ArticlePDF Available

Abstract and Figures

After no research in humans for >40 years, there is renewed interest in using lysergic acid diethylamide (LSD) in clinical psychiatric research and practice. There are no modern studies on the subjective and autonomic effects of LSD, and its endocrine effects are unknown. In animals, LSD disrupts prepulse inhibition (PPI) of the acoustic startle response, and patients with schizophrenia exhibit similar impairments in PPI. However, no data are available on the effects of LSD on PPI in humans. In a double-blind, randomized, placebo-controlled, crossover study, LSD (200 μg) and placebo were administered to 16 healthy subjects (8 women, 8 men). Outcome measures included psychometric scales; investigator ratings; PPI of the acoustic startle response; and autonomic, endocrine, and adverse effects. Administration of LSD to healthy subjects produced pronounced alterations in waking consciousness that lasted 12 hours. The predominant effects induced by LSD included visual hallucinations, audiovisual synesthesia, and positively experienced derealization and depersonalization phenomena. Subjective well-being, happiness, closeness to others, openness, and trust were increased by LSD. Compared with placebo, LSD decreased PPI. LSD significantly increased blood pressure, heart rate, body temperature, pupil size, plasma cortisol, prolactin, oxytocin, and epinephrine. Adverse effects produced by LSD completely subsided within 72 hours. No severe acute adverse effects were observed. In addition to marked hallucinogenic effects, LSD exerts methylenedioxymethamphetamine-like empathogenic mood effects that may be useful in psychotherapy. LSD altered sensorimotor gating in a human model of psychosis, supporting the use of LSD in translational psychiatric research. In a controlled clinical setting, LSD can be used safely, but it produces significant sympathomimetic stimulation. Copyright © 2014 Society of Biological Psychiatry. Published by Elsevier Inc. All rights reserved.
Content may be subject to copyright.
Archival Report
Acute Effects of Lysergic Acid Diethylamide in
Healthy Subjects
Yasmin Schmid, Florian Enzler, Peter Gasser, Eric Grouzmann, Katrin H. Preller,
Franz X. Vollenweider, Rudolf Brenneisen, Felix Müller, Stefan Borgwardt, and Matthias E. Liechti
ABSTRACT
BACKGROUND: After no research in humans for .40 years, there is renewed interest in using lysergic acid
diethylamide (LSD) in clinical psychiatric research and practice. There are no modern studies on the subjective and
autonomic effects of LSD, and its endocrine effects are unknown. In animals, LSD disrupts prepulse inhibition (PPI) of
the acoustic startle response, and patients with schizophrenia exhibit similar impairments in PPI. However, no data
are available on the effects of LSD on PPI in humans.
METHODS: In a double-blind, randomized, placebo-controlled, crossover study, LSD (200 μg) and placebo were
administered to 16 healthy subjects (8 women, 8 men). Outcome measures included psychometric scales;
investigator ratings; PPI of the acoustic startle response; and autonomic, endocrine, and adverse effects.
RESULTS: Administration of LSD to healthy subjects produced pronounced alterations in waking consciousness
that lasted 12 hours. The predominant effects induced by LSD included visual hallucinations, audiovisual
synesthesia, and positively experienced derealization and depersonalization phenomena. Subjective well-being,
happiness, closeness to others, openness, and trust were increased by LSD. Compared with placebo, LSD
decreased PPI. LSD signicantly increased blood pressure, heart rate, body temperature, pupil size, plasma cortisol,
prolactin, oxytocin, and epinephrine. Adverse effects produced by LSD completely subsided within 72 hours. No
severe acute adverse effects were observed.
CONCLUSIONS: In addition to marked hallucinogenic effects, LSD exerts methylenedioxymethamphetamine-like
empathogenic mood effects that may be useful in psychotherapy. LSD altered sensorimotor gating in a human model
of psychosis, supporting the use of LSD in translational psychiatric research. In a controlled clinical setting, LSD can
be used safely, but it produces signicant sympathomimetic stimulation.
Keywords: Adverse effects, Hormones, LSD, Prepulse inhibition, Subjective effects, Sympathomimetic effects
http://dx.doi.org/10.1016/j.biopsych.2014.11.015
Lysergic acid diethylamide (LSD) is a prototypical classic
hallucinogen (1,2). The psychotropic effects of LSD were
discovered in 1943 by Hofmann in Basel (3). In the 1950s
1970s, LSD was initially used as an experimental tool (psy-
chotomimetic) to study psychotic-like states and model
psychosis (4,5) and as an adjunct in psycholytic psychother-
apy.It has also been investigated for the treatment of
alcoholism (6), addiction (7), cluster headache (8), and anxiety
associated with terminal illness (911). Today, LSD is used
illicitly for recreational (personal or spiritual) purposes. The
lifetime prevalence of LSD use among adults is 6%8%
(12,13). Despite the widespread recreational use, no exper-
imental scientic pharmacologic studies have been conducted
with LSD in the last 40 years, until recently (14). After the initial
psychiatric investigation by Stoll (15), several case reports and
studies in the 1950s and 1960s described aspects of the
psychological effects of LSD (5,1618). However, these stud-
ies were not performed according to current research stand-
ards and did not include control conditions or the systematic
characterization of psychotropic effects. Many studies also
sought to describe the psychotomimetic effects of LSD but
were not designed to measure any positive subjective effects.
Modern experimental studies with hallucinogens in humans
resumed in the 1990s with N-N-dimethyltryptamine (DMT; also
known ayahuasca) (1922), ketamine (2224), and psilocybin
(25,26), but not with LSD. More recently, LSD and psilocybin
have been evaluated in pilot therapeutic studies as treatments
for anxiety in patients with life-threatening diseases (11,27).
Because of the continued popularity of LSD as a recreational
drug and renewed interest in its therapeutic use (11,28), we re-
examined the acute response to LSD in healthy subjects. To
allow for a better characterization of the subjective effects of
LSD, we used psychometric instruments that have been used
with other psychotropic drugs, including hallucinogens,
empathogens, and stimulants (21,22,2932).
