
inhibitory functions,40 whereas the more dorsal parts are thought to
be part of an ‘aversive-amplification circuit’.41 This mechanism has
been linked to negative affective bias in anxiety disorder.42,43
Consistent with our findings, serotonin depletion has been shown
to increase mPFC activity and functional connectivity between the
mPFC and the amygdala in response to fearful stimuli.44
The use of psychedelics as an additive in psychotherapy has
recently been rediscovered10,12,45 and our result is relevant for this
field of research. Processing biases towards negative stimuli are a
feature of several mental diseases, such as depression and social
anxiety disorder, and are associated with increased reactivity of
the amygdala.46,47 Resolving this processing bias might thus
reflect one important and potentially therapeutically useful effect
of psychedelic substances by, for example, facilitating the
therapeutic alliance48,49 and reducing perception of negative
emotions and social cognitive deficits. As we have recently
Figure 2. Relation between left amygdala BOLD activity during reported, LSD also exhibits some ‘empathogenic’ effects (such as
presentation of fearful faces under the LSD condition and visual increased openness and trust),2,20 which are usually ascribed to
analogue scale (VAS) for ‘any subjective drug effects’ of LSD substances like 3,4-methylenedioxymethamphetamine (MDMA).
(r = − 0.46, Po 0.05). BOLD, blood oxygen-level dependent; LSD, The attenuated amygdala reactivity observed in this study is in
lysergic acid diethylamide.
good accordance with those findings and possibly reflects a
neural basis for such effects, which might also be therapeutically
psychotherapy.12 Our results are consistent with our previously beneficial.49,50 However, and in contrast to substances like
reported findings in a facial emotion recognition task, showing selective serotonin reuptake inhibitors, the positive long-term
that LSD-impaired recognition of fearful faces compared with effects of psychedelics reported by recent studies8–12,15,51 outlast
placebo.20 Our results are also in line with findings obtained after the acute pharmacological effects. It should be further investi-
administration of psilocybin, where attenuated recognition of gated how psychological and biological factors, like
negative facial expressions21,22 and reduced amygdala blood neuroplasticity,52 contribute to these long-term effects.
oxygen-level dependent response to fearful faces23 were reported. Our study has several limitations. First, although the trial was
The psilocybin-induced attenuation of amygdala reactivity in formally double-blinded, assignment to placebo or LSD was
response to negative stimuli has consistently been shown to be unavoidably unblinded by the obvious psychedelic effects caused
related to the psilocybin-induced increase in positive mood.23 by the dose used. Second, we did not include in our analyses
It could be argued that the decreased responsiveness of the measures of negative affect. Third, we can only provide data about
amygdala under LSD was due to a drug-induced alteration in one moderate dose. Higher doses of psychedelics are possibly
visual perception, resulting in the inability to differentiate difficult to use with fMRI, because they are more likely to induce
between the presented facial expressions. However, our results anxiety,45 although the overall effects are still described as positive in
in two doses of LSD (100 μg and 200 μg, respectively) indicated, the higher doses investigated.2,45 The observed anxiolytic effect
that LSD specifically impaired recognition of fearful faces, while it probably also depends on personal and environmental factors and
did not significantly affect recognition of neutral, happy and angry might thus be different in the mentally ill or in uncontrolled settings.
faces.20 Furthermore, subjects in the present study performed well
in the gender differentiation task and our whole brain results
showed activation in regions typically involved in processing of CONFLICT OF INTEREST
neutral and fearful faces, respectively.14 The authors declare no conflict of interest.
We observed a significant effect of LSD on the left amygdala.
Several studies suggest, that the left amygdala might be
particulary involved in processing of negative facial ACKNOWLEDGMENTS
expressions.24,25 It has also been reported that the left amygdala We express our thanks to Dr Sarah Longhi. This work was supported by the Swiss
shows lesser habituation to fearful stimuli compared with the right National Science Foundation (grant no. 320030_170249 to MEL and SB).
amygdala, which might make it more likely to detect the blood
oxygen-level dependent changes in this area.26,27 However,
lateralization of the amygdala response is still controversial REFERENCES
discussed.28–30 The amygdala receives considerable serotonergic 1 Nichols DE. Psychedelics. Pharmacol Rev 2016; 68: 264–355.
2 Schmid Y, Enzler F, Gasser P, Grouzmann E, Preller KH, Vollenweider FX et al. Acute
innervation from the raphe nuclei.31,32 LSD mainly acts as an
effects of lysergic acid diethylamide in healthy subjects. Biol Psychiatry 2015; 78:
agonist at the serotonin 5-HT2A receptor,1,4 which is expressed in 544–553.
most parts of the amygdala.33 Accordingly, this might provide a 3 Carhart-Harris RL, Kaelen M, Bolstridge M, Williams TM, Williams LT, Underwood R
psychopharmacological basis for the observed effect of LSD on et al. The paradoxical psychological effects of lysergic acid diethylamide (LSD).
this structure. After administration of selective serotonin reuptake Psychol Med 2016; 46: 1–12.
inhibitors in healthy subjects, a decreased amygdala activity in 4 Vollenweider FX, Vollenweider-Scherpenhuyzen MF, Babler A, Vogel H, Hell D.
response to negative stimuli was reported.34,35 In depressed Psilocybin induces schizophrenia-like psychosis in humans via a serotonin-2
patients, amygdala hyperreactivity was resolved after treatment agonist action. Neuroreport 1998; 9: 3897–3902.
5 Carhart-Harris RL, Kaelen M, Whalley MG, Bolstridge M, Feilding A, Nutt DJ. LSD
with a selective serotonin reuptake inhibitor.36,37 Those findings
enhances suggestibility in healthy volunteers. Psychopharmacology (Berl) 2015;
indicate that the serotonin system is involved in the modulation of 232: 785–794.
the amygdala response to emotional stimuli.38 We also observed 6 Dolder PC, Schmid Y, Haschke M, Rentsch KM, Liechti ME. Pharmacokinetics and
decreased activity in the right medial prefrontal cortex (mPFC). concentration-effect relationship of oral LSD in humans. Int J Neuropsycho-
The mPFC is anatomically connected to the amygdala39 and pharmacol 2015; 19: pyv072.
67
Translational Psychiatry (2017), 1 – 5
Acute effects of LSD on amygdala activity in healthy subjects
F Mueller et al
5
7 Strajhar P, Schmid Y, Liakoni E, Dolder PC, Rentsch KM, Kratschmar DV et al. Acute 33 Bombardi C. Neuronal localization of the 5-HT2 receptor family in the amygdaloid
effects of LSD on circulating steroid levels in healthy subjects. J Neuroendocrinol complex. Front Pharmacol 2014; 5: 68.
2016; 28: 12374. 34 Murphy SE, Norbury R, O'Sullivan U, Cowen PJ, Harmer CJ. Effect of a single dose
8 Johnson MW, Garcia-Romeu A, Cosimano MP, Griffiths RR. Pilot study of the of citalopram on amygdala response to emotional faces. Br J Psychiatry 2009; 194:
5-HT2AR agonist psilocybin in the treatment of tobacco addiction. J Psycho- 535–540.
pharmacol 2014; 28: 983–992. 35 Harmer CJ, Mackay CE, Reid CB, Cowen PJ, Goodwin GM. Antidepressant drug
9 Bogenschutz MP, Forcehimes AA, Pommy JA, Wilcox CE, Barbosa PC, Strassman treatment modifies the neural processing of nonconscious threat cues. Biol Psy-
RJ. Psilocybin-assisted treatment for alcohol dependence: a proof-of- chiatry 2006; 59: 816–820.
concept study. J Psychopharmacol 2015; 29: 289–299. 36 Godlewska BR, Norbury R, Selvaraj S, Cowen PJ, Harmer CJ. Short-term SSRI
10 Grob CS, Danforth AL, Chopra GS, Hagerty M, McKay CR, Halberstadt AL et al. Pilot treatment normalises amygdala hyperactivity in depressed patients. Psychol Med
study of psilocybin treatment for anxiety in patients with advanced-stage cancer. 2012; 42: 2609–2617.
Arch Gen Psychiatry 2011; 68: 71–78. 37 Sheline YI, Barch DM, Donnelly JM, Ollinger JM, Snyder AZ, Mintun MA. Increased
11 Carhart-Harris RL, Bolstridge M, Rucker J, Day CM, Erritzoe D, Kaelen M et al. amygdala response to masked emotional faces in depressed subjects resolves
Psilocybin with psychological support for treatment-resistant depression: an with antidepressant treatment: an fMRI study. Biol Psychiatry 2001; 50: 651–658.
open-label feasibility study. Lancet Psychiatry 2016; 3: 619–627. 38 Bocchio M, McHugh SB, Bannerman DM, Sharp T, Capogna M. Serotonin, amyg-
12 Gasser P, Kirchner K, Passie T. LSD-assisted psychotherapy for anxiety associated dala and fear: assembling the puzzle. Front Neural Circuits 2016; 10: 24.
with a life-threatening disease: a qualitative study of acute and sustained sub- 39 Ghashghaei HT, Hilgetag CC, Barbas H. Sequence of information processing for
jective effects. J Psychopharmacol 2015; 29: 57–68. emotions based on the anatomic dialogue between prefrontal cortex and
13 Adolphs R. Fear, faces, and the human amygdala. Curr Opin Neurobiol 2008; 18: 166–172. amygdala. Neuroimage 2007; 34: 905–923.
14 Fusar-Poli P, Placentino A, Carletti F, Landi P, Allen P, Surguladze S et al. Functional 40 Etkin A, Egner T, Kalisch R. Emotional processing in anterior cingulate and medial
atlas of emotional faces processing: a voxel-based meta-analysis of 105 functional prefrontal cortex. Trends Cogn Sci 2011; 15: 85–93.
magnetic resonance imaging studies. J Psychiatry Neurosci 2009; 34: 418–432. 41 Robinson OJ, Charney DR, Overstreet C, Vytal K, Grillon C. The adaptive threat bias
15 Bedi G, Phan KL, Angstadt M, de Wit H. Effects of MDMA on sociability and neural in anxiety: amygdala-dorsomedial prefrontal cortex coupling and aversive
response to social threat and social reward. Psychopharmacology (Berl) 2009; 207: amplification. Neuroimage 2012; 60: 523–529.
73–83. 42 Robinson OJ, Krimsky M, Lieberman L, Allen P, Vytal K, Grillon C. Towards a
16 Thirion B, Pinel P, Meriaux S, Roche A, Dehaene S, Poline JB. Analysis of a large mechanistic understanding of pathological anxiety: the dorsal medial prefrontal-
fMRI cohort: Statistical and methodological issues for group analyses. Neuroimage amygdala 'aversive amplification' circuit in unmedicated generalized and social
2007; 35: 105–120. anxiety disorders. Lancet Psychiatry 2014; 1: 294–302.
17 Schmidt A, Borgwardt S, Gerber H, Wiesbeck GA, Schmid O, Riecher-Rossler A 43 Robinson OJ, Krimsky M, Lieberman L, Vytal K, Ernst M, Grillon C. Anxiety-
et al. Acute effects of heroin on negative emotional processing: relation of potentiated amygdala-medial frontal coupling and attentional control. Transl
amygdala activity and stress-related responses. Biol Psychiatry 2014; 76: 289–296. Psychiatry 2016; 6: e833.
18 Domes G, Heinrichs M, Kumbier E, Grossmann A, Hauenstein K, Herpertz SC. 44 Robinson OJ, Overstreet C, Allen PS, Letkiewicz A, Vytal K, Pine DS et al. The role of
Effects of intranasal oxytocin on the neural basis of face processing in autism serotonin in the neurocircuitry of negative affective bias: serotonergic modulation
spectrum disorder. Biol Psychiatry 2013; 74: 164–171. of the dorsal medial prefrontal-amygdala 'aversive amplification' circuit. Neuro-
19 Passie T, Halpern JH, Stichtenoth DO, Emrich HM, Hintzen A. The pharmacology of image 2013; 78: 217–223.
lysergic acid diethylamide: a review. CNS Neurosci Ther 2008; 14: 295–314. 45 Griffiths RR, Johnson MW, Richards WA, Richards BD, McCann U, Jesse R. Psilo-
20 Dolder PC, Schmid Y, Muller F, Borgwardt S, Liechti ME. LSD acutely impairs fear cybin occasioned mystical-type experiences: immediate and persisting dose-
recognition and enhances emotional empathy and sociality. Neuropsycho- related effects. Psychopharmacology (Berl) 2011; 218: 649–665.
pharmacology 2016; 41: 2638–2646. 46 Groenewold NA, Opmeer EM, de Jonge P, Aleman A, Costafreda SG. Emotional
21 Kometer M, Schmidt A, Bachmann R, Studerus E, Seifritz E, Vollenweider FX. valence modulates brain functional abnormalities in depression: evidence from a
Psilocybin biases facial recognition, goal-directed behavior, and mood state meta-analysis of fMRI studies. Neurosci Biobehav Rev 2013; 37: 152–163.
toward positive relative to negative emotions through different serotonergic 47 Etkin A, Wager TD. Functional neuroimaging of anxiety: a meta-analysis of
subreceptors. Biol Psychiatry 2012; 72: 898–906. emotional processing in PTSD, social anxiety disorder, and specific phobia. Am J
22 Schmidt A, Kometer M, Bachmann R, Seifritz E, Vollenweider F. The NMDA Psychiatry 2007; 164: 1476–1488.
antagonist ketamine and the 5-HT agonist psilocybin produce dissociable effects 48 Mithoefer MC, Wagner MT, Mithoefer AT, Jerome L, Doblin R. The safety and
on structural encoding of emotional face expressions. Psychopharmacology (Berl) efficacy of {+/-}3,4-methylenedioxymethamphetamine-assisted psychotherapy in
2013; 225: 227–239. subjects with chronic, treatment-resistant posttraumatic stress disorder: the first
23 Kraehenmann R, Preller KH, Scheidegger M, Pokorny T, Bosch OG, Seifritz E et al. randomized controlled pilot study. J Psychopharmacol 2011; 25: 439–452.
Psilocybin-induced decrease in amygdala reactivity correlates with enhanced 49 Oehen P, Traber R, Widmer V, Schnyder U. A randomized, controlled pilot study of
positive mood in healthy volunteers. Biol Psychiatry 2015; 78: 572–581. MDMA (±3,4-methylenedioxymethamphetamine)-assisted psychotherapy for
24 Morris JS, Frith CD, Perrett DI, Rowland D, Young AW, Calder AJ et al. A differential treatment of resistant, chronic post-traumatic stress disorder (PTSD). J Psycho-
neural response in the human amygdala to fearful and happy facial expressions. pharmacol 2013; 27: 40–52.
Nature 1996; 383: 812–815. 50 Mithoefer MC, Wagner MT, Mithoefer AT, Jerome L, Martin SF, Yazar-Klosinski B
25 Blair RJ, Morris JS, Frith CD, Perrett DI, Dolan RJ. Dissociable neural responses to et al. Durability of improvement in post-traumatic stress disorder symptoms and
facial expressions of sadness and anger. Brain 1999; 122(Pt 5): 883–893. absence of harmful effects or drug dependency after 3,4-methylenediox-
26 Wright CI, Fischer H, Whalen PJ, McInerney SC, Shin LM, Rauch SL. Differential ymethamphetamine-assisted psychotherapy: a prospective long-term follow-
prefrontal cortex and amygdala habituation to repeatedly presented emotional up study. J Psychopharmacol 2013; 27: 28–39.
stimuli. Neuroreport 2001; 12: 379–383. 51 Osorio Fde L, Sanches RF, Macedo LR, Santos RG, Maia-de-Oliveira JP, Wichert-Ana
27 Phillips ML, Medford N, Young AW, Williams L, Williams SC, Bullmore ET et al. Time L et al. Antidepressant effects of a single dose of ayahuasca in patients with
courses of left and right amygdalar responses to fearful facial expressions. Hum recurrent depression: a preliminary report. Rev Bras Psiquiatr 2015; 37: 13–20.
Brain Mapp 2001; 12: 193–202. 52 Vollenweider FX, Kometer M. The neurobiology of psychedelic drugs: implications
28 Sergerie K, Chochol C, Armony JL. The role of the amygdala in emotional pro- for the treatment of mood disorders. Nat Rev Neurosci 2010; 11: 642–651.
cessing: a quantitative meta-analysis of functional neuroimaging studies. Neurosci
Biobehav Rev 2008; 32: 811–830.
29 Baas D, Aleman A, Kahn RS. Lateralization of amygdala activation: a systematic This work is licensed under a Creative Commons Attribution 4.0
review of functional neuroimaging studies. Brain Res Brain Res Rev 2004; 45: 96–103. International License. The images or other third party material in this
30 Wager TD, Phan KL, Liberzon I, Taylor SF. Valence, gender, and lateralization of article are included in the article’s Creative Commons license, unless indicated
functional brain anatomy in emotion: a meta-analysis of findings from neuroi- otherwise in the credit line; if the material is not included under the Creative Commons
maging. Neuroimage 2003; 19: 513–531. license, users will need to obtain permission from the license holder to reproduce the
31 Hensler JG. Serotonergic modulation of the limbic system. Neurosci Biobehav Rev material. To view a copy of this license, visit http://creativecommons.org/licenses/
2006; 30: 203–214. by/4.0/
32 Asan E, Steinke M, Lesch KP. Serotonergic innervation of the amygdala: targets,
receptors, and implications for stress and anxiety. Histochem Cell Biol 2013; 139:
785–813. © The Author(s) 2017

68
Translational Psychiatry (2017), 1 – 5
3.6 Publication 6

Development and validation of an LC-MS/MS method to quantify


LSD, iso-LSD, 2-oxo-3-hydroxy LSD, and nor-LSD and identify novel
metabolites in plasma samples in a controlled clinical trial

Patrick C. Dolder1,2, Matthias E. Liechti2, Katharina M. Rentsch1

1 Laboratory Medicine, University Hospital Basel and University of Basel, Switzerland


2 Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of
Clinical Research, University Hospital Basel and University of Basel, Switzerland

69
|
Received: 22 December 2016    Accepted: 25 April 2017

DOI: 10.1002/jcla.22265

RESEARCH ARTICLE

Development and validation of an LC-­MS/MS method


to quantify lysergic acid diethylamide (LSD), iso-­LSD,
2-­oxo-­3-­hydroxy-­LSD, and nor-­LSD and identify novel
metabolites in plasma samples in a controlled clinical trial

Patrick C. Dolder1,2 | Matthias E. Liechti1 | Katharina M. Rentsch1

1
Laboratory Medicine, University Hospital and
University of Basel, Basel, Switzerland Background: Lysergic acid diethylamide (LSD) is a widely used recreational drug. The
2
Division of Clinical Pharmacology and aim of this study was to develop and validate a liquid chromatography tandem mass
Toxicology, Department of Biomedicine and spectrometry (LC-­
MS/MS) method for the quantification of LSD, iso-­
LSD,
Department of Clinical Research, University
Hospital and University of Basel, Basel, 2-­oxo-­3-­hydroxy LSD (O-­H-­LSD), and nor-­LSD in plasma samples from 24 healthy
Switzerland subjects after controlled administration of 100 μg LSD in a clinical trial. In addition,
Correspondence metabolites that have been recently described in in vitro studies, including lysergic
Katharina M. Rentsch, Laboratory Medicine, acid monoethylamide (LAE), lysergic acid ethyl-­2-­hydroxyethylamide (LEO), 2-­oxo-­LSD,
University Hospital Basel, Basel, Switzerland.
Email: [email protected] trioxylated-­LSD, and 13/14-hydroxy-LSD, should be identified.