Serotoninergic hallucinogens, including psilocybin, DMT,
and LSD, elicit mostly visual perceptual disturbances that
resemble perceptual disturbances observed in early schizo-
phrenia (22,3335). Hallucinogens also induce alterations in
information processing that are similar to those observed in
&2014 Society of Biological Psychiatry 1
ISSN: 0006-3223 Biological Psychiatry ]]], 2014; ]:]]]]]] www.sobp.org/journal
Biological
Psychiatry
SEE COMMENTARY ON PAGE
schizophrenia. Specically, prepulse inhibition (PPI) of the
acoustic startle response serves as an operational measure
of sensorimotor gating that can be assessed in animals and
humans (36). In schizophrenia, PPI is impaired in prodromal
states and early phases (3639), and hallucinogens such as
LSD acutely disrupt PPI in animals (4045). In animals, PPI
serves as a preclinical model of schizophrenia (46). The effects
of LSD on sensorimotor gating function have not yet been
explored in humans and were tested in the present study. We
hypothesized that LSD would produce alterations in waking
consciousness and impair PPI. Additionally, no data are
available on the acute autonomic and adverse effects of
LSD, and the endocrine effects of LSD in humans are
unknown. Up-to-date clinical safety data are mostly missing.
Because of the continued popularity of LSD as a recreational
drug and interest in its therapeutic use, we also examined the
acute somatic and endocrine response to LSD.
METHODS AND MATERIALS
Participants
We recruited 16 healthy subjects (8 men, 8 women; mean age
6SD, 28.6 66.2 years; range, 2551 years) by word of mouth
or an advertisement placed on the web market platform of the
University of Basel. All subjects provided written informed
consent and were paid for their participation. Additionally, we
considered the safety recommendations for high-dose halluci-
nogen research (47,48). The participant characteristics are
described in detail in Supplement 1. Seven subjects had used
a hallucinogen one to three times, and another four subjects
had prior experience with methylenedioxymethamphetamine
(MDMA) (two to four times).
Study Design
A double-blind, placebo-controlled, crossover design was
used with two experimental test sessions in balanced order.
The washout periods between sessions were at least 7 days.
The study was conducted in accordance with the Declaration
of Helsinki and International Conference on Harmonization
Guidelines in Good Clinical Practice and approved by the
Ethics Committee of the Canton of Basel, Switzerland, and
Swiss Agency for Therapeutic Products (Swissmedic). The
administration of LSD to healthy subjects was authorized by
the Swiss Federal Ofce for Public Health, Bern, Switzerland.
The study was registered at ClinicalTrials.gov (NCT01878942).
Drugs
Administration of LSD was in a single absolute dose of 200 mg,
corresponding to a dose of 2.84 6.13 mg/kg body weight
(mean 6SEM; range, 2.043.85 μg). The same dose was used
in LSD-assisted psychotherapy in a clinical study (11). The
dose was within the range of doses taken for recreational
purposes and expected to induce robust effects in humans (1).
The drug preparation is described in Supplement 1.
Study Procedures
The study included a screening visit with the study physician,
a separate psychiatric interview, an additional visit with the
study physician for familiarization, two 25-hour test ses-
sions, and an end-of-study visit. The sessions were con-
ducted in a calm laboratory environment. Only one research
subject and one or two investigators were present during the
test sessions. The test sessions began at 8:15 AM.Aurine
sample was taken to verify abstinence from drugs of abuse,
and a urine pregnancy test was performed in women, and all
subjects underwent baseline measurements. LSD (200 mg) or
placebo was administered at 9:00 AM. The outcome meas-
ures were repeatedly assessed for 24 hours. A standardized
lunch and dinner were served at 1:30 PM and 5:30 PM,
respectively. The subjects were under constant supervision
by the study physician until 1:00 AM. The subjects were never
alone during the rst 16 hours after drug administration,
and the investigator was in a room next to the subject for up
to 24 hours. The subjects were sent home the next day at
9:30 AM.
Subjective Drug Effects
Subjective measures included scores on the 5 Dimensions of
Altered States of Consciousness (5D-ASC) scale (29,49),
visual analog scales (VASs) (50), the Adjective Mood Rating
Scale (AMRS) (51), and the Addiction Research Center Inven-
tory (ARCI) (31). The 5D-ASC scale is designed to be used
retrospectively and was administered 24 hours after drug
administration to rate the peak drug effects. The VASs were
administered repeatedly for up to 24 hours to assess drug
effects over time. The AMRS and ARCI were administered
before and 3, 10, and 24 hours after drug administration. The
procedures are described in detail in Supplement 1.
Acoustic Startle Response Measurement
The eye-blink component of the acoustic startle response was
measured using an electromyographic startle system (EMG-
SR-Lab; San Diego Instruments, San Diego, California) as
described in detail elsewhere (36) and in Supplement 1. Briey,
the session included 16 pulse-alone stimuli (115 dB) and 32
similar pulse trials that were preceded by a 20-msec prepulse
(86 dB) and an interstimulus interval (ISI) of 30, 60, 120, or
2000 msec, resulting in four prepulse trial conditions.
Cardiovascular, Autonomic, Adverse, and Endocrine
Effects
Cardiostimulant (blood pressure and heart rate), autonomic
(body temperature and pupillary function), psychomotor per-
formance, endocrine measures (plasma cortisol, prolactin,
oxytocin, norepinephrine, and epinephrine), and adverse
effects were measured as described in Supplement 1.
Data Analysis
The data were analyzed using STATISTICA Version 12 soft-
ware (StatSoft, Inc, Tulsa, Oklahoma). Peak or peak change
from baseline values were determined for repeated measures.
Data were analyzed using repeated-measures analysis of
variance (ANOVA), with drug (LSD vs. placebo) as the within-
subjects factor. The PPI data were analyzed using repeated-
measures ANOVA, with drug and trial condition (30, 60, 120,
and 2000 msec) as within-subjects factors, followed by direct
Effects of LSD
2Biological Psychiatry ]]], 2014; ]:]]]]]] www.sobp.org/journal
Biological
Psychiatry
comparisons for each trial condition. Modulatory effects of sex
and hallucinogen experience were evaluated by including the
respective between-subjects factor into the ANOVA. Spear-
mans rank correlations were used to determine associations
between measures. The criterion for statistical signicance
was p,.05.