Funding information Methods: Separation of LSD and its metabolites was achieved on a reversed phase
This work was supported by the Swiss chromatography column after turbulent-­flow online extraction. For the identification
National Science Foundation (grant no.
320030_170249 to ML). and quantification, a triple-­stage quadrupole LC-­MS/MS instrument was used.
Results: The validation data showed slight matrix effects for LSD, iso-­LSD, O-­H-­LSD,
or nor-­LSD. Mean intraday and interday accuracy and precision were 105%/4.81% and
105%/4.35% for LSD, 98.7%/5.75% and 99.4%/7.21% for iso-­LSD, 106%/4.54% and
99.4%/7.21% for O-­H-­LSD, and 107%/5.82% and 102%/5.88% for nor-­LSD, respec-
tively. The limit of quantification was 0.05 ng/mL for LSD, iso-­LSD, and nor-­LSD and
0.1 ng/mL for O-­H-­LSD. The limit of detection was 0.01 ng/mL for all compounds.
Conclusion: The method described herein was accurate, precise, and the calibration
range within the range of expected plasma concentrations. LSD was quantified in the
plasma samples of the 24 subjects of the clinical trial, whereas iso-­LSD, O-­H-­LSD,
­nor-­LSD, LAE, LEO, 13/14-hydroxy-LSD, and 2-oxo-LSD could only sporadically be
­detected but were too low for quantification.

KEYWORDS
controlled study, LC-MS, LSD, lysergic acid diethylamide, metabolism, plasma

1 | INTRODUCTION reintroduced in psychiatric research2-16 and investigated as an adjunct


to psychotherapy.17,18 Therefore, information about its metabolism
Lysergic acid diethylamide (LSD) is a psychoactive substance that al- and pharmacokinetics after controlled intake has received increasing
ters states of consciousness and perception. Its psychedelic effects interest. Doses that were used in recent clinical studies ranged from
made it popular as a recreational drug, especially in the 1960s and 75 μg, i.v.,2-11 to 200 μg, p.o.,12-15,19 resulting in low blood and urine
1
1970s, but LSD is still widely used today. In addition, LSD has been concentrations.12,20 Dolder et al. and Steuer et al. recently showed

J Clin Lab Anal. 2017;e22265. wileyonlinelibrary.com/journal/jcla © 2017 Wiley Periodicals, Inc.  |  1 of 8
https://doi.org/10.1002/jcla.22265
|
2 of 8       DOLDER et al.

that LSD and its main urinary metabolite 2-­


oxo-­
3-­
hydroxy-­
LSD used for chromatographic separation. The online extraction system
­(O-­H-­LSD) were detectable in plasma after controlled intake of 200 μg was coupled to a TSQ Endura triple-­stage mass spectrometer (Thermo
LSD in 16 healthy subjects12,20 and clinical toxicological cases of acute Scientific) using APCI in positive mode because of its better perfor-
LSD overdose.21 Studies of in vitro metabolism have further identi- mance with regard to matrix effects.24,25
fied lysergic acid monoethylamide (LAE), lysergic acid ethyl-2-hy-
droxyethylamide (LEO), 2-­
oxo-­
LSD, nor-­
LSD, trioxylated-­
LSD, and
2.2.2 | Liquid chromatography method
13/14-hydroxy-LSD as glucuronides,22,23 but no systematic infor-
mation is available regarding their presence in human plasma after For LC, three mobile phases were used in gradient mode for extrac-
controlled intake of LSD. However, recent investigations confirmed tion and analytical chromatography. Mobile phase A consisted of
the presence of 2-­oxo-­LSD and 13/14-hydroxy-LSD (glucuronides) 20 mmol/L ammonium acetate in water and 0.1% formic acid. Mobile
in plasma samples after controlled intake of 200 μg LSD.20 The aim phase B consisted of 20 mmol/L ammonium acetate in methanol
of this study was to develop a sensitive turboflow liquid chromatog- and acetonitrile (1:1) that contained 0.1% formic acid. Mobile phase
raphy tandem mass spectrometry (LC-­MS/MS) method to quantify C was an organic mixture of acetonitrile, acetone, and 2-­propanol
LSD, iso-­LSD, O-­H-­LSD, and nor-­LSD and potentially identify LAE, (1:1:1). Chromatography was run in isocratic mode with 70% mobile
LEO, 2-­oxo-­LSD, trioxylated-­LSD, and 13/14-hydroxy-LSD (glucuro- phase A and 30% mobile phase B, with a run time of 11 minutes and
nides) in human plasma samples. The method was developed using four additional minutes for flushing and equilibration using mobile
a triple-­stage quadrupole LC-­MS/MS instrument in selected reaction phase C.
monitoring (SRM) mode after atmospheric pressure ionization (APCI).
Our method was established and successfully applied to the analysis
2.2.3 | Mass spectrometry conditions
of plasma samples from healthy volunteers after the intake of 100 μg
LSD in a controlled clinical study. The positive ion discharge current was set to 5 μA. The vaporizer
temperature was optimized to 400°C. Sheath and auxiliary gas pro-
vided the best results, with flow rates of 15 and 5 arbitrary units,
2 | MATERIALS AND METHODS
respectively. The temperature of the ion transfer tube was set to
300°C. The system was tuned and optimized for the detection of LSD.
2.1 | Chemicals and reagents
LSD and its metabolites were detected using SRM of the two to three
Acetonitrile, acetone, methanol, 2-­propanol, formic acid, and acetic most intense ion transitions. Analytes were identified when quanti-
acid with high-­
performance liquid chromatography (HPLC)-­
grade fier and qualifier ions were present within the given retention time.
purity were all purchased from Merck (Darmstadt, Germany). HPLC-­ Structures, transitions, and respective collision energies are shown in
grade ammonium acetate and ammonium carbonate were obtained Figure 1.
from Merck. Distilled water was obtained from an in-­house installed
purifier (ELGA, Bucks, UK). Drug-­free plasma samples (contain-
2.3 | Standard solutions
ing lithium-­heparin as an anticoagulant) serving as negative control,
and blank matrices were obtained from coworkers. LSD and LSD-­d3 Stock solutions that contained 100 μg/mL LSD, 100 μg/mL LSD-­d3,
as 1 mg/mL reference standards in acetonitrile were obtained from 10 μg/mL iso-­LSD, 10 μg/mL O-­H-­LSD, or 10 μg/mL nor-­LSD in ace-
Lipomed (Arlesheim, Switzerland). O-­H-­LSD and iso-­LSD as 0.1 mg/ tonitrile were prepared and stored in light-­protected brown glass vials
mL reference standards in acetonitrile were obtained from Cerilliant at −20°C. All of the solutions were prepared in duplicate to have dif-
(Round Rock, TX, USA). Nor-­LSD in powder form was obtained ferent sets for quality control (QC) and calibration samples. Working
from Toronto Research Chemicals (Toronto, Canada). The non-­ solutions of each analyte at 0.1 μg/mL in purified water/acetonitrile
commercially available metabolites LAE, LEO, 2-­oxo-­LSD, trioxylated-­ were used for the preparation of QC and calibration samples and ma-
LSD, and 13/14-hydroxy-LSD (glucuronides) were extracted from trix and selectivity experiments. Because of the instability of LSD and
pooled 24-­h urine samples as described in Results section. to minimize possible degradation by various freeze-­thaw cycles, 1 mL
aliquots of stock and working solutions were prepared.

2.2 | LC-­MS/MS analysis
2.4 | Sample preparation
2.2.1 | Equipment
Study samples were sorted according to drug condition (LSD or pla-
The HPLC system (Transcend TLX1 HPLC; Thermo Scientific, Basel, cebo) and subject (S1-­24). Calibrators, controls and subject samples
Switzerland) consisted of two Accela 1250 pumps for loading and were thawed once, and 100 μL aliquots was taken to minimize the
eluting. The autosampler and sample extraction system were con- freeze-­thaw cycles. To 100 μL of plasma, 110 μL of an acetonitrile/
trolled by the Aria MX 2.1 software (Thermo Scientific). A cyclone P LSD-­d3 solution (0.01 μg/mL) was added. The samples were then vig-
turboflow column (Thermo Scientific) was used for extraction, and a orously vortexed and centrifuged for 10 minutes at 13 200 g, and the
Zorbax Eclipse XDB-­C8 column (Agilent, Santa Clara, CA, USA) was supernatant was then transferred to 96-­well plates.
71
DOLDER et al. |
      3 of 8

Lysergic-acid-diethylamide (LSD) Iso-LSD Nor-LSD 2-oxo-3-hydroxy-LSD (O-H-LSD)

N N N N
H H H H H
O N O N O N O N
H H H H

OH

N N N N

Retenon me: Retenon me: Retenon me: Retenon me:


8.50 min 9.10 min 8.85 min 4.52 min
Transions: Collision Energy (V) Transions: Collision Energy (V) Transions: Collision Energy (V) Transions: Collision Energy (V)
324 → 223 24 324 → 223 24 310 → 237 21 356 → 338 14
324 → 208 31 324 → 208 31 310 → 209 24 356 → 265 17
324 → 197 24 324 → 197 24 310 → 192 39 356 → 237 22

Lysergic-acid-monoethylamide (LAE) Lysergic-acid-ethyl-hydroxyethylamide (LEO) 13/14-hydroxy-LSD 2-oxo-LSD


OH
H N N
N
N H H H
H
H O N O N
O N
O N H H
H
H

O
N N
N HO
N
Retenon me: Retenon me: Retenon me: Retenon me:
5.31 min 5.14 min 4.54 min 5.35 min
Transions: Collision Energy (V) Transions: Collision Energy (V) Transions: Collision Energy (V) Transions: Collision Energy (V)
296 → 253 24 340 → 223 24 358 → 340 24 340 → 223 24
296 → 223 24 340 → 239 22 358 → 239 22 340 → 208 30
296 → 208 30

F I G U R E   1   Structure, retention time, ion transitions, and collision energies of lysergic acid diethylamide (LSD) and selected metabolites

used in the present method, the extraction step comprised only pro-
2.5 | Experiments
tein precipitation. All of the samples were processed through the
turbulent-­flow extraction column. Five plasma samples were spiked
2.5.1 | Calibration
to concentrations between 0.05 and 10 ng/mL for LSD, iso-­LSD,
Six calibration standards were prepared by spiking plasma samples O-­H-­LSD, and nor-­LSD. The samples were measured before and
with LSD, iso-­LSD, and nor-­LSD to concentrations of 0.05, 0.1, 0.5, after extraction and in neat solution. The peak areas of the spiked
1, 5, and 10 ng/mL plus blank (matrix only) and zero sample (ma- samples after extraction were then compared with the area of the
trix plus internal standard). Five calibrators were used for O-­H-­LSD spiked mobile phase to calculate matrix effects. Recovery values
with concentrations of 0.05, 0.1, 0.5, 1, 5, and 10 ng/mL plus blank were calculated as the areas of standards that were spiked before
(matrix only) and zero sample (matrix plus internal standard). The extraction divided by the areas of standards that were spiked after
highest calibration point in plasma was adopted from our previ- extraction. The process efficiency was adopted from Matuszewski
ously developed method and pharmacokinetic-­
pharmacodynamic et al.27 and calculated as the ratio between the area of the stand-
12,21
data. The calibration curves were linearly fitted using a weight- ard spiked before extraction and the areas of the standard in neat
2
ing factor of 1/x . solution.

2.5.2 | Selectivity 2.5.4 | Stability
Following U.S. Food and Drug Administration validation guidelines,26 The determination of long-­term stability was based on Li et al. and
we collected plasma samples from six different healthy volunteers Klette et al., in which LSD is regarded as stable under storage condi-
and tested them for interference to establish selectivity. We further tions of −20°C.28,29 However, LSD is known to be very unstable and
analyzed samples from the placebo condition to confirm the absence vulnerable to air, light, and heat. Even ambient temperature (20-­25°C)
of LSD. and normal light conditions can lead to a decrease in LSD concentra-
tions. Therefore, we assessed bench-­top stability and autosampler
stability with multiple measurements of calibration and QC samples
2.5.3 | Matrix effects and recovery
within 24 h. For autosampler stability, the samples were kept in light-­
Matrix effects, recovery, and process efficiency were measured and protected, sealed, 96-­well deep-­well plates at 4°C in the autosampler
calculated according to Matuszewski et al.27 In regard of the vulner- until injection. During the study, the samples were drawn through
ability to light and air and because of the online extraction that was an intravenous catheter into lithium-­
heparin tubes and directly
72
4 of 8       | DOLDER et al.

(A) RT: 8.85


100
Nor-LSD
50
8.53 9.52
0
0 1 2 3 4 5 6 7 8 9 10 11 12
RT: 8.50
100 LSD RT: 9.14

50 iso-LSD LSD iso-LSD


0
0 1 2 3 4 5 6 7 8 9 10 11 12
RT: 4.52
100 2-oxo-3-hydroxy LSD
50

0
0 1 2 3 4 5 6 7 8 9 10 11 12
100 Lysergic acid monoethylamide RT: 5.31
50 (LAE)
6.30 8.63
7.82
0
0 1 2 3 4 5 6 7 8 9 10 11 12
100
Lysergic acid ethyl-2-hydroxyethylamide RT: 5.14
50 (LEO)
4.40 6.17
0
0 1 2 3 4 5 6 7 8 9 10 11 12
100 2-oxy LSD RT: 5.35

50
6.16
0
0 1 2 3 4 5 6 7 8 9 10 11 12
100 Dihydroxy LSD RT: 4.54
50
4.80 5.51
0
0 1 2 3 4 5 6 7 8 9 10 11 12
100 Trioxo LSD RT: 5.36
50

0
0 1 2 3 4 5 6 7 8 9 10 11 12

(B)
100 (1)
90

80

70
Relative Abundance

60

50

40 LSD-d3
30
(5)

20 (2)
(7) (4) (6) (8)
10

0
0 1 2 3 4 5 6 7 8 9 10 11 12
Time (min)

F I G U R E   2   (A) Chromatogram of selected metabolites. Lysergic acid diethylamide (LSD), iso-­LSD, nor-­LSD, and 2-­oxo-­3-­hydroxy-­LSD are
spiked at 1 ng/mL in plasma; the concentration of lysergic-­acid monoethylamide, lysergic-­acid-­ethyl-­2-­hydroxyethylamide, 13/14-hydroxy-LSD,
and 2-­oxo-­LSD is unknown. (B) Chromatogram of a healthy volunteer 4 h after administration of 100 μg LSD. Arrows are indicating peaks of
LSD (1), iso-­LSD (2), nor-­LSD (3) and 2-­oxo-­3-­hydroxy-­LSD (4), lysergic-­acid monoethylamide (5), lysergic-­acid-­ethyl-­2-­hydroxyethylamide (6),
13/14-hydroxy-LSD (7), and 2-­oxo-­LSD (8)

73
DOLDER et al. |
      5 of 8

centrifuged, and the plasma was stored at −20°C at the study site for LSD, iso-­LSD, and nor-­LSD, and 0.1, 1, and 10 ng/mL for O-­H-­LSD,
before transferring to the laboratory for analysis. Due to the known respectively.
vulnerability of LSD, calibrators and quality controls were freshly
weighted every week and single aliquots were stored at −20°C. A
new calibration was run every day and with every study subject. 3  | RESULTS

Lysergic acid diethylamide, LSD-­


d3, iso-­
LSD, and the metabolites
2.5.5 | Lower limits of detection and quantification
nor-­LSD, LAE, LEO, 2-­oxo-­LSD, trioxylated-­LSD, and 13/14-hydroxy-
Drug-­free plasma samples were spiked with different concentrations LSD (glucuronides) eluted between 4 and 11 minutes. The chroma-
of LSD, iso-­LSD, O-­H-­LSD, and nor-­LSD for determination of the limit tographic separation of spiked samples and selected metabolites is
of quantification (LOQ) and the limit of detection (LOD). The LOQ con- depicted in Figure 2A, and the chromatogram of a subject’s sample
centrations had to give a response at least five times greater than the 4 h after LSD administration is presented in Figure 2B.
blank. In addition, precision had to be <20%, and accuracy had to be
80%-­120% using at least five determinations per matrix and concen-
3.1 | Selectivity
tration. The LOD concentration was determined as the lowest discrim-
inable peak in the region of a signal-­to-­noise ratio greater than five. None of the six plasma samples showed any interference within the
measured mass range and time frame (Figure 3). Furthermore, none
of the measured plasma samples from the placebo condition showed
2.5.6 | Carryover
any interference.
For the determination of the carryover, different blank plasma sam-
ples were run between patient samples, highest calibrations, and qual-
3.2 | Matrix effects and recovery
ity controls.
The plasma matrix effects were 125% for LSD, 119% for iso-­LSD,
103% for O-­
H-­
LSD, and 118% for nor-­
LSD at concentrations of
2.5.7 | Reproducibility
10 ng/mL, consistent with a slight ion enhancement for LSD, iso-­LSD,
26
According to U.S. Food and Drug Administration guidelines, the and nor-­LSD. Recoveries were calculated as 70%-­90% for all sub-
reproducibility of quantification was determined by measuring each stances at 10 ng/mL. Process efficiencies were 113% for LSD, 86%
QC sample five times in 1 day to establish intraday precision and ac- for iso-­LSD, 77% for O-­H-­LSD, and 93% for nor-­LSD.
curacy. Each QC sample was also measured for five consecutive days
to determine interday precision and accuracy. All of the values had
3.3 | Stability
to meet the criteria of a coefficient of variation (CV) <15%, response
<20% at the LOQ, and accuracy of 80%-­120%. To demonstrate the The concentrations of the processed samples decreased up to
accuracy and precision of the method, we used three QCs (low, me- −60% within 24 hours at ambient temperature (20-­23°C). The
dium, and high). The QC concentrations were 0.05, 1, and 10 ng/mL concentrations of the extracted and sealed plasma samples that

100

90

80

70
Relative Abundance

60

50

40

30

20

10

0
0 1 2 3 4 5 6 7 8 9 10 11 12
Time (min)

F I G U R E   3   Chromatogram of 6 blank plasma samples from six different subjects, and a blank sample containing lysergic acid diethylamide-­d3
74
6 of 8       | DOLDER et al.

were stored within the closed autosampler at 4°C were stable up


3.7 | Reproducibility
to 24 hours.
All of the substances fulfilled the accuracy and precision criteria. The
mean intraday accuracy and precision were 105% and 4.81% for LSD,
3.4 | Lower limits of detection and quantification
98.7% and 5.75% for iso-­LSD, 106% and 4.54% for O-­H-­LSD, and
The LOQ was 0.05 ng/mL for LSD, iso-­LSD, and nor-­LSD. For O-­H-­ 107% and 5.82% for nor-­LSD, respectively. The mean interday accu-
LSD, the respective concentration was 0.1 ng/mL. The LODs were racy and precision were 105% and 4.35% for LSD, 99.4% and 7.21%
0.01 ng/mL for all compounds. for iso-­LSD, 99.4% and 7.21% for O-­H-­LSD, and 102% and 5.88% for
nor-­LSD, respectively.

3.5 | Carryover
3.8 | Identification of non-­commercially available
No carryover was found for LSD, iso-­LSD, O-­H-­LSD, or nor-­LSD in
LSD metabolites
the plasma samples. Despite these results as a preventive measure, a
consecutive blank was always run after the highest calibrator (10 ng/ Lysergic acid diethylamide metabolites were extracted by liquid-­liquid
mL) and QC (10 ng/mL) during method development and the meas- extraction from pooled LSD-­positive 24-­h urine samples (8 L) to reach
urement of the study samples. high concentrations. One part of the concentrated metabolites was
kept for eventual quantification, and the second part was extracted
using industrial separation by automated thin-­layer chromatography
3.6 | Linearity
and purification. Separation was performed with generous support
Calibration curves in plasma were linear over the respective calibra- from Camag (Muttenz, Switzerland). Parent masses and selected tran-
tion ranges, with a mean correlation coefficient (R2) of 0.99. The cali- sitions for LC-­MS were adopted from Cai et al.22 and Canezin et al.23
bration curves (mean ± SEM) are shown in Figure 4. and replicated by injecting a mixture of the concentrated, extracted

Calibration curve LSD Calibration curve O-H-LSD


12 12
1.2 1.2
R2 = 0.99
O-H-LSD measured [ng/ml]

10
1.0
R2 = 0.99 10
1.0
LSD measured [ng/ml]

0.8 0.8

0.6 0.6

8 0.4
8 0.4

0.2 0.2

0.0 0.0
6 0.0 0.2 0.4 0.6 0.8 1.0 1.2
6 0.0 0.2 0.4 0.6 0.8 1.0 1.2

4 4

2 2

0 0
0 2 4 6 8 10 0 2 4 6 8 10
LSD spiked [ng/ml] O-H-LSD spiked [ng/ml]

Calibration curve nor-LSD Calibration curve iso-LSD


12 12
1.2 1.2

R2 = 0.99
nor-LSD measured [ng/ml]

1.0 R2 = 0.99 1.0


iso-LSD measured [ng/ml]

10 0.8 10 0.8

0.6 0.6

8 0.4
8
0.4

0.2 0.2

0.0 0.0
6 0.0 0.2 0.4 0.6 0.8 1.0 1.2
6 0.0 0.2 0.4 0.6 0.8 1.0 1.2

4 4

2 2

0 0
0 2 4 6 8 10 0 2 4 6 8 10
nor-LSD spiked [ng/ml] iso-LSD spiked [ng/ml]

F I G U R E   4   Calibration curves of lysergic acid diethylamide (LSD), iso-­LSD, nor-­LSD, and 2-­oxo-­3-­hydroxy-­LSD in human plasma 75
DOLDER et al. |
      7 of 8

metabolites. All of the identified metabolites from concentrated urine

1.2
24
samples (LAE, LEO, 2-­oxo-­LSD, trioxylated-­LSD, and 13/14-hydroxy-

3
LSD) were added to the quantification method before validation, for

1.4
23
qualitative screening of the study samples.

2
T A B L E   1   Measured plasma concentrations (Cmax) and the corresponding time points (Tmax) following oral administration of 100 μg lysergic acid diethylamide in 24 healthy subjects

1.1
22

3
3.9 | Samples

1.9
LSD (100 μg) and placebo were administered to 24 healthy ­subjects

21

2
(12 women, 12 men) in a double-­
blind, randomized, placebo-­

1.8
controlled, cross-­over study. The study was conducted in ­accordance

20

2
with the Declaration of Helsinki and International Conference on

0.3
Harmonization Guidelines in Good Clinical Practice (ICH-­GCP) and

19

1
approved by the Ethics Committee Northwest Switzerland and
Swiss Federal Office for Public Health, Bern, Switzerland. The study

1.3
18

2
was registered at ClinicalTrials.gov (NCT02308969). Plasma sam-
ples were collected at baseline and 1, 2, 3, 4, 6, 8, 10, 12, 16, and

1.9
17

1
24 h after LSD ­
administration. Maximum LSD plasma concentra-
tions of 1.3 ± 0.17 ng/mL (mean ± SEM) were determined (Table

0.8
16

1
1). Nor-­LSD could only be quantified in two subjects (3 and 4 hours
post-­administration), and LAE, LEO, 2-­oxy LSD, and 13/14-hydroxy-

1.4
15

2
LSD were detected in some of the samples. 13/14-hydroxy-LSD
glucuronides were undetectable because they were cleaved dur-

0.9
14

2
ing ionization. Detailed study descriptions, pharmacokinetic data,
and pharmacokinetic-­
pharmacodynamic analyses will be published

0.8
13

1
elsewhere.

3.2
12

1
4 | DISCUSSION AND CONCLUSION

0.8
11

3
With mean maximum plasma concentrations of LSD of ~1 ng/mL, the 2.0
10

2
development of analytical methods for quantification remains a chal-
lenge and brings LC-­MS technologies to their limits. For separation
3.3

of the different analytes, various columns have been used. Especially,


9

the separation of LSD and iso-­LSD was challenging, and only achieved
1.9

using the Zorbax Eclipse XDB-­


C8 column. However, the method
8

was only developed to chromatographically separate LSD, iso-­LSD,


1.4

nor-­LSD, and O-­H-­LSD. The non-­commercially available metabolites


7

were not available in sufficient amounts for extensive experiments.


1.5

Further, to improve sensitivity, different sample preparation proce-


6

dures (eg, liquid-­liquid extraction using chlorobutane and tert-­butyl-­


2.1

methylether) have been performed but have not led to significant


5

changes in the LOQ. Considering the light and air sensitivity of LSD
2.0

and the manual workload that is caused by liquid-­liquid extraction or


4

solid-­phase extraction, simple and fast protein precipitation has been


1.2

favored instead. APCI was equally to ESI regarding signal intensity but
3

gave slightly better results regarding matrix effects and was there-
1.2

fore favored. Overall, quantifying plasma samples between 12 and


2

24 hours after LSD administration requires techniques that provide


1.7

precise and sensitive measurements within the low picogram range.


1

This poses a challenge to quantifying LSD concentrations and also


makes it impossible to quantify or even identify new metabolites in
(ng/mL)
Subject

Tmax (h)

plasma samples after controlled intake of 100 μg LSD. In our recent


Cmax

investigations,12 we detected quantifiable plasma levels of O-­H-­LSD 76


|
8 of 8       DOLDER et al.

after the administration of 200 μg LSD. Steuer et al.20 additionally 16. Schmid Y, Enzler F, Gasser P, et al. Acute effects of lysergic acid dieth-
identified O-­H-­LSD and 13/14-hydroxy-LSD (glucuronides). We did ylamide in healthy subjects. Biol Psychiatry. 2015;78:544‐553.
17. Gasser P, Holstein D, Michel Y, et al. Safety and efficacy of lysergic
not expect to detect quantifiable concentrations of LSD metabo-
acid diethylamide-­assisted psychotherapy for anxiety associated with
lites after the administration of 100 μg LSD. The metabolites did not life-­threatening diseases. J Nerv Ment Dis. 2014;202:513‐520.
reach the LOD of our or other methods. Nevertheless, we sporadi- 18. Gasser P, Kirchner K, Passie T. LSD-­assisted psychotherapy for anxiety
cally detected the presence of metabolites in some plasma samples associated with a life-­threatening disease: a qualitative study of acute
and sustained subjective effects. J Psychopharmacol. 2015;29:57‐68.
and could confirm the presence of O-­H-­LSD, nor-­LSD, LEO, LAE, and
19. Schmid Y, Dolder PC, Liechti ME. Acute autonomic and psychotropic
13/14-hydroxy-LSD in plasma. To investigate the metabolism of LSD effects of LSD in healthy subjects in a placebo-­controlled study. Clin
more comprehensively, further studies that use higher doses of LSD Toxicol. 2015;53:359.
are required and metabolites need to be commercially available to de- 20. Steuer AE, Poetzsch M, Stock L, et al. Development and validation of
an ultra-­fast and sensitive microflow liquid chromatography-­tandem
velop comprehensive analytical methods for their quantification.
mass spectrometry (MFLC-­MS/MS) method for quantification of LSD
and its metabolites in plasma and application to a controlled LSD
administration study in humans. Drug Test Anal. 2016. https://doi.
REFERENCES
org/10.1002/dta.2042
1. Passie T, Halpern JH, Stichtenoth DO, et al. The pharmacology of ly- 21. Dolder PC, Liechti ME, Rentsch KM. Development and validation of
sergic acid diethylamide: a review. CNS Neurosci Ther. 2008;14:295‐ a rapid turboflow LC-­MS/MS method for the quantification of LSD
314. and 2-­oxo-­3-­hydroxy LSD in serum and urine samples of emergency
2. Carhart-Harris RL, Kaelen M, Whalley MG, et al. LSD enhances sug- toxicological cases. Anal Bioanal Chem. 2015;407:1577‐1584.
gestibility in healthy volunteers. Psychopharmacology. 2015;232:785‐ 22. Cai J, Henion J. Elucidation of LSD in vitro metabolism by liquid chro-
794. matography and capillary electrophoresis coupled with tandem mass
3. Kaelen M, Barrett FS, Roseman L, et al. LSD enhances the emotional spectrometry. J Anal Toxicol. 1996;20:27‐37.
response to music. Psychopharmacology. 2015;232:3607‐3614. 23. Canezin J, Cailleux A, Turcant A, et al. Determination of LSD and its
4. Carhart-Harris RL, Kaelen M, Bolstridge M, et al. The paradoxical psy- metabolites in human biological fluids by high-­performance liquid
chological effects of lysergic acid diethylamide (LSD). Psychol Med. chromatography with electrospray tandem mass spectrometry. J
2016;46:1379‐1390. Chromatogr B Biomed Sci Appl. 2001;765:15‐27.
5. Carhart-Harris RL, Muthukumaraswamy S, Roseman L, et al. Neural 24. Caban M, Migowska N, Stepnowski P, et al. Matrix effects and re-
correlates of the LSD experience revealed by multimodal neuroimag- covery calculations in analyses of pharmaceuticals based on the de-
ing. Proc Natl Acad Sci USA. 2016;113:4853‐4858. termination of β-­blockers and β-­agonists in environmental samples. J
6. Kaelen M, Roseman L, Kahan J, et al. LSD modulates music-­ Chromatogr A. 2012;1258:117‐127.
induced imagery via changes in parahippocampal connectivity. Eur 25. Dams R, Huestis MA, Lambert WE, et  al. Matrix effect in bio-­
Neuropsychopharmacol. 2016;26:1099‐1109. analysis of illicit drugs with LC-­ MS/MS: influence of ionization
7. Lebedev AV, Kaelen M, Lovden M, et al. LSD-­induced entropic brain type, sample preparation, and biofluid. J Am Soc Mass Spectrom.
activity predicts subsequent personality change. Hum Brain Mapp. 2003;14:1290‐1294.
2016;37:3203‐3213. 26. Food and Drug Administration. 2015. Analytical Procedures and Methods
8. Roseman L, Sereno MI, Leech R, et al. LSD alters eyes-­closed func- Validation for Drugs and Biologics: Guidance for Industry. http://www.fda.
tional connectivity within the early visual cortex in a retinotopic fash- gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/
ion. Hum Brain Mapp. 2016;37:3031‐3040. Guidances/UCM386366.pdf. Accessed December 6, 2016.
9. Speth J, Speth C, Kaelen M, et al. Decreased mental time travel to 27. Matuszewski BK, Constanzer ML, Chavez-Eng CM. Strategies for the
the past correlates with default-­mode network disintegration under assessment of matrix effect in quantitative bioanalytical methods
lysergic acid diethylamide. J Psychopharmacol. 2016;30:344‐353. based on HPLC-­MS/MS. Anal Chem. 2003;75:3019‐3030.
10. Tagliazucchi E, Roseman L, Kaelen M, et al. Increased global functional 28. Klette KL, Horn CK, Stout PR, et al. LC-­mS analysis of human urine
connectivity correlates with LSD-­induced ego dissolution. Curr Biol. specimens for 2-­oxo-­3-­hydroxy LSD: method validation for potential
2016;26:1043‐1050. interferants and stability study of 2-­oxo-­3-­hydroxy LSD under various
11. Terhune DB, Luke DP, Kaelen M, et al. A placebo-­controlled investi- storage conditions. J Anal Toxicol. 2002;26:193‐200.
gation of synaesthesia-­like experiences under LSD. Neuropsychologia. 29. Li Z, McNally AJ, Wang H, et al. Stability study of LSD under various
2016;88:28‐34. storage conditions. J Anal Toxicol. 1998;22:520‐525.
12. Dolder PC, Schmid Y, Haschke M, et al. Pharmacokinetics and
concentration-­ effect relationship of oral LSD in humans. Int J
Neuropsychopharmacol. 2015;19:pyv072.
13. Dolder PC, Schmid Y, Mueller F, et al. LSD acutely impairs fear How to cite this article: Dolder PC, Liechti ME, Rentsch KM.
recognition and enhances emotional empathy and sociality. Development and validation of an LC-­MS/MS method to
Neuropsychopharmacology. 2016;41:2638‐2646.
quantify lysergic acid diethylamide (LSD), iso-­LSD, 2-­oxo-­3-­
14. Liechti ME, Dolder PC, Schmid Y. Alterations of conscious-
ness and mystical-­ type experiences after acute LSD in humans. hydroxy-­LSD, and nor-­LSD and identify novel metabolites in
Psychopharmacology. 2017;234:1499‐1510. plasma samples in a controlled clinical trial. J Clin Lab Anal.
15. Strajhar P, Schmid Y, Liakoni E, et  al. Acute effects of lysergic acid 2017;e22265. https://doi.org/10.1002/jcla.22265
diethylamide on circulating steroid levels in healthy subjects. J
Neuroendocrinol. 2016;28:12374.