RESULTS
Subjective Drug Effects
Altered States of Consciousness on the 5D-
ASC. Pronounced alterations of waking consciousness were
induced by LSD (Figure 1). Ratings of oceanic boundlessness
[F
1,15
592.3, p,.001] and visionary restructuralization [F
1,15
5243.5, p,.001] were most strongly increased by LSD. The
elevated ratings for oceanic boundlessness indicated that LSD
elicited a state of positively experienced derealization and
depersonalization with predominantly increased ratings for
experience of unity[F
1,15
560.2, p,.001] and blissful
state[F
1,15
568.1, p,.001]. Additionally, LSD produced
marked visionary restructuralization phenomena, including
increased ratings for elementary and complex imagery
[F
1,15
5123.8, p,.001, and F
1,15
555.9, p,.001,
respectively], audiovisual synesthesia[F
1,15
5156.8,
p,.001], and changed meaning of percepts[F
1,15
593.3,
p,.001]. Only minimal auditory alterations[F
1,15
534.5,
p,.001] were induced by LSD. Also, LSD moderately
increased ratings of anxious ego dissolution [F
1,15
516.1,
p,.01], mostly attributable to signicantly increased ratings
for disembodiment[F
1,15
534.4, p,.001] and impaired
control and cognition[F
1,15
525.3, p,.001], but not
anxiety[F
1,15
54.2, p5.06]. Profound anxiety or panic
was not experienced by any subject. However, two subjects
(one woman and one man) reacted with transient anxiety,
including fear of losing control, which completely resolved
without pharmacologic intervention within 23 hours. No sex
differences were observed in the effects of LSD on the 5D-
ASC scale.
Psychotropic Effects over Time on VASs. Subjective
effects on the VASs are shown in Figure 2, and maximal
effects are presented in Table S2 in Supplement 1. The
subjective effects began 3060 min after LSD administration.
Peak effects (any drug effects) were reported after (mean 6
SD) 1.75 6.82 hours. After 5 hours, the subjective effects of
LSD gradually subsided, but effects lasted up to 12 hours after
LSD administration. Three subjects rated the subjective effects
.50% of maximal possible effects at 12 hours. Compared
with placebo, LSD produced pronounced increases in all VAS
ratings, including any drug effects,”“good drug effect,”“drug
high,”“drug liking,and stimulated[all F
1,15
$1931, all
p,.001]. Peak effects for any drug effects,”“good drug
effect,and drug likingreached 90% of the maximal
possible score. Additionally, LSD signicantly increased rat-
ings of empathogenicdrug effects, including happy,
closeness,”“open,and trust[all F
1,15
$34, all p,.001].
LSD decreased subjective concentration [F
1,15
5212.5,
p,.001]. Compared with placebo, LSD induced small but
signicant increases in bad drug effectand fear[F
1,15
5
23.9, p,.001, and F
1,15
513.2, p5.003, respectively]. The
subjective effects of LSD did not differ between sexes.
AMRS. On the AMRS, LSD signicantly increased ratings
of well-being[F
1,15
58.2, p,.05], emotional excitation
Figure 1. Effects of lysergic acid
diethylamide (LSD) on the 5 Dimen-
sions of Altered States of Conscious-
ness scale. LSD predominantly
increased ratings of oceanic bound-
lessness and visionary restructuraliza-
tion. Increased oceanic boundlessness
ratings mostly resulted from higher
ratings for blissful state and experience
of unity. Increases in visionary restruc-
turalization ratings were attributable to
high ratings for audiovisual synesthe-
sia, elementary and complex imagery,
and changed meaning of percepts.
LSD produced only very little auditory
alterations. LSD-induced increases in
anxious ego dissolution were small
because of elevated ratings for disem-
bodiment, impaired control, and cog-
nition but not anxiety. Vigilance was
signicantly reduced by LSD com-
pared with placebo. The global Altered
States of Consciousness score con-
sists of the summation of the oceanic
boundlessness, anxious ego dissolu-
tion, and visionary restructuralization
scores. **p,.01, ***p,.001 com-
pared with placebo. Data are expres-
sed as mean 6SEM in 16 subjects. AA, auditory alterations; AED, anxious ego dissolution; ASC, Altered States of Consciousness; OB, oceanic boundlessness;
VIR, vigilance; VR, visionary restructuralization.
Effects of LSD
Biological Psychiatry ]]], 2014; ]:]]]]]] www.sobp.org/journal 3
Biological
Psychiatry
[F
1,15
517.4, p,.001], inactivity[F
1,15
510.8, p,.01],
introversion[F
1,15
516.9, p,.001], and dreaminess[F
1,15
557.9, p,.001] compared with placebo (Figure 3 and Table
S2 in Supplement 1). Ratings of extroversionor anxiety
were not altered by LSD. No sex differences were observed in
the effects of LSD on the AMRS.
ARCI. Subjective effects on the ARCI are presented in Table
S2 and Figure S1 in Supplement 1. LSD signicantly increased
ratings on the amphetamine group scale [F
1,15
515.8, p5
.001], with a trend toward signicantly reduced ratings on the
benzedrine group scale [F
1,15
53.8, p5.07]. Also, LSD
signicantly increased ratings of euphoria and drug liking on
the morphine-benzedrine group scale [F
1,15
531.3, p,.001],
sedation on the pentobarbital-alcohol group scale [F
1,15
5
52.6, p,.001], and ratings on the LSD group scale [F
1,15
5
24.4, p,.001], a measure of dysphoric and psychotomimetic
changes. No sex differences were observed in the effects of
LSD on the ARCI.
Investigator-Rated Drug Effects. The investigator-rated
drug effects are shown in Table S2 and Figure S2 in
Supplement 1. Investigator ratings of any drug effect[F
1,15
5449.7, p,.001], distance from reality[F
1,15
521.7,
p,.001], happiness[F
1,15
537.4, p,.001], and non-
speech vocalization[F
1,15
56.9, p,.05] were increased by
LSD. Ratings for anxietyor paranoid thinkingwere not
signicantly increased. LSD did not alter the percentage of
time talking with the investigatorcompared with placebo.
Acoustic Startle Response
The effects of LSD on PPI and startle response habituation are
shown in Figure 4. The data from one participant were
excluded because of technical reasons. The two-way ANOVA,
with drug and prepulse trial condition as within-subject
factors, revealed a signicant drug 3prepulse trial interaction
[F
3,42
53.0, p,.05]. LSD signicantly reduced PPI in the 30-
msec and 60-msec trial conditions [F
1,14
55.5, p,.05, and
F
1,14
55.1, p,.05, respectively] and tended to reduce PPI in
the 120-msec trial condition [F
1,14
53.4, p5.09] (Figure 3A).