77
3.7 Publication 7

Pharmacokinetics and Pharmacodynamics of Lysergic Acid


Diethylamide in Healthy Subjects

Patrick C. Dolder1,3, Yasmin Schmid1, Andrea E. Steuer2, Thomas Kraemer2,


Katharina M. Rentsch3, Felix Hammann1, Matthias E. Liechti1

1 Division of Clinical Pharmacology and Toxicology, Department of Biomedicine and Department of


Clinical Research, University Hospital Basel and University of Basel, Switzerland
2 Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine,
University of Zurich, Zurich, Switzerland
3 Laboratory Medicine, University Hospital Basel and University of Basel, Switzerland

78
Clin Pharmacokinet
DOI 10.1007/s40262-017-0513-9

ORIGINAL RESEARCH ARTICLE

Pharmacokinetics and Pharmacodynamics of Lysergic Acid


Diethylamide in Healthy Subjects
Patrick C. Dolder1,3 • Yasmin Schmid1 • Andrea E. Steuer2 • Thomas Kraemer2 •

Katharina M. Rentsch3 • Felix Hammann1 • Matthias E. Liechti1

Ó The Author(s) 2017. This article is published with open access at Springerlink.com

Abstract repeatedly assessed. Pharmacokinetic parameters were


Background and Objective Lysergic acid diethylamide determined using compartmental modeling. Concentration-
(LSD) is used recreationally and in clinical research. The effect relationships were described using pharmacokinetic-
aim of the present study was to characterize the pharma- pharmacodynamic modeling.
cokinetics and exposure–response relationship of oral LSD. Results Geometric mean (95% confidence interval) maxi-
Methods We analyzed pharmacokinetic data from two mum plasma concentration values of 1.3 (1.2–1.9) and 3.1
published placebo-controlled, double-blind, cross-over (2.6–4.0) ng/mL were reached 1.4 and 1.5 h after admin-
studies using oral administration of LSD 100 and 200 lg in istration of 100 and 200 lg LSD, respectively. The plasma
24 and 16 subjects, respectively. The pharmacokinetics of half-life was 2.6 h (2.2–3.4 h). The subjective effects las-
the 100-lg dose is shown for the first time and data for the ted (mean ± standard deviation) 8.2 ± 2.1 and
200-lg dose were reanalyzed and included. Plasma con- 11.6 ± 1.7 h for the 100- and 200-lg LSD doses, respec-
centrations of LSD, subjective effects, and vital signs were tively. Subjective peak effects were reached 2.8 and 2.5 h
after administration of LSD 100 and 200 lg, respectively.
A close relationship was observed between the LSD con-
centration and subjective response within subjects, with
moderate counterclockwise hysteresis. Half-maximal
effective concentration values were in the range of 1 ng/
mL. No correlations were found between plasma LSD
concentrations and the effects of LSD across subjects at or
near maximum plasma concentration and within dose
groups.
Electronic supplementary material The online version of this Conclusions The present pharmacokinetic data are
article (doi:10.1007/s40262-017-0513-9) contains supplementary important for the evaluation of clinical study findings (e.g.,
material, which is available to authorized users. functional magnetic resonance imaging studies) and the
& Matthias E. Liechti
interpretation of LSD intoxication. Oral LSD presented
[email protected] dose-proportional pharmacokinetics and first-order elimi-
nation up to 12 h. The effects of LSD were related to
1
Division of Clinical Pharmacology and Toxicology, changes in plasma concentrations over time, with no evi-
Department of Biomedicine and Department of Clinical
Research, University Hospital Basel, Hebelstrasse 2, 4031
dence of acute tolerance.
Basel, Switzerland Trial registration: NCT02308969, NCT01878942.
2
Department of Forensic Pharmacology and Toxicology,
Zurich Institute of Forensic Medicine, University of Zurich,
Zurich, Switzerland
3
Laboratory Medicine, University Hospital Basel, Basel,
Switzerland

79
P. C. Dolder et al.

second goal was to link the plasma concentration changes


Key Points over time within subjects to the acute subjective and
autonomic effects of LSD to derive half-maximal effective
The pharmacokinetics of lysergic acid diethylamide concentration (EC50) values using standard pharmacoki-
was dose proportional and the subjective effects netic-pharmacodynamic modeling.
were related to the time course of plasma Researchers have correlated subjective drug effects with
concentrations within subjects, with no evidence of brain functional magnetic resonance imaging (fMRI) data
acute tolerance. [12, 13, 16, 17]. This approach likely detects significant
correlations for subjective effects that show large between-
Between-subject differences in plasma subject variance but not for subjective effects of the sub-
concentrations of lysergic acid diethylamide did not stance that are consistently present in all subjects. Plasma
predict the subjective response within a dose group concentrations of LSD have not been determined in any of
and when plasma concentrations were above the the published LSD fMRI studies to date; therefore, it is
half-maximal effective concentration of the response unclear how LSD exposure in the body is linked to sub-
measures. jective effects in these studies. Therefore, a further goal of
the present study was to assess associations across subjects
between plasma exposure to LSD and the pharmacody-
namic effects at corresponding times.
1 Introduction The present study combined data from two similar
clinical studies that tested 100- and 200-lg doses of LSD in
Lysergic acid diethylamide (LSD) is the prototypical hal- 24 and 16 healthy subjects, respectively. The pharma-
lucinogen [1, 2]. Lysergic acid diethylamide has seen cokinetic data and concentration–effect relationship of
worldwide interest with regard to pharmacology, psychia- 100 lg LSD are presented. Similar data on 200 lg LSD
try, and society at large. Lysergic acid diethylamide con- have been previously reported [23]. In the present study,
tinues to be used for recreational and personal purposes [3]. plasma concentrations after 200 lg LSD administration
Additionally, considerable interest has been seen in its were newly measured using a more sensitive and specific
therapeutic potential [4–9], and experimental clinical analytical method. The results were included for compar-
research with LSD has recently been reinitiated [10–23]. isons with the 100-lg data and to newly evaluate
However, basic pharmacokinetic information on LSD is dose/concentration–response effects. The subjective effects
largely missing. A small study in five male subjects of LSD have been reported for both doses, but relationships
reported a mean plasma elimination half-life of LSD of to plasma exposure were not evaluated [21].
175 min after intravenous administration (2 lg/kg) [24].
Another non-systematic study sampled blood after admin-
istration of LSD 160 lg in 13 subjects up to 2.5–5 h but 2 Methods
because of sparse and short sampling could not derive
pharmacokinetic parameters [25]. We recently reported the 2.1 Study Design
first pharmacokinetic data for orally administered LSD
(200 lg) in 16 male and female subjects [23]. The con- We performed the pharmacokinetic data analysis on two
centrations of LSD were maximal after 1.5 h (median) and similar previously performed studies [21–23] using double-
gradually declined to very low levels by 12 h, with an blind, placebo-controlled, cross-over designs with two
elimination half-life of 3.6 h [23]. experimental test sessions (LSD and placebo) in a balanced
Recent studies have reported the effects of LSD on order. Study 1 used a dose of LSD 100 lg and placebo in
various neuronal correlates of brain activation 24 subjects. Study 2 used LSD 200 lg and placebo in 16
[12, 13, 16, 17]. However, plasma exposure and thus the subjects. The washout periods between sessions were at
actual presence of LSD in the body have not been docu- least 7 days. The studies were registered at ClinicalTri-
mented in any of these studies to date. Unknown are the als.gov (NCT02308969, NCT01878942).
time point at which peak concentrations are reached and
the actual or predicted concentrations of LSD at the time 2.2 Participants
point at which pharmacodynamic outcomes were collected.
Therefore, the primary goal of the present study was to Forty healthy participants were recruited from the
describe the pharmacokinetics of a controlled administra- University of Basel campus via an online advertisement.
tion of oral LSD by assessing the plasma concentration- Twenty-four subjects [12 men, 12 women; age
time profile of two doses of LSD (100 and 200 lg). A 33 ± 11 years (mean ± standard deviation); range

80
Pharmacokinetics-Pharmacodynamics of LSD

25–60 years; body weight: 68 ± 8 kg, 55–85 kg) partici- 2.4 Study Drug
pated in Study 1 (100 lg), and 16 subjects (eight men,
eight women; age 29 ± 6 years; range 25–51 years; body Lysergic acid diethylamide (d-lysergic acid diethylamide
weight: 72 ± 12 kg, 52–98 kg) participated in Study 2 hydrate, high-performance liquid chromatography pur-
(200 lg). The inclusion and exclusion criteria were iden- ity [99%; Lipomed AG, Arlesheim, Switzerland) was
tical for both studies. The exclusion criteria were administered in a single oral dose of 100 or 200 lg as a
age \25 years or [65 years, pregnancy (urine pregnancy capsule (Bichsel Laboratories, Interlaken, Switzerland).
test at screening and before each test session), personal or Both doses were within the range of doses that are taken for
family (first-degree relative) history of major psychiatric recreational purposes [1]. The 200-lg dose (the same
disorders (assessed by the semi-structured clinical inter- capsules) was also used in LSD-assisted psychotherapy in
view for Diagnostic and Statistical Manual of Mental patients [6], and intravenous doses of 75–100 lg have been
Disorders, 4th edition, Axis I disorders by the study used in fMRI studies in healthy subjects [13].
physician and an additional interview by a trained psy-
chiatrist), use of medications that may interfere with the 2.5 Measures
study drug, chronic or acute physical illness (abnormal
physical examination, electrocardiogram, or hematological 2.5.1 Blood Sampling
and chemical blood analyses), tobacco smoking (more than
ten cigarettes/day), lifetime prevalence of illicit drug use Blood was collected into lithium heparin tubes before and
more than ten times (except for tetrahydrocannabinol), 0.5, 1, 1.5, 2, 2.5, 3, 4, 6, 8, 10, 12, 16, and 24 h after LSD
illicit drug use within the previous 2 months, and illicit drug administration. The 0.5-, 1.5-, and 2.5-h samples were not
use during the study. We performed urine drug tests at collected in Study 1 (100 lg). The blood samples were
screening and before each test session, and no substances immediately centrifuged, and the plasma was rapidly stored
were detected during the study. The subjects were asked to at -20 °C and later at -80 °C until analysis within
abstain from excessive alcohol consumption between test 12 months. Long-term stability has been shown for LSD
sessions and particularly limit their use to one standard drink when kept under refrigerated or frozen conditions [26, 27].
on the day before the test sessions. Additionally, the par- Samples were thawed for the first time for both analyses,
ticipants were not allowed to drink xanthine-containing this was also the case for study 2 (200 lg) because separate
liquids after midnight before the study day. The participants sets of samples were stored and used for the present [28]
did not regularly use medications that could potentially and previous [29] analyses.
interact with the study drug. No other medications aside
from LSD were used during the study sessions. Eleven 2.5.2 Analysis of Lysergic Acid Diethylamide
subjects had previously used a hallucinogen, including LSD Concentrations
(six participants), one to three times during their lives, and
most of the subjects (29) were hallucinogen naive. Lysergic acid diethylamide concentrations in plasma were
determined using sensitive and validated liquid chro-
2.3 Study Procedures matography-tandem mass spectrometry methods as repor-
ted in detail elsewhere [28, 29]. The lower limit of
Each study included a screening visit, a psychiatric inter- quantification was 0.05 ng/mL in Study 1 (100 lg) [29]
view, two 25-h experimental sessions, and an end-of-study and 0.01 ng/mL in Study 2 (200 lg) [28].
visit. The experimental sessions were conducted in a quiet
standard hospital patient room. The participants were resting 2.5.3 Subjective Mood
in hospital beds except when going to the restroom. Only one
research subject and one investigator were present during Visual analog scales (VASs) were repeatedly used to assess
the experimental sessions. The participants could interact subjective effects over time [21, 22]. The VASs included
with the investigator, rest quietly, and/or listen to music via separate measures for ‘‘any drug effect,’’ ‘‘good drug
headphones, but no other entertainment was provided. LSD effect,’’ and ‘‘bad drug effect’’ and were presented as
or placebo was administered at 9:00 A.M. A standardized 100-mm horizontal lines (0–100%) marked from ‘‘not at
lunch and dinner was served at 1:30 P.M. and 5.30 P.M., all’’ on the left to ‘‘extremely’’ on the right. The VASs
respectively. The subjects were never alone during the first were administered 1 h before and 0, 0.5, 1, 1.5, 2, 2.5, 3, 4,
12 h after drug administration, and the investigator was in a 5, 6, 7, 8, 9, 10, 11, 12, 16, and 24 h after drug adminis-
room next to the subject for up to 24 h while the subject was tration. The 0.5- and 2.5-h ratings were not collected in
asleep (mostly from 1:00 A.M. to 8:00 A.M.). Study 1 (100 lg).

81
P. C. Dolder et al.

2.5.4 Vital Signs estimated effect-site concentrations and the effects of LSD
compared with a simple Emax model (plot inspection and
Blood pressure, heart rate, and body temperature were Akaike information criteria). Examples of diagnostic plots
assessed repeatedly 1 h before and 0, 0.5, 1, 1.5, 2, 3, 4, 5, are shown in Figs. S8 and S9.
6, 7, 8, 9, 10, 11, 12, and 24 h after drug administration.
Diastolic and systolic blood pressure and heart rate were 2.7 Statistical Analyses
measured using an automatic oscillometric device
(OMRON Healthcare Europe NA, Hoofddorp, Nether- The LSD-induced subjective and autonomic effects were
lands). The measurements were performed in duplicate at determined as a difference from placebo in the same sub-
an interval of 1 min and after a resting time of at least ject at the corresponding time point to control for circadian
10 min. The averages were calculated for analysis. Core changes and placebo effects [22]. The pharmacodynamic
(tympanic) temperature was measured using a GENIUSTM effect changes after LSD administration for each time point
2 ear thermometer (Tyco Healthcare Group LP, Water- were plotted over time (effect-time curves) and against the
town, NY, USA). The 0.5- and 2.5-h measures were not respective plasma concentrations of LSD and graphed as
collected in Study 1 (100 lg). concentration-effect curves. The onset, time to maximum
plasma concentration (Tmax), offset, and effect duration
2.6 Pharmacokinetic Analyses were assessed for the model-predicted ‘‘any drug effect’’
and Pharmacokinetic-Pharmacodynamic VAS effect-time plots after LSD using a threshold of 10%
Modeling of the maximal possible effect of 100% using Phoenix
WinNonlin 6.4. Associations between concentrations and
All of the analyses were performed using Phoenix effects were assessed using Pearson correlations, and
WinNonlin 6.4 (Certara, Princeton, NJ, USA). Pharma- multiple regression analysis was used to exclude effects of
cokinetic parameters were estimated using compartmental sex and body weight (Statistica 12 software; StatSoft,
modeling. A one-compartment model was used with first- Tulsa, OK, USA).
order input, first-order elimination, and no lag time. Initial
estimates for apparent volume of distribution and k were
derived from non-compartmental analyses. 3 Results
The model fit was not relevantly improved by a two-
compartment model based on visual inspection of the plots. 3.1 Pharmacokinetics
The one-compartment model showed better Akaike infor-
mation criterion values in all subjects than a two-com- The plasma concentration-time curves for the two LSD
partment model. The pharmacokinetic model was first doses are shown in Fig. 1a. The pharmacokinetic parame-
fitted and evaluated. The predicted concentrations were ters are shown in Table 1. In Study 1 (100 lg), LSD could
then used as inputs to the pharmacodynamic model, treat- be quantified up to 8, 10, 12, 16, and 24 h in 24, 23, 22, 9,
ing the pharmacokinetic parameters as fixed and using the and one subject, respectively. In Study 2 (200 lg), LSD
classic pharmacokinetic/pharmacodynamic link model could be quantified up to 16 h in all 16 subjects and up to
module in WinNonlin. The model uses a first-order equi-
librium rate constant (keo) that related the observed phar- Fig. 1 Pharmacokinetics and pharmacodynamics of lysergic acid c
diethylamide (LSD). a LSD plasma concentration-time curves. The
macodynamic effects of LSD to the estimated LSD corresponding semi-log plot is shown in Fig. S3. LSD effect-time
concentrations at the effect site (Fig. S1) and accounts for curves for Visual Analog Scale ratings (0–100%) of b ‘‘any drug
the lag between the plasma- and effect-site concentration effect,’’ d ‘‘good drug effect,’’ and f ‘‘bad drug effect.’’ c, e, g In the
curves [30]. Initial estimates for keo values were obtained LSD concentration-effect plots (hysteresis curves), the subjective
effects of LSD showed moderate counterclockwise hysteresis,
using semi-compartmental modeling by collapsing the indicating a relatively short delay in the effect of LSD relative to
hysteresis loop in the Ce vs. effect plots in WinNonlin. A the changes in plasma concentration over time. The plasma concen-
sigmoid maximum effect (Emax) model (EC50, Emax, c) was tration-effect site equilibration half-lives were in the range of
selected for all pharmacodynamic effects. EC50 and Emax 21–48 min according to the pharmacokinetic-pharmacodynamic link
model (Table 2). ‘‘Any drug effect’’ and ‘‘good drug effect’’ were
estimates were taken from the pharmacokinetic-pharma- robustly and markedly increased in all subjects and paralleled the
codynamic plots. Lower and upper limits for Emax were set changes in LSD concentration, whereas the mean ‘‘bad drug effect’’
to 0 and 100%, respectively, for all the VAS scores. Upper increased only moderately after LSD owing to transient increases.
limits for Emax for changes in heart rate, body temperature, ‘‘Bad drug effect’’ occurred mostly at the onset of the drug effect in
some subjects but also later in time in others. The data are expressed
and diastolic and systolic blood pressure were set to as the mean ± standard error of the mean in 24 and 16 subjects after
100/min, 2 °C, 50 and 80 mm Hg, respectively. The sig- administration of 100 and 200 lg LSD, respectively. The time of
moidal Emax model best described the relationship between sampling is noted next to each point. LSD was administered at t = 0

82
Pharmacokinetics-Pharmacodynamics of LSD

83
P. C. Dolder et al.

Table 1 Pharmacokinetic parameters for LSD based on compartmental modeling


Dose N k01 (1/h) k (1/h) Vd (L) Cmax (ng/ tmax (h) t1/2 (h) AUC? CL/F (L/h)
mL) (ngh/mL)

100 lg 24 Geometric mean 1.4 0.27 46 1.3 1.4 2.6 8.1 12.3
(95% CI) (1.2–4.1) (0.24–0.31) (35–76) (1.2–1.9) (1.3–2.1) (2.4–3.0) (7.5–11.1) (7.8–24)
Range 0.31–9.9 0.17–0.50 24–270 0.3–3.7 0.4–3.2 1.4–4.2 1–19 5.2–103
200 lg 16 Geometric mean 1.2 0.27 37 3.1 1.5 2.6 20.3 9.9
(95% CI) (0.68–4.6) (0.22–0.35) (32–46) (2.6–4.0) (1.3–2.4) (2.2–3.4) (17.3–26.2) (8.3–12.8)
Range 0.27–10 0.12–0.59 18–66 1.9–7.1 0.4–3.8 1.2–5.6 11–39 5.1–18.5
AUC? area under the plasma concentration-time curve from time zero to infinity, Cmax estimated maximum plasma concentration, t1/2 estimated
plasma elimination half-life, tmax estimated time to reach Cmax, k01 first-order absorption coefficient, k first order elimination coefficient, Vd
volume of distribution

24 h in 15 subjects (Fig. S2). Mean maximum plasma 2.5 ± 1.2 h (range 0.8–4.4 h). LSD increased diastolic and
concentration (Cmax) and area under the concentration-time systolic blood pressure, heart rate, and body temperature
curve values were approximately twice as high for the compared with placebo to similar extents for both doses
200-lg dose compared with the 100-lg dose. Dose-nor- (Fig. 2). The corresponding peak effect data and dose-re-
malized Cmax and area under the concentration-time curve sponse statistics have been previously reported [21].
values were not statistically different between the dose
groups and the Tmax and plasma half-lives were also sim- 3.3 Pharmacokinetic-Pharmacodynamic Modeling
ilar, consistent with dose-proportional pharmacokinetics
(Table 1). Consistent with the fit of the one-compartment Figures 1 and 2 show the subjective, cardiovascular, and
model, inspection of the semi-logarithmic concentration- thermogenic effects of LSD plotted against the plasma
time curves showed linear elimination kinetics for both concentration over time. A close relationship was found
doses (Fig. S3) up to 12 h as previously reported for the between LSD concentrations and LSD effects over time.
200-lg dose [23]. The individual-observed and model- Counterclockwise hysteresis was observed during the
predicted LSD concentrations are shown in Fig. S2. Plasma assumed drug distribution phase (\2 h), especially for
concentrations varied considerably between subjects, body temperature (Fig. 2h). Model-predicted effects of
especially at the lower 100-lg dose (Table 1; Fig. S2). LSD on the VASs for ‘‘any drug effect,’’ ‘‘good drug
effect,’’ and ‘‘bad drug effect’’ are illustrated for each
3.2 Pharmacodynamics subject in Figs. S4–6, respectively. Table 2 shows the
predicted concentrations of LSD at the effect site that
Lysergic acid diethylamide produced robust increases in produced half-maximal effects (EC50 values). Mean EC50
‘‘any drug effect’’ (Fig. 1b, Fig. S4) and ‘‘good drug
effect’’ (Fig. 1d, Fig. S5). Transient ‘‘bad drug effect’’ was Fig. 2 Pharmacokinetics and autonomic effects in response to c
reported in some subjects, resulting in a moderate increase lysergic acid diethylamide (LSD). The figure shows LSD effect-time
in mean group ratings (Fig. 1f, Fig. S6). The corresponding curves for a diastolic blood pressure, c systolic blood pressure, e heart
subjective peak effects have previously been reported and rate, and g changes in body temperature and corresponding b, d, f,
h LSD concentration-effect plots (hysteresis curves). The cardiovas-
were shown to be dose dependent [21]. ‘‘Any drug effect,’’ cular stimulant effects of LSD at the higher 200-lg dose showed only
‘‘good drug effect,’’ and ‘‘bad drug effect’’ ratings for each little counterclockwise hysteresis, indicating a short delay in the
subject are shown in Figs. S4–6, respectively. After effect of LSD relative to the changes in plasma concentration over
administration of the 100-lg dose of LSD, the times of time and thus a close relationship between LSD concentration and
changes in cardiovascular effects over time within subjects. The
onset and offset of the subjective response, assessed by the plasma concentration-effect site equilibration half-lives were in the
‘‘any drug effect’’ VAS, were (mean ± standard deviation) range of 13–34 min according to the pharmacokinetic-pharmacody-
0.8 ± 0.4 h (range 0.1–1.7 h) and 9.0 ± 2.0 h (range namic link model (Table 2). In contrast, marked counterclockwise
6.1–14.5 h), respectively. The mean effect duration was hysteresis was observed in the LSD concentration-body temperature
change plot, indicating that the LSD-induced changes in body
8.2 ± 2.1 h (range 5–14 h). The time to peak drug effect temperature manifested themselves slowly and with a mean plasma
was 2.8 ± 0.8 h (range 1.2–4.6 h). After administration of concentration-effect site equilibration half-life of 136 min for the
the 200-lg dose of LSD, the times of onset and offset of 200-lg dose (Table 2). The data are expressed as the mean ± stan-
the subjective response were 0.4 ± 0.3 h (range dard error of the meant in 24 and 16 subjects after administration of
LSD 100 and 200 lg, respectively. The pharmacodynamic values are
0.04–1.2 h) and 11.6 ± 4.2 h (range 7.0–19.5 h), respec- the mean ± standard error of the mean differences from placebo at
tively. The mean effect duration was 11.2 ± 4.2 h (range each time point. The time of sampling is noted next to each point.
6.4–19.3 h). The time to the subjective peak response was LSD was administered at t = 0

84
Pharmacokinetics-Pharmacodynamics of LSD

85
P. C. Dolder et al.

Table 2 Pharmacodynamic parameter estimates (PK-PD link model)


Effect Dose EC50 (ng/mL) Emax (%) c keo (1/h) T1/2keo (min)

Any drug effect 100 lg 0.75 ± 0.4 95 ± 9 4.2 ± 1.2 1.8 ± 1.3 35 ± 23
200 lg 1.2 ± 0.7 97 ± 5 3.4 ± 1.5 3.4 ± 1.7 21 ± 17
Good drug effect 100 lg 0.71 ± 0.5 89 ± 15 3.9 ± 1.5 2.0 ± 1.6 39 ± 37
200 lg 0.94 ± 0.5 93 ± 9 3.2 ± 1.6 2.4 ± 1.8 32 ± 29
Bad drug effect 100 lg 1.5 ± 1.1 32 ± 37 4.7 ± 2.3 2.7 ± 2.2 42 ± 37
200 lg 2.5 ± 1.6 34 ± 35 3.2 ± 2.1 2.8 ± 2.0 48 ± 66
Heart rate increase 100 lg 0.67 ± 0.5 22 ± 25 3.7 ± 2.0 2.5 ± 1.9 46 ± 52
200 lg 1.9 ± 1.2 33 ± 28 2.7 ± 1.8 4.0 ± 2.0 13 ± 8
Body temperature increase 100 lg 0.75 ± 0.4 1.1 ± 0.6 2.2 ± 1.8 1.5 ± 1.6 107 ± 121
200 lg 1.8 ± 1.1 1.0 ± 0.6 3.6 ± 2.0 1.7 ± 1.9 136 ± 155
Diastolic blood pressure increase 100 lg 0.9 ± 0.6 23 ± 14 2.0 ± 1.6 2.6 ± 1.9 53 ± 70
200 lg 1.6 ± 0.9 18 ± 11 3.5 ± 1.6 3.4 ± 1.9 31 ± 42
Systolic blood pressure increase 100 lg 0.8 ± 0.5 30 ± 17 1.9 ± 1.6 2.6 ± 1.7 51 ± 78
200 lg 1.9 ± 1.4 30 ± 17 2.9 ± 1.9 3.2 ± 1.9 34 ± 41
Values are means ± standard deviations. T1/2keo = ln2/keo, calculated for each individual value
EC50 maximal effect predicted by the PK-PD link model, EC50 predicted drug concentration at effect site producing a half-maximal effect, c
sigmoid shape parameter, keo first-order rate constant for the equilibration process between plasma concentration and effect site (PK-PD model
link parameter), t1/2keo (min) plasma-effect-site equilibration half-life

values were in the range of 0.67–2.5 ng/mL and lower for effects of LSD and any of these possible predictors. Thus,
‘‘good drug effect’’ than for ‘‘bad drug effect’’ (Table 2). the plasma concentrations of LSD did not predict the
‘‘Any drug effect’’ and ‘‘good drug effect’’ could be effects of LSD during the time it produced robust and
modeled in all of the subjects, whereas no ‘‘bad drug similar effects in all of the subjects (i.e., little between-
effect’’ (ratings \5% at any time point) was reported in subject variability). In contrast, a close relationship was
eight (33%) and five (31%) subjects after 100 and 200 lg, found over time within subjects, as shown in the pharma-
respectively. Thus, the EC50 and keo values could not be cokinetic-pharmacodynamic analysis (Figs. 1, 2).
determined in these subjects. Similarly, vital signs did not
change sufficiently in a few subjects (one to three/outcome)
to determine these values. 4 Discussion
The predicted Cmax of LSD did not correlate with the
predicted maximal response on the ‘‘any drug effect’’ VAS The present study describes the pharmacokinetics and
when analyzed across subjects and separately for the two concentration–effect relationship after oral administration
dose groups (Rp = 0.38, p = 0.08, and Rp = 0, p = 0.9, of LSD 100 lg. Additionally, the previously reported
for the 100- and 200-lg doses, respectively). There was a pharmacokinetics and concentration–effect relationship for
significant correlation in the pooled sample (Rp = 0.38, the 200-lg dose of LSD [23] were reanalyzed and included
p \ 0.05, n = 40, Fig. S7). The predicted area under the for comparison with the 100-lg dose. Compartmental
concentration-time curve of LSD did not correlate with the modeling predicted geometric mean peak plasma concen-
predicted area under the concentration-time curve for ‘‘any trations of 1.3 ng/mL, 1.4 h after administration of the
drug effect’’, a measure of the overall pharmacodynamic 100-lg dose. Mean Cmax values of 3.1 ng/mL were reached
response (Rp = 0, p = 0.9, and Rp = 0.27, p = 0.4, after 1.5 h after administration of the 200-lg dose. The
respectively). Additionally, there were generally no corre- predicted mean half-lives of LSD were 2.6 h after both
lations between plasma LSD concentrations and different doses. The plasma half-life in the present study was com-
pharmacodynamic effects for matched time points across parable to the value of 2.9 h after intravenous administra-
subjects within dose groups (Table 3). A few correlations tion of 2 lg/kg of LSD [24] but shorter than the 3.6-h value
were significant at the beginning (1 h) and end (8 and 12 h) previously determined using non-compartmental analysis
of the LSD effect. However, no significant associations [23]. Additionally, the plasma concentrations after admin-
were found between plasma concentrations and effects istration of the 200-lg dose in the present study were lower
during the peak response to LSD (3–6 h). Multiple than those that were previously published in the same
regression analysis, including LSD concentration, body research subjects [23]. This can be explained by the dif-
weight, and sex, revealed no associations between the ferent analytical methods and modeling approach that were

86
Pharmacokinetics-Pharmacodynamics of LSD

Table 3 Correlations between plasma levels of LSD and its pharmacodynamic effects at the corresponding time points after administration
Effect 1h 2h 3h 4h 6h 8h 10 h 12 h

Any subjective drug effect 100 lg N = 24 0.17 0.13 -0.02 -0.04 -0.18 0.09 0.01 -0.03
200 lg N = 16 0.21 0.17 0.1 0.13 0.2 0.16 0.33 0.42
Both N = 40 0.36 0.35 0.19 0.04 0.06 0.41 0.46 0.49
Good drug effect 100 lg N = 24 0.6 0.3 0.23 0.15 -0.13 -0.2 -0.03 0.04
200 lg N = 16 0 -0.23 0.32 0.27 0.28 0.55 0.39 0.17
Both N = 40 0.39 0.34 0.36 0.31 0.24 0.42 0.35 0.23
Bad drug effect 100 lg N = 24 0.06 -0.11 -0.23 -0.1 -0.08 -0.03 0 -0.15
200 lg N = 16 0.34 -0.32 -0.27 0.07 0.2 0.35 -0.26 -0.16
Both N = 40 0.36 -0.16 0 0 0.1 0.29 0.05 0.07
Heart rate increase 100 lg N = 24 0.41 0.3 0.4 0.27 0.1 0.26 -0.4 0.027
200 lg N = 16 0.3 0.21 0.3 -0.06 -0.08 0.19 -0.16 -0.52
Both N = 40 0.44 0.41 0.33 0.08 -0.05 -0.02 0.03 -0.2
Body temperature increase 100 lg N = 24 0.12 -0.27 0.14 0.07 0.18 -0.06 -0.2 0.41
200 lg N = 16 0.09 -0.11 0.54 -0.1 -0.02 0.37 0.15 -0.19
Both N = 40 -0.08 -0.18 0.25 -0.15 -0.09 -0.12 0.02 0.06
Diastolic blood pressure increase 100 lg N = 24 0.16 -0.09 0.14 0.04 0.17 0.15 0.28 0.13
200 lg N = 16 -0.53 -0.22 0.2 -0.13 0.09 0.27 0.09 0.47
Both N = 40 -0.2 -0.03 0.07 0.03 -0.06 -0.01 0.01 0.07
Systolic blood pressure increase 100 lg N = 24 0.1 0.05 0.06 0 0.2 0.23 0.29 0.21
200 lg N = 16 -0.03 -0.4 -0.1 0.25 0 0.54 -0.02 0.19
Both N = 40 0 0.07 0.03 0.07 -0.07 0.11 0.05 0.08
Data are Pearson correlation coefficients between the LSD concentration in plasma and the corresponding time-matched effect of LSD. Bold
values indicate significant associations (p \ 0.05)

used in the present study, which predicts lower Cmax values and route of administration [10–19]. The intravenous 75-lg
than the observed values. Overall, we observed linear dose dose of LSD produced comparably strong alterations in
and elimination kinetics of LSD up to 12 h after drug consciousness to the 100-lg dose in the present study
administration. [10, 31]. Additionally, the time-concentration curve for the
The present data on the plasma concentration-time 75-lg intravenous preparation remains unknown. Specifi-
curves of LSD are important because many experimental cally, an intravenous bolus dose of LSD would be expected
and therapeutic studies are currently being conducted or to result in peak effects shortly after administration.
have been published without this detailed information on Indeed, early studies reported that intravenous adminis-
the presence of LSD in the human body. Specifically, the tration of LSD tartrate salt at a higher dose (2 lg/kg of
effects of LSD on emotion processing after 100 and 200 lg base) produced a rapid onset within seconds to minutes and
have been reported [23], but no pharmacokinetic data were peak effects that occurred approximately 30 min after
reported. Additionally, fMRI data were obtained in Study 1 administration [24, 32–34].
(100 lg) in Basel and in an additional study in Zurich In the more recent studies that used the 75-lg dose
(n = 22) that did not perform blood sampling. Doses of administered as the base, subjective drug effects reportedly
100 lg were used in both studies. Thus, the present study began within 5–15 min and peaked 45–90 min after
provides estimates of LSD concentrations in plasma over intravenous dosing, although further details were not
time for these studies and the observed and predicted time reported [13, 19]. Other hallucinogens with mechanisms of
courses of the subjective and autonomic effects of LSD. action that are similar to those of LSD (e.g., serotonin
The 200-lg dose preparation of LSD has been used in 5-HT2A receptor stimulation [35]), such as dimetyl-
patients [5, 6], and the present phase I study provides the tryptamine or psilocybin, also produced subjective and
pharmacokinetic data for these phase II studies. autonomic effects almost instantaneously and peak effects
In contrast, no data are currently available on the plasma within 2–5 min after intravenous administration [36–38].
concentrations of LSD after intravenous administration of In the present study, the mean effect onset and peak were
75 lg of LSD base in saline [11], despite the publication of 48 and 170 min, respectively, after oral administration of
extensive pharmacodynamic data using this preparation LSD 100 lg. Thus, the effect began and peaked an average

87
P. C. Dolder et al.

of 30 and 100 min later, respectively, after oral adminis- comparable for the increases in heart rate and blood pres-
tration compared with intravenous administration of an sure but longer for the thermogenic response. No clockwise
equivalent dose [13, 19]. Magnetic resonance imaging hysteresis was found for any of the pharmacodynamic
scanning correctly started at approximately 70 and 150 min outcome measures, and thus no evidence was found of
in the studies that used intravenous [13] and oral (unpub- acute tolerance as described for other psychoactive sub-
lished data from Study 1, 100 lg) routes of LSD admin- stances, such as methylenedioxymethamphetamine [39] or
istration, respectively, coinciding with the maximal cocaine [40], or for repeated administration of LSD [41].
response to LSD. Nevertheless, the plasma concentrations Thus, as long as relevant concentrations of LSD were
of LSD and associated time-matched subjective responses present in plasma, subjective and autonomic effects were
after intravenous LSD administration should also be observed. The mean durations of the subjective effects of
determined to better evaluate the considerable research LSD was 8 and 11 h after administration of the 100- and
data that have been generated with this formulation. 200-lg doses, respectively, and the difference corre-
After intravenous administration, a drug is rapidly sponded to the plasma half-life of LSD.
diluted and distributed within the blood. Peak plasma The present analyses typically found no correlations
concentrations are typically reached rapidly, and elimina- between LSD concentrations and the effects of LSD across
tion begins immediately. Using the model parameters k and subjects within dose groups, likely because of the relatively
keo from the present study, the Tmax for ‘‘any drug effect’’ high concentrations of LSD and generally consistently high
after intravenous administration can be predicted to occur subjective response ratings in most subjects. If relatively
at approximately 70 and 50 min for the 100- and 200-lg high and similar doses of LSD are used that result in plasma
doses and are thus similar to the recently observed times to concentrations above the EC50 of a particular response
peak effects [13, 19]. In our model, the relatively long Tmax measures, then responses do not vary across subjects
of the effect of LSD is represented by the lag that is because responses are close to maximal. This would typi-
attributable to distribution of the drug from plasma to the cally also be the case with measures with a maximal effect
hypothetical effect compartment. The cause for this lag is limit such as VAS ratings and some physiological effects
unclear. Additional studies are needed to determine whe- such as pupil size [42]. In fact, responses to LSD or other
ther LSD is distributed slowly because it is present only in drugs in a standardized experimental setting may vary only
small concentrations or slowly penetrates the blood–brain if the response is not induced consistently in all subjects
barrier or whether there is a lag in the response mechanism. (e.g., at the beginning and end of the response) because of
The present study showed that LSD produced robust and individual differences in drug absorption/distribution and
high subjective ‘‘any drug effect’’ and ‘‘good drug effect’’ elimination. Correlations of plasma concentrations with the
in almost all of the subjects. The estimates of the corre- subjective and cardiovascular effects of LSD or 3,4-
sponding EC50 values were in the range of 0.71–1.2 ng/mL methylenedioxymethamphetamine [42] across subjects are
and lower than the mean LSD Cmax values (1.3 and 3.1 ng/ only weak during the peak response. This finding needs to
mL for the 100- and 200-lg doses, respectively) observed be considered when interpreting associations between sub-
in the present study. ‘‘Bad drug effects’’ were moderate and jective responses and other measures, such as fMRI
not present in every subject. Consistent with this finding, parameters. fMRI findings may reflect the variance in LSD
the EC50 values were higher than those for ‘‘good drug plasma concentrations. The likelihood of detecting corre-
effect’’ and ‘‘any drug effect’’ (1.5–2.5 ng/mL). As previ- lations within a dose group increases for effects that are not
ously reported, the subjective effects were dose dependent, robustly induced in all subjects.
whereas the autonomic effects were comparable at both The present study has limitations. First, the two doses of
doses [21]. When analyzed within subjects using pharma- LSD were evaluated in two separate studies in different
cokinetic-pharmacodynamic modeling, a close relationship participants and not within subjects. Second, the plasma
was found between plasma concentrations of LSD and the samples were analyzed in different laboratories. Nonethe-
effects of LSD, with moderate counterclockwise hysteresis. less, the pharmacokinetic data were consistent across the
Counterclockwise hysteresis typically reflects the time lag two studies and laboratories.
that is caused by drug distribution to the effect site and the
response time associated with the mechanism of action.
The present study showed that the subjective and auto- 5 Conclusion
nomic effects establish themselves relatively slowly. On
average, the subjective ‘‘any drug effect’’ peak was reached We gathered pharmacokinetic data for oral LSD that are
2.8 and 2.5 h after administration of the 100- and 200-lg essential for interpreting the findings of clinical studies and
doses, respectively, and 1.1 and 0.6 h after the respective LSD intoxication. LSD had dose-proportional pharma-
peak LSD concentrations were reached. The lag times were cokinetics and first-order elimination up to 12 h. A close