Compared with placebo, LSD nonsignicantly increased the
startle response (mean reaction amplitude over all pulse-alone
trials [mean 6SD], 571 6321 units and 469 6190 units after
administration of LSD and placebo, respectively). The two-way
ANOVA for pulse-alone trials, with drug and block (time) as
factors, showed a signicant main effect of block, indicating
habituation of the startle response over time [F
3,42
512.8,
p,.001]. No drug 3block interaction was observed,
indicating similar habituation of the response over time in the
LSD and placebo conditions (Figure 4B). Similarly, LSD did not
affect percentage of habituation compared with placebo. No
associations were found between percentage of PPI disruption
and any subjective effect ratings assessed shortly before or
after the startle measurement.
Figure 2. Subjective effects of lysergic acid diethylamide (LSD) over time on the visual analog scales. LSD or placebo was administered at t 50 hours. The
subjective effects began 3060 min after LSD administration, peaked after 15 hours, gradually subsided after 5 hours, and were increased up to 12 hours.
LSD produced signicant changes in all visual analog scale ratings. However, bad drug effectsand fearwere only minimally elevated. LSD also increased
ratings that are typically increased by empathogens, including ratings for happy,”“closeness,”“open,and trust.Data are expressed as mean 6SEM %
maximal values in 16 subjects.
Effects of LSD
4Biological Psychiatry ]]], 2014; ]:]]]]]] www.sobp.org/journal
Biological
Psychiatry
Cardiovascular, Autonomic, Adverse, and Endocrine
Effects
Peak values and statistics are shown in Table S2 in
Supplement 1. Compared with placebo, LSD signicantly
increased systolic [F
1,15
523.77, p,.001] and diastolic
[F
1,15
525.19, p,.001] blood pressure, heart rate [F
1,15
5
15.27, p5.001], and body temperature [F
1,15
511.61,
p5.004] (Figure 5). LSD signicantly increased the pupil size
in the dark and after a light stimulus [F
1,15
522.71 and F
1,15
5
36.33, respectively, both p,.001] (Figure S3 in Supplement 1).
Participantsability to balance on one foot was signicantly
impaired by LSD [F
1,15
526.1, p5.001] (Figure S4 in
Supplement 1). The plasma concentrations of cortisol [F
1,15
5
198.03, p,.001], prolactin [F
1,15
510.13, p,.01], oxytocin
[F
1,15
59.40, p,.01], and epinephrine [F
1,15
58.95, p,.01]
were signicantly increased by LSD (Figure 6). Compared with
placebo, LSD signicantly increased the total acute (010 hours)
[F
1,15
513.67, p,.01] and subacute (1024 hours) [F
1,15
5
7.19, p,.05] adverse effects but not adverse effects at 2472
hours. Adverse effects at 2472 hours did not differ between
LSD and placebo. The frequently reported acute adverse effects
of LSD are presented in Table S3 in Supplement 1. There were
no severe acute effects. The somatic and endocrine effects of
LSD did not differ between sexes.
Figure 3. Subjective effects on the Adjective Mood Rating Scale. Lysergic acid diethylamide (LSD) or placebo was administered at t 50 hours. LSD
induced increases in general well-be ing (A), emotional excitation (B), inactivity (C), introversion (D), and dreaminess (F). LSD did not induce signicant anxiety (E).
Data are expressed as mean 6SEM change from baseline in 16 subjects. *p,.05, **p,.01, ***p,.001 compared with placebo.
Figure 4. Effects of lysergic acid
diethylamide (LSD) on the percentage
of prepulse inhibition of the acoustic
startle response (A) and startle
response habituation over time (B).
LSD signicantly reduced percentage
of prepulse inhibition in trials with
prepulses that were presented 30
msec or 60 msec before the startle
pulse compared with placebo (A).
A trend toward a signicant reduction
of percentage of prepulse inhibition
was observed for the 120-msec pre-
pulse trial condition. LSD did not
signicantly alter the startle response
or startle response habituation com-
pared with placebo (B). Data are
expressed as mean 6SEM in 15 subjects. *p,. 05,
(
*
)
p5.09 compared with placebo.
Effects of LSD
Biological Psychiatry ]]], 2014; ]:]]]]]] www.sobp.org/journal 5
Biological
Psychiatry
DISCUSSION
The subjective effects of LSD began 3060 min after admin-
istration and peaked at 1.75 hours but remained high for 35
hours before gradually declining. LSD induced a pronounced
alteration in waking consciousness, including visual percep-
tual alterations, audiovisual synesthesia, and positively expe-
rienced derealization and depersonalization. LSD did not
induce pronounced anxiety and overall produced high ratings
of good drug effects and low ratings of bad drug effects.
Feelings of well-being, happiness, closeness to others, open-
ness, and trust were also increased by LSD, effects typically
associated with the empathogen MDMA (Ecstasy) (52).
The acute psychological effects of LSD lasted 12 hours in
most subjects and up to 16 hours in some, which is longer
Figure 5. Effect of lysergic acid
diethylamide (LSD) on vital signs.
LSD or placebo was administered at
t50 hours. LSD signicantly inc-
reased systolic (A) and diastolic (B)
blood pressure, heart rate (C), and
body temperature (D) compared with
placebo. Comparisons for each time
point revealed that the cardiostimu-
lant (A, B) and thermogenic (D)
changes induced by LSD were signif-
icant up to 5 hours after drug admin-
istration compared with placebo, but
moderate trend changes remained up
to 11 hours before the levels returned
to baseline. Data are expressed as
mean 6SEM in 16 subjects. *p,.05,
**p,.01, ***p,.001 compared with
placebo.
Figure 6. Endocrine effects of lysergic acid diethylamide (LSD). LSD signicantly increased the plasma concentrations of prolactin (A), cortisol (B),oxytocin(C),
and epinephrine (D). LSD did not signicantly alter the plasma levels of norepinephrine (E). LSD or placebo was administered at t 50 min. Data are expressed as
mean 6SEM in 16 subjects. **p,.01, ***p,.001 compared with placebo.
Effects of LSD
6Biological Psychiatry ]]], 2014; ]:]]]]]] www.sobp.org/journal
Biological
Psychiatry
than the 610 hours or 12 hours reported by other authors
(1,17,53); this could be attributable to the relatively high dose
of LSD or more sensitive psychometric measures used in the
present study. The effects of LSD lasted twice as long as the
effects of psilocybin (6 hours) (54,55), lasted longer than the
effects of DMT (,1 hour) (19), and possibly lasted a similar
duration as the effects of mescaline (18,56).