88
Pharmacokinetics-Pharmacodynamics of LSD

plasma concentration–effect relationship was found within associated with life-threatening diseases. J Nerv Ment Dis.
subjects over time, with moderate counterclockwise hys- 2014;202(7):513–20.
7. Baumeister D, Barnes G, Giaroli G, Tracy D. Classical hallu-
teresis because of a short lag of the response. Generally, no cinogens as antidepressants? A review of pharmacodynamics and
association was found between plasma LSD concentrations putative clinical roles. Ther Adv Psychopharmacol.
and its robust effects when analyzed across different sub- 2014;4(4):156–69.
jects and within a dose group. This has implications for 8. Krebs TS, Johansen PO. Lysergic acid diethylamide (LSD) for
alcoholism: meta-analysis of randomized controlled trials. J Psy-
studies that interrelate different effects of LSD. chopharmacol. 2012;26(7):994–1002.
9. Kupferschmidt K. High hopes. Science. 2014;345(6192):18–23.
Acknowledgements The authors acknowledge the proofreading 10. Carhart-Harris RL, Kaelen M, Bolstridge M, et al. The para-
assistance of Michael Arends, a professional freelance English editor, doxical psychological effects of lysergic acid diethylamide
the services of whom were paid for by the authors. (LSD). Psychol Med. 2016;46:1379–90.
11. Tagliazucchi E, Roseman L, Kaelen M, et al. Increased global
Author contributions PD designed the research, performed the functional connectivity correlates with LSD-induced ego disso-
research, and analyzed the data. YS designed the research and per- lution. Curr Biol. 2016;26(8):1043–50.
formed the research. AES performed the research and analyzed the 12. Kaelen M, Roseman L, Kahan J, et al. LSD modulates music-
data. TK, FH and KMR analyzed the data. MEL designed the induced imagery via changes in parahippocampal connectivity.
research, analyzed the data, and wrote the manuscript. Eur Neuropsychopharmacol. 2016;26:1099–109.
13. Carhart-Harris RL, Muthukumaraswamy S, Roseman L, et al.
Neural correlates of the LSD experience revealed by multimodal
Compliance with Ethical Standards neuroimaging. Proc Natl Acad Sci. 2016;113:4853–8.
14. Terhune DB, Luke DP, Kaelen M, et al. A placebo-controlled
Funding This work was supported by the Swiss National Science investigation of synaesthesia-like experiences under LSD. Neu-
Foundation (Grant No. 320030_170249 to ML) and the University ropsychologia. 2016;88:28–34.
Hospital Basel. 15. Speth J, Speth C, Kaelen M, et al. Decreased mental time travel to
the past correlates with default-mode network disintegration
Conflict of interest Patrick C. Dolder, Yasmin Schmid, Andrea E. under lysergic acid diethylamide. J Psychopharmacol.
Steuer, Thomas Kraemer, Katharina M. Rentsch, Felix Hammann, 2016;30(4):344–53.
and Matthias E. Liechti declare no conflicts of interest. 16. Roseman L, Sereno MI, Leech R, et al. LSD alters eyes-closed
functional connectivity within the early visual cortex in a
Ethics approval and consent to participate The studies were con- retinotopic fashion. Hum Brain Mapp. 2016;37:3031–40.
ducted in accordance with the Declaration of Helsinki and approved 17. Lebedev AV, Kaelen M, Lovden M, et al. LSD-induced entropic
by the local ethics committee. The administration of LSD to healthy brain activity predicts subsequent personality change. Hum Brain
subjects was authorized by the Swiss Federal Office for Public Health, Mapp. 2016;37:3203–13.
Bern, Switzerland. All of the subjects provided written consent before 18. Carhart-Harris RL, Kaelen M, Whalley MG, et al. LSD enhances
participating in either of the studies, and they were paid for their suggestibility in healthy volunteers. Psychopharmacology.
participation. 2015;232(4):785–94.
19. Kaelen M, Barrett FS, Roseman L, et al. LSD enhances the
Open Access This article is distributed under the terms of the emotional response to music. Psychopharmacology.
Creative Commons Attribution-NonCommercial 4.0 International 2015;232(19):3607–14.
License (http://creativecommons.org/licenses/by-nc/4.0/), which per- 20. Strajhar P, Schmid Y, Liakoni E, et al. Acute effects of lysergic
mits any noncommercial use, distribution, and reproduction in any acid diethylamide on circulating steroid levels in healthy subjects.
medium, provided you give appropriate credit to the original J Neuroendocrinol. 2016;28:12374.
author(s) and the source, provide a link to the Creative Commons 21. Dolder PC, Schmid Y, Mueller F, et al. LSD acutely impairs fear
license, and indicate if changes were made. recognition and enhances emotional empathy and sociality.
Neuropsychopharmacology. 2016;41:2638–46.
22. Schmid Y, Enzler F, Gasser P, et al. Acute effects of lysergic acid
diethylamide in healthy subjects. Biol Psychiatry.
2015;78(8):544–53.
References 23. Dolder PC, Schmid Y, Haschke M, et al. Pharmacokinetics and
concentration-effect relationship of oral LSD in humans. Int J
1. Passie T, Halpern JH, Stichtenoth DO, et al. The pharmacology of Neuropsychopharmacol. 2015;19:pii: pyv072.
lysergic acid diethylamide: a review. CNS Neurosci Ther. 24. Aghajanian GK, Bing OH. Persistence of lysergic acid diethy-
2008;14(4):295–314. lamide in the plasma of human subjects. Clin Pharmacol Ther.
2. Nichols DE. Psychedelics. Pharmacol Rev. 2016;68(2):264–355. 1964;5:611–4.
3. Krebs TS, Johansen PO. Over 30 million psychedelic users in the 25. Upshall DG, Wailling DG. The determination of LSD in human
United States. F1000Res. 2013;2:98. plasma following oral administration. Clin Chim Acta.
4. Davenport WJ. Psychedelic and nonpsychedelic LSD and psilo- 1972;36(1):67–73.
cybin for cluster headache. CMAJ. 2016;188(3):217. 26. Klette KL, Horn CK, Stout PR, Anderson CJ. LC-MS analysis of
5. Gasser P, Kirchner K, Passie T. LSD-assisted psychotherapy for human urine specimens for 2-oxo-3-hydroxy LSD: method vali-
anxiety associated with a life-threatening disease: a qualitative dation for potential interferants and stability study of 2-oxo-3-
study of acute and sustained subjective effects. J Psychopharma- hydroxy LSD under various storage conditions. J Anal Toxicol.
col. 2015;29(1):57–68. 2002;26(4):193–200.
6. Gasser P, Holstein D, Michel Y, et al. Safety and efficacy of 27. Martin R, Schurenkamp J, Gasse A, et al. Determination of psi-
lysergic acid diethylamide-assisted psychotherapy for anxiety locin, bufotenine, LSD and its metabolites in serum, plasma and

89
P. C. Dolder et al.

urine by SPE-LC-MS/MS. Int J Legal Med. with classic hallucinogens. Eur Neuropsychopharmacol.
2013;127(3):593–601. 2016;26:1327–37.
28. Steuer AE, Poetzsch M, Stock L, et al. Development and vali- 36. Gallimore AR, Strassman RJ. A model for the application of
dation of an ultra-fast and sensitive microflow liquid chro- target-controlled intravenous infusion for prolonged immersive
matography-tandem mass spectrometry (MFLC-MS/MS) method DMT psychedelic experience. Front Pharmacol. 2016;14(7):211.
for quantification of LSD and its metabolites in plasma and doi:10.3389/fphar.2016.00211.
application to a controlled LSD administration study in humans. 37. Strassman RJ, Qualls CR, Uhlenhuth EH, Kellner R. Dose-re-
Drug Test Anal. 2016. doi:10.1002/dta.2042 (Epub ahead of sponse study of N, N-dimethyltryptamine in humans: II. Subjec-
print). tive effects and preliminary results of a new rating scale. Arch
29. Dolder PC, Liechti ME, Rentsch KM. Development and valida- Gen Psychiatry. 1994;51(2):98–108.
tion of a rapid turboflow LC-MS/MS method for the quantifica- 38. Carhart-Harris RL, Williams TM, Sessa B, et al. The adminis-
tion of LSD and 2-oxo-3-hydroxy LSD in serum and urine tration of psilocybin to healthy, hallucinogen-experienced vol-
samples of emergency toxicological cases. Anal Bioanal Chem. unteers in a mock-functional magnetic resonance imaging
2015;407:1577–84. environment: a preliminary investigation of tolerability. J Psy-
30. Sheiner LB, Stanski DR, Vozeh S, et al. Simultaneous modeling chopharmacol. 2011;25(11):1562–7.
of pharmacokinetics and pharmacodynamics: application to 39. Hysek CM, Simmler LD, Ineichen M, et al. The norepinephrine
d-tubocurarine. Clin Pharmacol Ther. 1979;25(3):358–71. transporter inhibitor reboxetine reduces stimulant effects of
31. Liechti ME, Dolder PC, Schmid Y. Alterations in conciousness MDMA (‘‘ecstasy’’) in humans. Clin Pharmacol Ther.
and mystical-type experiences after acute LSD in humans. Psy- 2011;90(2):246–55.
chopharmacology (Berl). 2016;. doi:10.1007/s00213-016-4453-0 40. Van Dyke C, Ungerer J, Jatlow P, et al. Intranasal cocaine: dose
(Epub ahead of print). relationships of psychological effects and plasma levels. Int J
32. Sokoloff L, Perlin S, Kornetsky C, Kety SS. The effects of Psychiatry Med. 1982;12(1):1–13.
D-lysergic acid diethylamide on cerebral circulation and over-all 41. Belleville RE, Fraser HF, Isbell H, et al. Studies on lysergic acid
metabolism. Ann NY Acad Sci. 1957;66(3):468–77. diethylamide (LSD-25): I. Effects in former morphine addicts and
33. Wagner JG, Aghajanian GK, Bing OH. Correlation of perfor- development of tolerance during chronic intoxication. AMA.
mance test scores with ‘‘tissue concentration’’ of lysergic acid Arch Neurol Psychiatry. 1956;76(5):468–78.
diethylamide in human subjects. Clin Pharmacol Ther. 42. Hysek CM, Liechti ME. Effects of MDMA alone and after pre-
1968;9(5):635–8. treatement with reboxetine, duloxetine, clonidine, carvedilol, and
34. Hoch PH. Studies in routes of administration and counteracting doxazosin on pupillary light reflex. Psychopharmacology.
drugs: lysergic acid diethylmide and mescaline in experimental 2012;224:363–76.
psychosis. New York: Grune and Stratton; 1956.
35. Rickli A, Moning OD, Hoener MC, Liechti ME. Receptor
interaction profiles of novel psychoactive tryptamines compared

90
Supplementary Figures

Figure S1. Schematic representation of the pharmacokinetic-pharmacodynamic link model.

91
92
93
94
Figure S2. LSD plasma concentration-time curves. LSD was orally administered at a dose of
100 µg (panels 1-16) or 200 µg (panels 17-40) at t = 0. The data represent individual
observed LSD plasma concentrations as measured at the different time points (□ for 100 µg
and ● for 200 µg LSD) and the LSD concentrations predicted by the one-compartment
pharmacokinetic model (black lines). Note the interindividual variance in plasma
concentrations, especially within the 100 µg LSD dose group (panels 1-16).

95
10

L S D ( n g /m L )
1

0 .1
3
0 1 2 3 4 6 8 10 12
L S D ( n g /m L )

tim e (h )

0
0 1 2 3 4 6 8 10 12
tim e (h )

Figure S3. Plasma concentration-time curves of LSD. Filled circles (●) and empty squares

(□) indicate the mean ± SEM concentrations after 100 and 200 µg LSD. The inset shows the

semilogarithmic plot. First-order kinetics were observed up to 12 h. LSD was administered at

t = 0.

96
97
98
Figure S4. Subjective responses to LSD. LSD was orally administered at a dose of 100 µg
(panels 1-16) or 200 µg (panels 17-40) at t = 0. The data represent individual observed LSD
responses on the “any drug effect” Visual Analog Scale (rated 0-100%) at the different time
points (□ for 100 µg LSD and ● for 200 µg LSD) and the pharmacokinetic-pharmacodynamic
model-predicted effect (black lines).

99
100
101
102
Figure S5. Subjective responses to LSD. LSD was orally administered at a dose of 100 µg
(panels 1-16) or 200 µg (panels 17-40) at t = 0. The data represent individual observed LSD
responses on the “good drug effect” Visual Analog Scale (rated 0-100%) at the different time
points (□ for 100 µg and ● for 200 µg LSD) and the pharmacokinetic-pharmacodynamic
model-predicted effect (black lines).

103
104
105
106
Figure S6. “Bad drug effect” of LSD. LSD was orally administered at a dose of 100 µg
(panels 1-16) or 200 µg (panels 17-40) at t = 0. The data represent individual observed LSD
responses on the “bad drug effect” Visual Analog Scale (rated 0-100%) at the different time
points (□ for 100 µg and ● for 200 µg LSD) and the pharmacokinetic-pharmacodynamic
model-predicted effect (black lines).

107
Figure S7. Association of LSD plasma concentrations (predicted Cmax levels) and peak
subjective effects (predicted any drug effects) for both doses (100 and 200 µg) pooled
(N=40, circles, left panel) and for the 200 µg dose alone (N=16, rectangles, right panel).
Plasma concentrations of LSD are significantly correlated with its subjective effects across
subjects in the pooled sample (Rp = 0.38, p < 0.05, N = 40, left panel). However, plasma
peak concentrations are not significantly correlated with the subjective peak response within
the 100 µg (Rp = 0.38, p = 0.08, N = 24) or the 200 µg (Rp = -0.04, p > 0.8, N = 16) dose
groups.

108
Figure S8. Diagnostic plots for a representative subject of the 100 µg LSD dose study group.
upper left panel: Observed and predicted concentrations of LSD vs. time. (see Figure S2 for
all plots) upper right panel: Observed vs. predicted concentrations of LSD. Middle left panel:
Residual vs. predicted concentrations of LSD. Middle right panel: Observed and predicted
effects of LSD vs. time. (see Figure S4 for all plots). Lower left panel: Observed vs. predicted
effects of LSD. Lower right panel: Residual vs. predicted effects of LSD.

109
Figure S9. Diagnostic plots for a representative subject of the 200 µg LSD dose study group.
upper left panel: Observed and predicted concentrations of LSD vs. time. (see Figure S2 for
all plots) upper right panel: Observed vs. predicted concentrations of LSD. Middle left panel:
Residual vs. predicted concentrations of LSD. Middle right panel: Observed and predicted
effects of LSD vs. time. (see Figure S4 for all plots). Lower left panel: Observed vs. predicted
effects of LSD. Lower right panel: Residual vs. predicted effects of LSD.

110
4. Discussion

4.1 Pharmacokinetics

After no research in humans since the 1970s, we have successfully conducted two
double-blind, placebo-controlled, randomized, cross-over phase I studies in healthy
subjects. We investigated a high dose of 200 µg of LSD in 16 subjects and a
lower dose of 100 µg of LSD in 24 healthy subjects, and characterized psycho-
logical, physiological, and pharmacokinetic effects (56, 68-71).
The development of a sensitive method for the measurement of LSD and its
metabolites was an analytical challenge. Due to the high potency of the substance,
only very low doses are administered and thus result in very low plasma and urine
concentrations. Additionally, the vulnerability of the compound to light and air
demands careful handling. Therefore, we decided to evade purification procedures
with solid-phase or liquid-liquid extraction. These can certainly increase the
concentration and lead to better sensitivity of the LC-MS/MS method, but also form a
time consuming procedure. We established a fast and reliable method for application
in emergency toxicological cases where time is a crucial factor. This method was
then successfully applied in five toxicology cases where consumption of LSD could
be confirmed four times in serum and once in urine (65). Further, we successfully
quantified concentrations of LSD, and its major (urinary) metabolite 2-oxo-3-hydroxy
LSD. Following the controlled administration of 200 µg LSD in our first study, the
metabolite was found to be present at around 10% of the LSD concentration in
plasma, and up to 20-fold the LSD concentration in urine. Confirmation of this
metabolite following the dose of 100 µg was difficult as peak plasma concentrations
of LSD were around 1.5 ng/ml what corresponds to 2-oxo-3-hydroxy LSD
concentrations of 0.15 ng/ml. This is already near to the limit of detection of many
LC-MS/MS methods. Nonetheless, we could confirm the presence of some in-vitro
identified metabolites (54) using more specific LC-MS/MS methods (67). We and
another group (79) were able to detect nor-LSD, LAE, LEO, 13- or 14-hydroxy-LSD,
and 2-oxo LSD in some of the plasma and urine samples after 100 and 200 µg LSD
(67, 79). Nevertheless, the complete metabolic faith of LSD, including involved
enzymes, is still unknown. Figure 1 gives an overview of currently identified and

111
possible metabolites. Future studies should address this issue and use higher doses
of LSD in humans for quantification of metabolites. Further,in vitro/in vivo studies
should clear up the metabolism of LSD including the involved enzyme mechanisms.
Additionally, metabolites need to be commercially available to develop
comprehensive analytical methods for their quantification.

2-oxo-LSD
Iso-LSD Present in rat, mice, guinea pigs,
Synthetic by-product, inactive
monkeys, and humans (41-44)
Identical metabolism to LSD (47)
Inactive in humans (41)
13-OH-LSD > 14-OH-LSD
Nor-LSD
Present in humans (67,79)
Identified in-vitro (47)
Lysergic-acid-ethyl-hydroxyethylamide
(LEO)
Present in humans (67)
pH>9

?
“naphthostyril compound”
10-Hydroxy-9,10-dihydro-LSD (lumi-LSD) Formed out of 2-oxo-LSD
Proposed but unconfirmed metabolite (43) Present in monkey (43)
Formed under UV light (48,49)
d-Lysergic-acid-diethylamide (LSD)

13- or 14-hydroxy-LSD 2-oxo-3-hydroxy-LSD


Major metabolite in rat, guinea pigs (43) Major human metabolite (46,65,67,79)
Present in humans (glucuronides) (67) Lysergic-acid-monoethylamide (LAE)
13-OH-LSD possibly active (43) Present in rat, guinea pigs, monkeys, humans
(43,67)
Active in animal and human

Figure 2 shows possible and already identified metabolites of LSD.


112
With the established LC-MS/MS methods we have assessed data on the plasma
concentration-time curves of LSD. This is crucial because many experimental and
therapeutic studies, some of which have started simultaneously to our studies, did
not determine plasma concentrations. Thus no information on the presence of LSD in
their subjects sample is available.
Maximum plasma concentrations (Cmax) and areas under the curve (AUC) values
were approximately twice as high for the 200 µg dose compared with the 100 µg
dose. Time point of peak plasma concentrations (Tmax) and plasma half-lifes were
similar, consistent with dose-proportional pharmacokinetics. Compartmental
modeling predicted a geometric mean Cmax of 1.3 ng/ml, 1.4 h after the administration
of 100 µg LSD. Geometric mean Cmax values of 3.1 ng/ml were reached 1.5 h after
the administration of 200 µg LSD. The predicted mean half-live of LSD was 2.6 h
after both doses and was thus comparable to the value of 2.9 hours found after
intravenous administration of 2 µg/kg LSD in the 1960s (48, 50-52) but shorter than
the 3.6 hours that we have determined using non-compartmental analysis (68).
Overall, we observed linear dose and elimination kinetics of LSD up to 12 hours after
drug administration.
The present data on the plasma concentration-time curves of LSD are important as
many studies that started investigating LSD did not perform blood sampling. There
are no data available on the plasma concentrations after intravenous administration
of 75 µg LSD base in saline (80), despite the publication of extensive
pharmacodynamic data (75, 80-87).

113
4.2 Pharmacodynamics

In the present studies, LSD produced robust and high subjective drug effects in
almost all of the subjects. The subjective effects lasted 8.2 ± 2.1 hours and 11.6 ± 1.7
hours (mean ± SD) for the 100 and 200 µg LSD doses, respectively. Subjective peak
effects were reached 2.8 hours and 2.5 hours after administration of 100 and 200 µg
LSD, respectively (68, 70, 71).
Both doses of LSD induced subjective feelings of well-being, happiness, closeness to
others, openness, and trust (70). LSD induced a profound altered state of
consciousness on the five dimensions of altered consciousness questionnaire (5D-
ASC) including visual hallucinations, audiovisual synesthesia, positively experienced
derealisation, and depersonalization phenomena (69). These mind altering effects
were dose dependent and have recently been replicated by other research groups
using different doses and routes of administration (12, 69, 75) (Figure 2). Recent
investigations have further shown that these alterations in consciousness are
completely blocked by pretreatment with the selective 5-HT2A antagonist ketanserin
(12, 13). Thus, the 5-HT2A receptor is the main responsible receptor, whereas others
only play a minor role in LSD’s mind altering effects. Further, the overall alterations of
consciousness (5D-ASC total score) were significantly correlated with ratings of
mystical experience on the Mystical Effects Questionnaire (MEQ) (69). These effects
are of importance because strong mystical experiences were associated with positive
long-term effects on mood and personality in healthy subjects and better therapeutic
outcomes in patients with anxiety, depression, and substance use disorder in various
psilocybin studies (24, 88-90). However, in our study with 200 µg LSD, we rarely
observed strong mystical experiences. This raises questions regarding expectancy
effects, placebo responses, and the role of the supervisor and therapist in mystical
experiences. Ratings for a “Bad drug effect” were not present in every subject and
inconsistently occurred throughout the sessions and were typically mild and short
lasting. Adverse effects produced by doses of 100 µg and 200 µg LSD mostly
included complaints like difficulty in concentrating, headache, dizziness, lack off
appetite, nausea, and imbalance (70). All adverse effects completely subsided within
24 - 72 hours. No severe acute adverse effects were observed in both studies and no
reports of flash-back phenomena were registered.
114
Figure 3 shows the mind altering effects of different LSD doses assessed across different research groups with the 5
dimensions of altered states of consciousness questionnaire.
200 µg LSD oral, Basel N=16, 100 µg LSD oral, Basel N=24, 100 µg LSD oral, Zürich N= 22, 75 µg LSD i.v. London N=20

115
4.3 Pharmacokinetics - Pharmacodynamics

The estimates of the corresponding half maximal effective concentration (EC50)


values were in the range of 0.71 - 1.2 ng/ml for positive experienced subjective
effects, and between 1.5 - 2.5 ng/ml for ratings of bad drug effects (68, 71). In our
studies, where relatively high and similar doses of LSD were used, the resulting
plasma concentrations were above the EC50 of the particular response measures.
Therefore responses did not vary across subjects because responses are close to
the maximum. This is important to note and explains why we typically found no
correlations between LSD concentrations and effects across subjects within dose
groups. Probably because of the relatively high concentrations of LSD and the
consistent very high subjective response ratings in most subjects. This finding needs
to be considered when interpreting associations between subjective responses and
other measures. However, these correlations of plasma concentrations with the
subjective and cardiovascular effects of LSD are only weak during the peak response
and typically the case with measures with a maximal effect limit such as subjective
drug effect ratings across different questionnaires and some physiological effects like
pupil size. Still we observed a close relationship between the LSD plasma
concentration and subjective effects.
In both studies, we found no evidence of acute pharmacological tolerance
(represented by a counterclockwise hysteresis, shown in Figure 3) within 12 hours
after the 100 μg dose and within 24 hours after the 200 μg dose. In contrast, other
psychoactive substances, such as 3,4-methylenedioxymethamphetamine (MDMA),
exhibit very marked acute pharmacological tolerance (represented by a clockwise
hysteresis, shown in Fig 3), with a rapid decline of subjective and physiological
effects of MDMA within 4 hours despite continuously high plasma levels.

116
Figure 4 shows the plasma concentration – effect relationships with counterclockwise hysteresis (no acute
tolerance) of LSD and clockwise hysteresis (acute tolerance) of MDMA.

117
4.4 Emotion Recognition and Empathy

Because of the use of LSD, mainly in psychiatric settings and the recreational use,
information about the effects of LSD on social cognition are important. Social
cognition includes aspects of emotion recognition and empathy which describe the
ability to infer another’s thoughts, feelings, and intentions. Those are relevant for a
better understanding of the human brain structures and functioning, as well as simple
social interactions during clinical studies and mainly in psychotherapeutic settings.
Results from earlier studies were primarily observational and thus very subjective.
To allow for a better characterization of the social-cognitive effects of LSD, we used
validated psychometric instruments that have been used with other drugs, such as
MDMA, methylphenidate, and psilocybin (72-74, 76, 77).
Interestingly, 100 and 200 µg LSD positively altered the processing of emotional
information by decreasing the recognition of fearful faces and tended to impair the
recognition of sad faces (70). Further, 200 µg LSD significantly enhanced emotional
empathy whereas the effects of 100 µg LSD did nearly not reach significance.
Overall, these effects are similar to those observed following MDMA administration
which similarly impaired the correct recognition of negative emotions and induced
strong feelings of well-being, and empathy. Similar to LSD, psilocybin decreased the
recognition of negative facial expressions and increased emotional empathy (76, 77).
These findings indicate that LSD affects emotion processing similarly to MDMA and
psilocybin. In line with the findings of impaired recognition of fear following LSD
administration, we found that 100 µg of LSD reduced left amygdala reactivity to the
presentation of fearful faces relative to placebo (78). Similarly, psilocybin and MDMA
decreased amygdala reactivity to negative facial stimuli (91, 92).
The emotional effects during the later phase of the acute LSD response (6-10 h) and
the reduced perception of negative emotions/amygdala reactivity are likely beneficial
in psychotherapeutic settings. Future research should therefore address the relative
contributions of the empathic and emotional effects of LSD to its potential therapeutic
effects.

118
5. Summary and Outlook

The findings of the two clinical studies about the effects of LSD in healthy participants
have translational relevance for further medical investigations. First, we have shown
that LSD can be safely administrated to healthy subjects when closely monitored and
supervised by experienced investigators. Second, the PK-PD relationship shows that
the subjective effects are directly related to the plasma concentrations and that LSD
does not produce acute tolerance. Third, the increase in emotional empathy and the
bias towards the recognition of positive emotions in line with a decreased amygdala
reactivity, might reflect a potentially therapeutic effect by reducing perception of
negative emotions and facilitating the therapeutic alliance in LSD-assisted
psychotherapy (e.g. in anxiety disorders). Currently LSD-assisted psychotherapy is
offered by two psychiatrists to selected patients in Switzerland in the context of
compassionate use which is legally-authorized, but demands case-by-case
authorization by the Swiss Federal Office for Public Health (BAG).

Due to the study results and the pioneer work in the field of compassionate use, we
were able to get the approval for a new clinical phase II study, investigating LSD in
40 patients with anxiety (with or without life threatening diseases). Because the
higher dose of 200 µg LSD produced stronger subjective and emotional effects while
having comparable cardiovascular stimulation as the lower 100 µg dose, it was
selected to be used in this phase II trial. The study has just started and uses a dou-
ble-blind, placebo-controlled, within-subject, cross-over design with two LSD and
two placebo sessions. Our hypothesis is that LSD will significantly reduce anxiety in
these patients. The study will last until 2021.

119
References

1. Hollister LE. Human pharmacology of lysergic acid diethylamide.


Psychopharmacology - A review of progress 1957 - 1967. 1967;Proceedings of the
6th annual meething of the ACNP. San Juan, Puerto Rico. Der. 12-15, 1967:1253.
2. Hoffer A. D-Lysergic Acid Diethylamide (LSD): A Review of its Present Status.
Clin Pharmacol Ther. 1965;6:183.
3. Victor Rossi G. LSD - A pharmacologic profile. Amer.J.Pharm. 1971;143:38.
4. Greiner T, Burch N, Edelberg R. Trheshold doses of LSD in human subjects.
Fed. Proc. 1957;16:303.
5. Abramson HA, Jarvik ME, Kaufman MR, Kornetsky C, Levine A, Wagner M.
Lysergic Acid Diethylamide (LSD-25): I. Physiological and Perceptual Responses.
The Journal of Psychology. 1955;39(1):3.
6. Watts VJ, Lawler CP, Fox DR, Neve KA, Nichols DE, Mailman RB. LSD and
structural analogs: pharmacological evaluation at D1 dopamine receptors.
Psychopharmacology (Berl). 1995;118(4):401.
7. Giacomelli S, Palmery M, Romanelli L, Cheng CY, Silvestrini B. Lysergic acid
diethylamide (LSD) is a partial agonist of D2 dopaminergic receptors and it
potentiates dopamine-mediated prolactin secretion in lactotrophs in vitro. Life Sci.
1998;63(3):215.
8. Halberstadt AL, Geyer MA. Multiple receptors contribute to the behavioral
effects of indoleamine hallucinogens. Neuropharmacology. 2011;61(3):364.
9. Nichols DE. Hallucinogens. Pharmacol Ther. 2004;101(2):131.
10. Egan CT, Herrick-Davis K, Miller K, Glennon RA, Teitler M. Agonist activity of
LSD and lisuride at cloned 5HT2A and 5HT2C receptors. Psychopharmacology
(Berl). 1998;136(4):409.
11. Gresch PJ, Barrett RJ, Sanders-Bush E, Smith RL. 5-Hydroxytryptamine
(serotonin)2A receptors in rat anterior cingulate cortex mediate the discriminative
stimulus properties of d-lysergic acid diethylamide. J Pharmacol Exp Ther.
2007;320(2):662.
12. Preller KH, Herdener M, Pokorny T, Planzer A, Kraehenmann R, Stampfli P, et
al. The Fabric of Meaning and Subjective Effects in LSD-Induced States Depend on
Serotonin 2A Receptor Activation. Curr Biol. 2017;27(3):451.
120
13. Kraehenmann R, Pokorny D, Vollenweider L, Preller KH, Pokorny T, Seifritz E,
et al. Dreamlike effects of LSD on waking imagery in humans depend on serotonin
2A receptor activation. Psychopharmacology (Berl). 2017;10.1007/s00213-017-4610-
0.
14. Passie T, Halpern JH, Stichtenoth DO, Emrich HM, Hintzen A. The
pharmacology of lysergic acid diethylamide: a review. CNS Neurosci Ther.
2008;14(4):295.
15. Abramson HA, Jarvik ME, Gorin MH, Hirsch MW. Lysergic Acid Diethylamide
(LSD-25): XVII. Tolerance Development and its Relationship to a Theory of
Psychosis. The Journal of Psychology. 1956;41(1):81.
16. Isbell H, Fraser H, Wikler A, Belleville R. Tolerance to diethylamide of lysergic
acid (LSD-25). Fed. Proc. 1955;14:355.
17. Isbell H. Comparison of the reactions induced by psilocybin and LSD-25 in
man. Psychopharmacologia. 1959;1(1):29.
18. Abramson HA, Rolo A, Sklarofsky B, Stache J. Production of Cross-Tolerance
to Psychosis-Producing Doses of Lysergic Acid Diethylamide and Psilocybin. The
Journal of Psychology. 1960;49(1):151.
19. McGlothlin WH, Arnold DO, Freedman DX. Organicity measures following
repeated lsd ingestion. Archives of General Psychiatry. 1969;21(6):704.
20. Anastasopoulos G, Photiades H. Effects of LSD-25 on Relatives of
Schizophrenic Patients. The British Journal of Psychiatry. 1962;108(452):95.
21. McGlothlin W, Cohen S, McGlothlin MS. Long lasting effects of LSD on
normals. Arch Gen Psychiatry. 1967;17(5):521.
22. Hofmann A. How LSD originated. J Psychedelic Drugs. 1979;11(1-2):53.
23. Nichols DE. Psychedelics. Pharmacol Rev. 2016;68(2):264.
24. Nichols DE, Johnson MW, Nichols CD. Psychedelics as Medicines: An
Emerging New Paradigm. Clin Pharmacol Ther. 2017;101(2):209.
25. Pahnke WN, Kurland AA, Unger S, Savage C, Grof S. The experimental use
of psychedelic (LSD) psychotherapy. Jama. 1970;212(11):1856.
26. Grof S, Goodman LE, Richards WA, Kurland AA. LSD-assisted psychotherapy
in patients with terminal cancer. Int Pharmacopsychiatry. 1973;8(3):129.
27. Geyer MA, Vollenweider FX. Serotonin research: contributions to
understanding psychoses. Trends Pharmacol Sci. 2008;29(9):445.
121
28. Halberstadt AL, Geyer MA. Serotonergic hallucinogens as translational models
relevant to schizophrenia. Int J Neuropsychopharmacol. 2013;16(10):2165.
29. Anderson EW, Rawnsley K. Clinical studies of lysergic acid diethylamide.
Monatsschr Psychiatr Neurol. 1954;128(1-2):38.
30. Stoll WA. Lysergsäureäthylamid, ein Phantastikum aus der Mutterkorngruppe.
Schweiz Arch Neurol Psychiatr. 1947;60:279.
31. Chwelos N, Blewett D, Smith C, Hoffer A. Use of d-lysergic acid diethylamide
in the treatment of alcoholism. QJ Stud Alcohol. 1959;20:577.
32. Pahnke WN, Kurland AA, Goodman LE, Richards WA. LSD-assisted
psychotherapy with terminal cancer patients. Curr Psychiatr Ther. 1969;9:144.
33. Kast E. LSD and the dying patient. The Chicago Medical School quarterly.
1965;26(2):80.
34. Raffaelli E, Martins OJ, Dágua Filho AdSP. Lisuride in cluster headache.
Headache: The Journal of Head and Face Pain. 1983;23(3):117.
35. Cheek FE. Exploratory study of drugs and social interaction. Archives of
General Psychiatry. 1963;9(6):566.
36. Cheek FE, Holstein CM. Lysergic acid diethylamide tartrate (LSD-25) dosage
levels, group differences and social interaction. J Nerv Ment Dis. 1971;153(2):133.
37. Levine A, Abramson HA, Kaufman MR, Kornetsky C, Markham S. Lysergic
acid diethylamide (LSD-25): XIV. Effect on personality as observed in psychological
tests. Journal of Psychology.. - Provincetown, Mass., etc. : Journal Press, etc.
1955;40(2):351.
38. Lennard H, Jarvik ME, Abramson HA. Lysergic Acid Diethylamide (LSD-25):
XII. A Preliminary Statement of its Effects Upon Interpersonal Communication. The
Journal of Psychology. 1956;41(1):185.
39. Slater PE, Morimoto K, Hyde RW. The effects of LSD upon group interaction.
Arch Gen Psychiatry. 1963;8:564.
40. Stoll A, Rothlin E, Rutschmann J, Schalch WR. Distribution and fate of14C-
labeled lysergic acid diethylamide (LSD 25) in the animal body. Experientia.
1955;11(10):396.
41. Axelrod J, Brady RO, Witkop B, Evarts EV. Metabolism of Lysergic Acid
Diethylamide. Nature. 1956;178(4525):143.