In the present study, LSD produced higher scores on the
5D-ASC scale compared with psilocybin in a similar popula-
tion of healthy subjects (55). In particular, LSD produced 30%
higher ratings for oceanic boundlessness (mostly blissful
state), 30% higher ratings for anxious ego dissolution, and
63% higher ratings for visionary restructuralization (mostly
greater audiovisual synesthesia) compared with a high dose of
psilocybin (55,57). Compared with DMT and ketamine, LSD
produced 50% higher ratings for oceanic boundlessness, 50%
higher ratings for visionary restructuralization, and comparably
high ratings for anxious ego dissolution (22,29). On the AMRS,
LSD produced similar ratings for emotional excitation, inacti-
vation, and dreaminess compared with high-dose psilocybin
(55). Similar to LSD, mean group anxiety scores were not
appreciably increased by psilocybin (55). On the ARCI, LSD
increased ratings on the amphetamine group scale and
morphine-benzedrine group scale, suggesting stimulant and
euphoric subjective effects that were similar to MDMA (58).
In contrast, LSD reduced ratings on the benzedrine group
scale, suggesting reduced energy and focus (58). LSD had
overall similar effects to psilocybin on the ARCI (59). Sub-
jective VAS ratings for happy, open, closeness to others, and
trust were increased by LSD. Similarly, the investigators rated
subjects as being happier after administration of LSD com-
pared with placebo. Similar subjective effects are typically
produced by empathogens, such as MDMA, but not by
stimulants (30,60).
Altogether, the psychometric ndings indicate that LSD
produced stronger perceptual alterations than the doses of
other psychotropic drugs tested so far as well as MDMA-like
empathogenic mood effects. Additionally, LSD increased
plasma oxytocin levels. Oxytocin is thought to contribute to
the empathogenic and prosocial effects of MDMA (61) and
may have similar effects in the case of LSD. Although LSD and
MDMA were not compared in the same subjects, the present
ndings indicate that LSD exerts partially MDMA-like empa-
thogenic effects that may be associated with common sero-
toninergic or oxytocinergic properties (52). MDMA produces
weak LSD-like perceptual alterations, likely via similar
5-hydroxytryptamine 2A (5-HT
2A
) receptor stimulation (62).
Pharmacologically, LSD acts as a direct partial agonist at
serotoninergic receptors (2,63), whereas MDMA mostly acts
as an indirect serotoninergic agonist by releasing serotonin via
the serotonin transporter (64).
The primary safety concerns with hallucinogen research
are psychological rather than somatic adverse effects (1).
In laboratory studies that use psilocybin, ketamine, or MDMA,
moderate anticipatory anxiety is common at the beginning of
the onset of the drugs effects (49,65,66). Acute anxiety was
also infrequently reported when LSD was administered at the
same dose as the one used in the present study for LSD-
assisted psychotherapy in patients with anxiety associated
with life-threatening diseases (11). In the present study, LSD
produced anxiety in two subjects, which resolved spontane-
ously with verbal support from the investigators. Anxiety was
related to fear of loss of thought control, disembodiment, and
loss of self as similarly described for psilocybin (55). Some
subjects also had to be reminded of the transient state of the
drug-induced experience. None of the subjects had a current
or past history of major psychiatric disorders, and all were well
informed about the setting and acquainted with and constantly
supervised by the same investigator. Also, only half of the
subjects in the present study were hallucinogen-naïve, and the
other half had very limited prior experience with hallucinogenic
drugs. We found no differences in the quality and extent of the
response to LSD between the hallucinogen-naïve and moder-
ately experienced subjects. Consistent with this nding, prior
experience with hallucinogenic drugs affected the response to
psilocybin only moderately in a similar research setting (66).
In line with our hypothesis, LSD disrupted PPI and pro-
duced sensorimotor decits similar to decits observed in
schizophrenia (3639). In animals, LSD (4042) and other
serotoninergic hallucinogens (4345) reduce PPI. Also, LSD
potentiated the startle magnitude and impaired habituation of
the startle response in rats (67). Similar decits in habituation
were reported in patients with schizophrenia (36,38). Consis-
tent with the preclinical ndings, LSD reduced PPI in the
present study at the 30120 msec ISI. The startle response
amplitude or its habituation was not signicantly altered by
LSD. Psilocybin reduced PPI at a short ISI (30 msec), had no
effect at a medium ISI (60 msec), and increased PPI at long
ISIs (1202000 msec), without changing startle reactivity or
habituation (68,69). The effects of LSD and psilocybin on the
acoustic startle response and its modulation were quite
similar. Additionally, the disruption of PPI induced by psilocy-
bin in humans at a short ISI (30 msec) was prevented by
administration of a 5-HT
2A
receptor antagonist (70), consistent
with similar preclinical studies of LSD (42). In contrast to the
ndings with LSD and psilocybin, DMT or ayahuasca had no
effects on PPI, startle reactivity, or habituation in humans
(20,71). Altogether, the effects of LSD on PPI in normal
humans were consistent with the PPI decits after LSD
administration in animals and sensorimotor gating decits in
patients with schizophrenia.
Serotoninergic hallucinogens, including LSD, are hypothe-
sized to act at the 5-HT
2A
receptor (2,54), which is upregulated
in patients with schizophrenia (72). Genetic variations in the
5-HT
2A
receptor gene inuence PPI (73), and PPI decits
induced by psilocybin in humans depend on 5-HT
2A
receptor
stimulation (70). In animals, LSD disrupts PPI (4042) also via
5-HT
2A
receptor stimulation (42). To characterize further the
role of 5-HT
2
receptors and other receptors in the subjective
and sensorimotor psychotomimetic effects of LSD in humans,
future studies should investigate the effects of receptor
antagonists on the response to LSD using a similar exper-
imental setting. The present ndings lend support to the use of
LSD to study the neurobiological basis of psychotic states in
humans. To date, brain activation patterns have not been
studied using LSD in neuroimaging studies, in contrast to
several modern investigations that used psilocybin to model
psychotic states (25,26).