122
42. Axelrod J, Brady RO, Witkop B, Evarts EV. The distribution and metabolism of
lysergic acid diethylamide. Ann N Y Acad Sci. 1957;66(3):435.
43. Siddik ZH, R.D B, Graham Dring L, Smith RL, Tecwyn Williams R. The fate of
lysergic acid di[14C]ethylamide ([14C]LSD) in the rat, guinea pig and rhesus monkey
and of [14C]iso-LSD in rat. Biochem Pharmacol. 1979;28:3093.
44. Boyd ES. Metabolism of lysergic acid diethylamide. Arch. Int. Pharmacodyn.
1959;120:292.
45. Kiechel J-R. Pharmacocinétique et métabolsime des dérivés de l'ergot.
J.Pharmacol. (Paris). 1979;10:533.
46. Poch GK, Klette KL, Hallare DA, Manglicmot MG, Czarny RJ, McWhorter LK,
et al. Detection of metabolites of lysergic acid diethylamide (LSD) in human urine
specimens: 2-oxo-3-hydroxy-LSD, a prevalent metabolite of LSD. J Chromatogr B
Biomed Sci Appl. 1999;724(1):23.
47. Niwaguchi T, Inoue T, Nakahara Y. Studies on enzymatic dealkylation of D-
lysergic acid diethylamide (LSD). Biochem Pharmacol. 1974;23(6):1073.
48. Aghajanian GK, Bing OH. Persistence of lysergic acid diethylamide in the
plasma of human subjects. Clin Pharmacol Ther. 1964;5:611.
49. Upshall DG, Wailling DG. The determination of LSD in human plasma
following oral administration. Clin.chim.Acta. 1972;36:67.
50. Levy G, Gibaldi M, Jusko WJ. Multicompartment pharmacokinetic models
pharmacologic effects. J.pharm.Sci. 1969;50:422.
51. Wagner JG, Aghajanian GK, Bing OH. Correlation of performance test scores
with “tissue concentration” of lysergic acid diethylamide in human subiects. Clinical
Pharmacology & Therapeutics. 1968;9(5).
52. Levy G. Pharmacokinetics of LSD effect. Clinical Pharmacology &
Therapeutics. 1969;10(1):134.
53. Tonini G, Montanari C. Effetti psichici della monoetilamide dell'acido lisergico
(LAE32). Giorn.psichiatr.Ferrara. 1955;83:355.
54. Cai J, Henion J. Elucidation of LSD in vitro metabolism by liquid
chromatography and capillary electrophoresis coupled with tandem mass
spectrometry. J Anal Toxicol. 1996;20(1):27.
55. Canezin J, Cailleux A, Turcant A, Le Bouil A, Harry P, Allain P. Determination
of LSD and its metabolites in human biological fluids by high-performance liquid
123
chromatography with electrospray tandem mass spectrometry. J Chromatogr B
Biomed Sci Appl. 2001;765(1):15.
56. Schmid Y, Enzler F, Gasser P, Grouzmann E, Preller KH, Vollenweider FX, et
al. Acute Effects of Lysergic Acid Diethylamide in Healthy Subjects. Biol Psychiatry.
2015;78(8):544.
57. Gasser P. Die Psycholytische Therapie in der Schweiz von 1988-1993.
Schweiz Arch Neurol Psychiatr. 1996;147:59.
58. Sewell RA, Halpern JH, Pope HG, Jr. Response of cluster headache to
psilocybin and LSD. Neurology. 2006;66(12):1920.
59. Karst M, Halpern JH, Bernateck M, Passie T. The non-hallucinogen 2-bromo-
lysergic acid diethylamide as preventative treatment for cluster headache: an open,
non-randomized case series. Cephalalgia. 2010;30(9):1140.
60. Krebs TS, Johansen PO. Lysergic acid diethylamide (LSD) for alcoholism:
meta-analysis of randomized controlled trials. J Psychopharmacol. 2012;26(7):994.
61. Gasser P, Holstein D, Michel Y, Doblin R, Yazar-Klosinski B, Passie T, et al.
Safety and efficacy of lysergic acid diethylamide-assisted psychotherapy for anxiety
associated with life-threatening diseases. J Nerv Ment Dis. 2014;202(7):513.
62. EMCDDA. European Drug Report 2013. European Monitoring Center for
Drugs and Drug Addiction (EMCDDA). 2013;www.emcdda.europa.eu.
63. Krebs TS, Johansen PO. Over 30 million psychedelic users in the United
States. F1000Res. 2013;2:98.
64. Liakoni E, Dolder PC, Rentsch K, Liechti ME. Acute health problems due to
recreational drug use in patients presenting to an urban emergency department in
Switzerland. Swiss Med Wkly. 2015;145:w14166.
65. Dolder PC, Liechti ME, Rentsch KM. Development and validation of a rapid
turboflow LC-MS/MS method for the quantification of LSD and 2-oxo-3-hydroxy LSD
in serum and urine samples of emergency toxicological cases. Anal Bioanal Chem.
2015;407(6):1577.
66. Liakoni E, Dolder PC, Rentsch KM, Liechti ME. Presentations due to acute
toxicity of psychoactive substances in an urban emergency department in
Switzerland: a case series. BMC Pharmacol Toxicol. 2016;17(1):25.
67. Dolder PC, Liechti ME, Rentsch KM. Development and validation of an LC-
MS/MS method to quantify LSD, iso-LSD, 2-oxo-3-hydroxy LSD, and nor-LSD and
124
identify novel metabolites in plasma samples in a controlled clinical trial.
Anal Bioanal Chem. 2017
68. Dolder PC, Schmid Y, Haschke M, Rentsch KM, Liechti ME. Pharmacokinetics
and Concentration-Effect Relationship of Oral LSD in Humans. Int J
Neuropsychopharmacol. 2015;19(1).
69. Liechti ME, Dolder PC, Schmid Y. Alterations of consciousness and mystical-
type experiences after acute LSD in humans. Psychopharmacology (Berl).
2016;10.1007/s00213-016-4453-0.
70. Dolder PC, Schmid Y, Muller F, Borgwardt S, Liechti ME. LSD Acutely Impairs
Fear Recognition and Enhances Emotional Empathy and Sociality.
Neuropsychopharmacology. 2016;41(11):2638.
71. Dolder PC, Schmid Y, Steuer AE, Kraemer T, Rentsch KM, Hammann F, et al.
Pharmacokinetics and Pharmacodynamics of Lysergic Acid Diethylamide in Healthy
Subjects. Clin Pharmacokinet. 2017;10.1007/s40262-017-0513-9.
72. Schmid Y, Hysek CM, Simmler LD, Crockett MJ, Quednow BB, Liechti ME.
Differential effects of MDMA and methylphenidate on social cognition. J
Psychopharmacol. 2014;28:847.
73. Hysek CM, Schmid Y, Simmler LD, Domes G, Heinrichs M, Eisenegger C, et
al. MDMA enhances emotional empathy and prosocial behavior. Soc Cogn Affect
Neurosci. 2014;9:1645.
74. Liechti ME, Schmid Y, Dolder PC, Hysek C. Effects of MDMA on social
cognition. European Neuropsychopharmacology: Elsevier; 2016. p. S248.
75. Carhart-Harris RL, Kaelen M, Bolstridge M, Williams TM, Williams LT,
Underwood R, et al. The paradoxical psychological effects of lysergic acid
diethylamide (LSD). Psychol Med. 2016;46(7):1379.
76. Preller KH, Pokorny T, Krähenmann R, Dziobek I, Stämpfli P, Vollenweider
FX. The Effect of 5-HT2A/1a Agonist Treatment On Social Cognition, Empathy, and
Social Decision-making. European Psychiatry; //2015. p. 22.
77. Kometer M, Schmidt A, Bachmann R, Studerus E, Seifritz E, Vollenweider FX.
Psilocybin biases facial recognition, goal-directed behavior, and mood state toward
positive relative to negative emotions through different serotonergic subreceptors.
Biol Psychiatry. 2012;72(11):898.

125
78. Mueller F, Lenz C, Dolder PC, Lang UE, Schmidt A, Liechti ME, et al. Acute
effects of LSD on amygdala activity during processing of fearful stimuli in healthy
subjects. Transl Psychiatry. 2017;doi:10.1038/tp.2017.54(7).
79. Steuer AE, Poetzsch M, Stock L, Eisenbeiss L, Schmid Y, Liechti ME, et al.
Development and validation of an ultra-fast and sensitive microflow liquid
chromatography-tandem mass spectrometry (MFLC-MS/MS) method for
quantification of LSD and its metabolites in plasma and application to a controlled
LSD administration study in humans. Drug Test Anal. 2016;10.1002/dta.2042.
80. Tagliazucchi E, Roseman L, Kaelen M, Orban C, Muthukumaraswamy SD,
Murphy K, et al. Increased global functional connectivity correlates with LSD-induced
ego dissolution. Curr Biol. 2016;26(8):1043.
81. Kaelen M, Roseman L, Kahan J, Santos-Ribeiro A, Orban C, Lorenz R, et al.
LSD modulates music-induced imagery via changes in parahippocampal connectivity.
Eur Neuropsychopharmacol. 2016;26:1099.
82. Carhart-Harris RL, Muthukumaraswamy S, Roseman L, Kaelen M, Droog W,
Murphy K, et al. Neural correlates of the LSD experience revealed by multimodal
neuroimaging. Proc Natl Acad Sci U S A. 2016;113:4853.
83. Terhune DB, Luke DP, Kaelen M, Bolstridge M, Feilding A, Nutt D, et al. A
placebo-controlled investigation of synaesthesia-like experiences under LSD.
Neuropsychologia. 2016;88:28.
84. Speth J, Speth C, Kaelen M, Schloerscheidt AM, Feilding A, Nutt DJ, et al.
Decreased mental time travel to the past correlates with default-mode network
disintegration under lysergic acid diethylamide. J Psychopharmacol. 2016;30(4):344.
85. Roseman L, Sereno MI, Leech R, Kaelen M, Orban C, McGonigle J, et al. LSD
alters eyes-closed functional connectivity within the early visual cortex in a retinotopic
fashion. Hum Brain Mapp. 2016;37(8):3031.
86. Lebedev AV, Kaelen M, Lovden M, Nilsson J, Feilding A, Nutt DJ, et al. LSD-
induced entropic brain activity predicts subsequent personality change. Hum Brain
Mapp. 2016;37(9):3203.
87. Kaelen M, Barrett FS, Roseman L, Lorenz R, Family N, Bolstridge M, et al.
LSD enhances the emotional response to music. Psychopharmacology (Berl).
2015;232(19):3607.

126
88. Griffiths RR, Johnson MW, Richards WA, Richards BD, McCann U, Jesse R.
Psilocybin occasioned mystical-type experiences: immediate and persisting dose-
related effects. Psychopharmacology (Berl). 2011;218(4):649.
89. MacLean KA, Johnson MW, Griffiths RR. Mystical experiences occasioned by
the hallucinogen psilocybin lead to increases in the personality domain of openness.
J Psychopharmacol. 2011;25(11):1453.
90. Garcia-Romeu A, Griffiths RR, Johnson MW. Psilocybin-occasioned mystical
experiences in the treatment of tobacco addiction. Curr Drug Abuse Rev.
2014;7(3):157.
91. Bedi G, Phan KL, Angstadt M, de Wit H. Effects of MDMA on sociability and
neural response to social threat and social reward. Psychopharmacology (Berl).
2009;207(1):73.
92. Kraehenmann R, Preller KH, Scheidegger M, Pokorny T, Bosch OG, Seifritz E,
et al. Psilocybin-Induced Decrease in Amygdala Reactivity Correlates with Enhanced
Positive Mood in Healthy Volunteers. Biol Psychiatry. 2015;78(8):572.

127
Curriculum Vitae
Contact Information:

Name: Patrick Christian Dolder


Adress: Division of Clinical Pharmacology & Toxicology
University Hospital Basel
Schanzenstrasse 55
4056 Basel
E-Mail: [email protected]
Personal Information:

Date of Birth: August, 10th 1987


Place of Birth: Geneva
Citizenship: Swiss
Education:

2014 – 2017 PhD in Clinical Research, University Hospital and University Of Basel
Group Prof. Dr. Matthias Liechti, Clinical Pharmacology
Group Prof. Dr. Katharina Rentsch, Clinical Chemistry
2012 – 2014 MSc Pharmaceutical Sciences, University Of Basel
2009 – 2012 BSc Pharmaceutical Sciences, University Of Basel
2008 – 2009 Swiss Academy of Fitness and Sports, Zürich
2004 – 2008 Swiss Olympic Sports School, Gymnasium Liestal

Certifications:

2014 Good Clinical Practice Certifications


(Basic/Sponsor-Investigator/Study Manager)
Awards:

2017 PPHS Stipend Award of the Medical Faculty, University of Basel


2014 Amedis Prize, Pharmaceutical Sciences, University of Basel

128
Publications:

Dolder PC, Mueller F, Schmid Y, Borgwardt S, Liechti ME. Direct comparison of the acute
subjective, emotional, autonomic, and endocrine effects of MDMA, methylphenidate, and
modafinil in healthy subjects. Psychopharmacolgy. 2017. DOI 10.1007/s00213-017-4650-5

Schmidt A , Mueller F, Dolder PC, Schmid Y, Zanchi D, Liechti M, Borgwardt S.


Comparative effects of methylphenidate, modafinil and MDMA on response inhibition neural
networks in healthy subjects. Int J Neuropsychopharmacol. 2017 May 19. doi:
10.1093/ijnp/pyx037.

Dolder PC, Liechti ME, Rentsch KM. Development and validation of an LC-MS/MS method
to quantify LSD, iso-LSD, 2-oxo-3-hydroxy LSD, and nor-LSD and identify novel metabolites
in plasma samples in a controlled clinical trial. J Clin Lab Anal. 2017 May 26. doi:
10.1002/jcla.22265

Binz TM, Williner E, Strajhar P, Dolder PC, Liechti ME, Baumgartner MR, et al. Chiral
Analysis of Amphetamines in Hair by Liquid Chromatography-Tandem Mass Spectrometry:
Compliance-Monitoring of attention deficit hyperactivity disorder (ADHD) patients under
Elvanse(R) therapy and identification after controlled low dose application. Drug Test Anal.
2017;10.1002/dta.2208.

Mueller F, Lenz C, Dolder PC, Lang UE, Schmidt A, Liechti ME, et al. Acute effects of LSD
on amygdala activity during processing of fearful stimuli in healthy subjects. Transl
Psychiatry. 2017;doi:10.1038/tp.2017.54(7).

Kuypers PC, Dolder PC, Ramaekers JG, Liechti ME. Multifaceted empathy of healthy
volunteers after single doses of MDMA: A pooled sample of placebo-controlled studies. J
Psychopharmacol. 2017;https://doi.org/10.1177/026988111769.

Dolder PC, Holze F, Liakoni E, Harder S, Liechti ME. Alcohol acutely enhances decoding of
positive emotions and emotional concern for positive stimuli and facilitates the viewing of
sexual images. Psychopharmacology (Berl). 2017;234(1):41.

Dolder PC, Schmid Y, Muller F, Borgwardt S, Liechti ME. LSD Acutely Impairs Fear
Recognition and Enhances Emotional Empathy and Sociality. Neuropsychopharmacology.
2016;41(11):2638.

Dolder PC, Schmid Y, Steuer AE, Kraemer T, Rentsch KM, Hammann F, et al.
Pharmacokinetics and Pharmacodynamics of Lysergic Acid Diethylamide in Healthy
Subjects. Clin Pharmacokinet. 2017;10.1007/s40262-017-0513-9.

Stoller A, Dolder PC, Bodmer M, Hammann F, Rentsch KM, Exadaktylos AK, et al.
Mistaking 2C-P for 2C-B: What a Difference a Letter Makes. J Anal Toxicol. 2017;41(1):77.

Liakoni E, Dolder PC, Rentsch KM, Liechti ME. Presentations due to acute toxicity of
psychoactive substances in an urban emergency department in Switzerland: a case series.
BMC Pharmacol Toxicol. 2016;17(1):25.

129
Liakoni E, Dolder PC, Rentsch K, Liechti ME. Acute health problems due to recreational
drug use in patients presenting to an urban emergency department in Switzerland. Swiss
Med Wkly. 2015;145:w14166.

Liechti ME, Dolder PC, Schmid Y. Alterations of consciousness and mystical-type


experiences after acute LSD in humans. Psychopharmacology (Berl).
2016;10.1007/s00213-016-4453-0.

Mauermann E, Filitz J, Dolder P, Rentsch KM, Bandschapp O, Ruppen W. Does Fentanyl


Lead to Opioid-induced Hyperalgesia in Healthy Volunteers?: A Double-blind, Randomized,
Crossover Trial. Anesthesiology. 2016;124(2):453.

Mueller F, Lenz C, Steiner M, Dolder PC, Walter M, Lang UE, et al. Neuroimaging in
moderate MDMA use: A systematic review. Neurosci Biobehav Rev. 2016;62:21.

Strajhar P, Schmid Y, Liakoni E, Dolder PC, Rentsch KM, Kratschmar DV, et al. Acute
Effects of Lysergic Acid Diethylamide on Circulating Steroid Levels in Healthy Subjects. J
Neuroendocrinol. 2016;28(3):12374

Dolder PC, Schmid Y, Haschke M, Rentsch KM, Liechti ME. Pharmacokinetics and
Concentration-Effect Relationship of Oral LSD in Humans. Int J Neuropsychopharmacol.
2015;19(1).

Dolder PC, Liechti ME, Rentsch KM. Development and validation of a rapid turboflow LC-
MS/MS method for the quantification of LSD and 2-oxo-3-hydroxy LSD in serum and urine
samples of emergency toxicological cases. Anal Bioanal Chem. 2015;407(6):1577.

130

You might also like