Signicant sympathomimetic effects, including increases in
blood pressure, heart rate, and pupil size, were produced by
Effects of LSD
Biological Psychiatry ]]], 2014; ]:]]]]]] www.sobp.org/journal 7
Biological
Psychiatry
LSD. Similar ndings were reported in early studies in the
1950s (7478). In contrast, LSD (200 μg administered orally)
did not alter diastolic or systolic blood pressure or heart rate in
a recent study in eight patients with different chronic life-
threatening illnesses (11). Overall, the cardiostimulant effects
of LSD were moderate and smaller than the effects seen with
empathogens and stimulants (30). The LSD-induced increase
in epinephrine levels in the present study was similar to the
effect produced by MDMA (79).
Body temperature was increased by LSD in the present
study. In animals, LSD is thermogenic (80), and hyperthermia
has been reported to be a consequence of massive LSD
overdose in humans (81). Other serotoninergic hallucinogens,
including psilocybin and DMT, produce similar cardiostimulant
and autonomic responses to LSD (18,55,59,8284).
In the present study, LSD increased circulating levels of
cortisol and prolactin. LSD binds to dopaminergic D
2
recep-
tors (85). Studies in rats showed that LSD inhibited prolactin
secretion by rat pituitary cells (86) and decreased plasma
levels of prolactin in rats (87). These ndings led to the
suggestion that LSD acts as a dopamine D
2
receptor agonist
in the pituitary. However, the present study in humans found
that LSD increased the plasma levels of prolactin and cortisol,
which are markers of serotoninergic activity (88,89). Our
ndings suggest that the serotoninergic stimulant effects of
LSD on prolactin regulation usurp any dopamine D
2
receptor
mediated inhibition in humans at the dose used in the present
study. Other serotoninergic drugs, including psilocybin (55),
DMT (84), ayahuasca (90), and MDMA (30,91), increased the
plasma levels of prolactin and cortisol in humans.
The present study has several limitations. First, we used
only a single dose of LSD, and we cannot provide dose-
response data. We used a relatively high dose of LSD (200 μg),
which produced a full and representative LSD response (1).
The same dose of LSD was also used recently in patients with
anxiety associated with terminal illness (11). Second, although
we used formal blinding, the overt subjective effects of LSD
unblinded the treatment assignment. Additionally, expecta-
tions may have inuenced the psychological effects of LSD
because all of the subjects knew that they would receive LSD
or placebo and not another active drug. The psychological
effects and risks of LSD are likely to be different from effects
described herein if LSD is used recreationally in unsupervised
settings or in subjects with psychiatric disorders. Third,
endocrine measures were performed only at two time points
during the expected peak drug effect, not allowing for a full
characterization of the endocrine effects of LSD over a longer
time interval.
In conclusion, LSD produced marked effects on perception
and subjective effects on mood that were similar to effects
reported for MDMA and increased plasma oxytocin, suggest-
ing empathogenic properties that may be useful in psycho-
therapy (11). Consistent with preclinical data and the
sensorimotor decits seen in schizophrenia, LSD acutely
decreased PPI of the acoustic startle response. The present
experimental human study may serve as an interface for the
translation of preclinical research with hallucinogens to clinical
research ndings in patients with schizophrenia and vice
versa. Also, LSD may be useful for further study of alterations
in consciousness and information processing in humans.
The present study showed that LSD can be safely adminis-
tered in an experimental research setting in humans, forming a
basis for further psychopharmacologic studies. However, the
sympathomimetic stimulant effects need to be considered
when LSD is to be used in patients with hypertension or heart
disease.
ACKNOWLEDGMENTS AND DISCLOSURES
This work was supported by the University Hospital Basel, Switzerland, and
the Swiss National Science Foundation Grant No. 320030_1449493.
The authors report no biomedical nancial interests or potential conicts
of interest.
ClinicalTrials.gov: Psychological, Physiological, Endocrine, and Pharma-
cokinetic Effects of LSD in a Controlled Study; http://clinicaltrials.gov/ct2/
show/NCT01878942.
ARTICLE INFORMATION
From Psychopharmacology Research, Clinical Pharmacology and Toxicol-
ogy, Department of Biomedicine and Department of Clinical Research (YS,
FE, MEL), University Hospital Basel, Basel; Private practice for Psychiatry
and Psychotherapy (PG), Solothurn; Biomedicine Service (EG), University
Hospital Lausanne, Lausanne; Neuropsychopharmacology and Brain Imag-
ing and Heffter Research Center, Department of Psychiatry, Psychotherapy
and Psychosomatics (KHP, FXV), University Hospital of Psychiatry Zurich,
Zurich; Department of Clinical Research (RB), University of Bern, Bern; and
Department of Psychiatry (FM, SB), University of Basel, Basel, Switzerland.
Address correspondence to Matthias E. Liechti, M.D., Clinical Pharma-
cology, University Hospital Basel, Hebelstrasse 2, Basel, CH-4031, Switzer-
land; E-mail: matthias.liechti@usb.ch.
Received Sep 30, 2014; revised Oct 28, 2014; accepted Nov 11, 2014.
Supplementary material cited in this article is available online at http://
dx.doi.org/10.1016/j.biopsych.2014.11.015.
REFERENCES
1. Passie T, Halpern JH, Stichtenoth DO, Emrich HM, Hintzen A (2008):
The pharmacology of lysergic acid diethylamide: A review. CNS
Neurosci Ther 14:295314.
2. Nichols DE (2004): Hallucinogens. Pharmacol Ther 101:131181.
3. Hofmann A (1979): How LSD originated. J Psychedelic Drugs 11:
5360.
4. Koelle GB (1958): The pharmacology of mescaline and D-lysergic acid
diethylamide (LSD). N Engl J Med 258:2532.
5. Bercel NA, Travis LE, Olinger LB, Dreikurs E (1956): Model psychoses
induced by LSD-25 in normals. I. Psychophysiological investigations,
with special reference to the mechanism of the paranoid reaction.
AMA Arch Neurol Psychiatry 75:588611.
6. Krebs TS, Johansen PO (2012): Lysergic acid diethylamide (LSD) for
alcoholism: Meta-analysis of randomized controlled trials. J Psycho-
pharmacol 26:9941002.
7. Savage C, McCabe OL (1973): Residential psychedelic (LSD) therapy
for narcotic addict: A controlled study. Arch Gen Psychiatry 28:
808814.
8. Sewell RA, Halpern JH, Pope HG Jr (2006): Response of cluster
headache to psilocybin and LSD. Neurology 66:19201922.
9. Grof S, Goodman LE, Richards WA, Kurland AA (1973): LSD-assisted
psychotherapy in patients with terminal cancer. Int Pharmacopsychia-
try 8:129144.
10. Pahnke WN, Kurland AA, Goodman LE, Richards WA (1969): LSD-
assisted psychotherapy with terminal cancer patients. Curr Psychiatr
Ther 9:144152.
11. Gasser P, Holstein D, Michel Y, Doblin R, Yazar-Klosinski B, Passie T,
et al. (2014): Safety and efcacy of lysergic acid diethylamide-assisted
psychotherapy for anxiety associated with life-threatening diseases.
J Nerv Ment Dis 202:513520.
Effects of LSD
8Biological Psychiatry ]]], 2014; ]:]]]]]] www.sobp.org/journal
Biological
Psychiatry
12. European Monitoring Center for Drugs and Drug Addiction. European
drug report 2014. Available at: www.emcdda.europa.eu. Accessed
August 13, 2014.
13. Johnston LD, O Malley PM, Bachmann JG, Schulenberg JE, Miech RA
(2014): Monitoring the Future: National Survey Results on Drug Use,
1975-2013: Volume 2, College Students and Adults Ages 19-55. Ann
Arbor: Institute for Social Research, University of Michigan.
14. Carhart-Harris RL, Kaelen M, Whalley MG, Bolstridge M, Feilding A,
Nutt DJ (2014): LSD enhances suggestibility in healthy volunteers.
Psychopharmacology. http://dx.doi/10.1007/s00213-014-3714-z.
15. Stoll WA (1947): Lysergsäureäthylamid, ein Phantastikum aus der
Mutterkorngruppe. Schweiz Arch Neurol Psychiatr 60:279323.
16. Rothlin E (1957): Lysergic acid diethylamide and related substances.
Ann N Y Acad Sci 66:668676.
17. Salvatore S, Hyde RW (1956): Progression of effects of lysergic acid
diethylamide (LSD). AMA Arch Neurol Psychiatry 76:5059.
18. Hollister LE, Hartman AM (1962): Mescaline, lysergic acid diethyla-
mide and psilocybin comparison of clinical syndromes, effects on
color perception and biochemical measures. Compr Psychiatry 3:
235242.
19. Strassman RJ, Qualls CR, Uhlenhuth EH, Kellner R (1994): Dose-
response study of N,N-dimethyltryptamine in humans. II. Subjective
effects and preliminary results of a new rating scale. Arch Gen
Psychiatry 51:98108.
20. Riba J, Rodriguez-Fornells A, Barbanoj MJ (2002): Effects of aya-
huasca on sensory and sensorimotor gating in humans as measured
by P50 suppression and prepulse inhibition of the startle reex,
respectively. Psychopharmacology 165:1828.
21. Dos Santos RG, Valle M, Bouso JC, Nomdedeu JF, Rodriguez-
Espinosa J, McIlhenny EH, et al. (2011): Autonomic, neuroendocrine,
and immunological effects of ayahuasca: A comparative study with
d-amphetamine. J Clin Psychopharmacol 31:717726.
22. Gouzoulis-Mayfrank E, Heekeren K, Neukirch A, Stoll M, Stock C,
Obradovic M, et al. (2005): Psychological effects of (S)-ketamine and
N,N-dimethyltryptamine (DMT): A double-blind, cross-over study in
healthy volunteers. Pharmacopsychiatry 38:301311.
23. Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD,
et al. (1994): Subanesthetic effects of the noncompetitive NMDA
antagonist, ketamine, in humans: Psychotomimetic, perceptual, cogni-
tive, and neuroendocrine responses. Arch Gen Psychiatry 51:199214.
24. Vollenweider FX, Leenders KL, Scharfetter C, Antonini A, Maguire P,
Missimer J, et al. (1997): Metabolic hyperfrontality and psychopathol-
ogy in the ketamine model of psychosis using positron emission
tomography (PET) and [18F]uorodeoxyglucose (FDG). Eur Neuro-
psychopharmacol 7:924.
25. Carhart-Harris RL, Erritzoe D, Williams T, Stone JM, Reed LJ,
Colasanti A, et al. (2012): Neural correlates of the psychedelic state
as determined by fMRI studies with psilocybin. Proc Natl Acad Sci U S
A 109:21382143.
26. Vollenweider FX, Leenders KL, Scharfetter C, Maguire P, Stadelmann
O, Angst J (1997): Positron emission tomography and uorodeox-
yglucose studies of metabolic hyperfrontality and psychopathology in
the psilocybin model of psychosis. Neuropsychopharmacology 16:
357372.
27. Grob CS, Danforth AL, Chopra GS, Hagerty M, McKay CR, Halber-
stadt AL, et al. (2011): Pilot study of psilocybin treatment for anxiety in
patients with advanced-stage cancer. Arch Gen Psychiatry 68:7178.
28. Kupferschmidt K (2014): High hopes. Science 345:1823.
29. Studerus E, Gamma A, Vollenweider FX (2010): Psychometric evalua-
tion of the altered states of consciousness rating scale (OAV). PLoS
One 5:e12412.
30. Hysek CM, Simmler LD, Schillinger N, Meyer N, Schmid Y, Donzelli M,
et al. (2014): Pharmacokinetic and pharmacodynamic effects of
methylphenidate and MDMA administered alone and in combination.
Int J Neuropsychopharmacol 17:371381.
31. Martin WR, Sloan JW, Sapira JD, Jasinski DR (1971): Physiologic,
subjective, and behavioral effects of amphetamine, methamphet-
amine, ephedrine, phenmetrazine, and methylphenidate in man. Clin
Pharmacol Ther 12:245258.
32. Hill HE, Haertzen CA, Wolbach AB Jr, Miner EJ (1963): The Addiction
Research Center Inventory: Standardization of scales which evaluate
subjective effects of morphine, amphetamine, pentobarbital, alcohol,
LSD-25, pyrahexyl and chlorpromazine. Psychopharmacologia 4:
167183.
33. Gouzoulis-Mayfrank E, Habermeyer E, Hermle L, Steinmeyer A, Kunert
H, Sass H (1998): Hallucinogenic drug induced states resemble acute
endogenous psychoses: Results of an empirical study. Eur Psychiatry
13:399406.
34. Geyer MA, Vollenweider FX (2008): Serotonin research: Contributions
to understanding psychoses. Trends Pharmacol Sci 29:445453.
35. Langs RJ, Barr HL (1968): Lysergic acid diethylamide (LSD-25) and
schizophrenic reactions: A comparative study. J Nerv Ment Dis 147:
163172.
36. Braff DL, Grillon C, Geyer MA (1992): Gating and habituation of the
startle reex in schizophrenic patients. Arch Gen Psychiatry 49:
206215.
37. Kumari V, Soni W, Mathew VM, Sharma T (2000): Prepulse inhibition
of the startle response in men with schizophreni: Effects of age of
onset of illness, symptoms, and medication. Arch Gen Psychiatry 57:
609614.
38. Ludewig K, Geyer MA, Vollenweider FX (2003): Decits in prepulse
inhibition and habituation in never-medicated, rst-episode schizo-
phrenia. Biol Psychiatry 54:121128.
39. Quednow BB, Frommann I, Berning J, Kuhn KU, Maier W, Wagner M
(2008): Impaired sensorimotor gating of the acoustic startle response
in the prodrome of schizophrenia. Biol Psychiatry 64:766773.
40. Halberstadt AL, Geyer MA (2010): LSD but not lisuride disrupts
prepulse inhibition in rats by activating the 5-HT
2A
receptor. Psycho-
pharmacology (Berl) 208:179189.
41. Palenicek T, Hlinak Z, Bubenikova-Valesova V, Novak T, Horacek J
(2010): Sex differences in the effects of N,N-diethyllysergamide (LSD)
on behavioural activity and prepulse inhibition. Prog Neuropsycho-
pharmacol Biol Psychiatry 34:588596.
42. Ouagazzal A, Grottick AJ, Moreau J, Higgins GA (2001): Effect of LSD
on prepulse inhibition and spontaneous behavior in the rat: A
pharmacological analysis and comparison between two rat strains.
Neuropsychopharmacology 25:565575.
43. Sipes TE, Geyer MA (1997): DOI disrupts prepulse inhibition of startle in
rats via 5-HT
2A
receptors in the ventral pallidum. Brain Res 761:97104.
44. Varty GB, Higgins GA (1995): Examination of drug-induced and
isolation-induced disruptions of prepulse inhibition as models to
screen antipsychotic drugs. Psychopharmacology (Berl) 122:1526.
45. Krebs-Thomson K, Ruiz EM, Masten V, Buell M, Geyer MA (2006): The
roles of 5-HT
1A
and 5-HT
2
receptors in the effects of 5-MeO-DMT on
locomotor activity and prepulse inhibition in rats. Psychopharmacol-
ogy (Berl) 189:319329.
46. Halberstadt AL, Geyer MA (2013): Serotonergic hallucinogens as
translational models relevant to schizophrenia. Int J Neuropsycho-
pharmacol 16:21652180.
47. Johnson M, Richards W, Grifths R (2008): Human hallucinogen
research: Guidelines for safety. J Psychopharmacol 22:603620.
48. Gouzoulis-Mayfrank E, Schneider F, Friedrich J, Spitzer M, Thelen B,
Sass H (1998): Methodological issues of human experimental research
with hallucinogens. Pharmacopsychiatry 31(suppl 2):114118.
49. Dittrich A (1998): The standardized psychometric assessment of
altered states of consciousness (ASCs) in humans. Pharmacopsy-
chiatry 31(suppl 2):8084.
50. Farre M, Abanades S, Roset PN, Peiro AM, Torrens M, OʼMathuna B,
et al. (2007): Pharmacological interaction between 3,4-methylenedioxy-
methamphetamine (ecstasy) and paroxetine: Pharmacological effects
and pharmacokinetics. J Pharmacol Exp Ther 323:954962.
51. Janke W, Debus G (1978): Die Eigenschaftswörterliste. Göttingen:
Hogrefe
52. Hysek CM, Schmid Y, Simmler LD, Domes G, Heinrichs M, Eiseneg-
ger C, et al. (2014): MDMA enhances emotional empathy and
prosocial behavior. Soc Cogn Affect Neurosci 9:16451652.
53. Abraham HD, Aldridge AM, Gogia P (1996): The psychopharmacology
of hallucinogens. Neuropsychopharmacology 14:285298.
Effects of LSD
Biological Psychiatry ]]], 2014; ]:]]]]]] www.sobp.org/journal 9
Biological
Psychiatry
54. Vollenweider FX, Vollenweider-Scherpenhuyzen MF, Babler A, Vogel
H, Hell D (1998): Psilocybin induces schizophrenia-like psychosis in
humans via a serotonin-2 agonist action. Neuroreport 9:38973902.
55. 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. Psy-
chopharmacology 172:145156.
56. Shulgin AT (1973): Mescaline: The chemistry and pharmacology of its
analogs. Lloydia 36:4658.
57. Vollenweider FX, Kometer M (2010): The neurobiology of psychedelic
drugs: Implications for the treatment of mood disorders. Nat Rev
Neurosci 11:642651.
58. Hysek CM, Domes G, Liechti ME (2012): MDMA enhances mind
readingof positive emotions and impairs mind readingof negative
emotions. Psychopharmacology (Berl) 222:293302.
59. Grifths RR, Richards WA, McCann U, Jesse R (2006): Psilocybin can
occasion mystical-type experiences having substantial and sustained
personal meaning and spiritual signicance. Psychopharmacology
(Berl) 187:268283.
60. Schmid Y, Hysek CM, Simmler LD, Crockett MJ, Quednow BB, Liechti
ME (2014): Differential effects of MDMA and methylphenidate on
social cognition. J Psychopharmacol 28:847856.
61. Ramos L, Hicks C, Kevin R, Caminer A, Narlawar R, Kassiou M, et al.
(2013): Acute prosocial effects of oxytocin and vasopressin when
given alone or in combination with 3,4-methylenedioxymethamphet-
amine in rats: involvement of the V
1A
receptor. Neuropsychopharma-
cology 38:22492259.
62. Liechti ME, Saur MR, Gamma A, Hell D, Vollenweider FX (2000):
Psychological and physiological effects of MDMA (Ecstasy) after
pretreatment with the 5-HT
2
antagonist ketanserin in healthy humans.
Neuropsychopharmacology 23:396404.
63. Titeler M, Lyon RA, Glennon RA (1988): Radioligand binding evidence
implicates the brain 5-HT
2
receptor as a site of action for LSD and
phenylisopropylamine hallucinogens. Psychopharmacology (Berl) 94:
213216.
64. Hysek CM, Simmler LD, Nicola V, Vischer N, Donzelli M, Krähenbühl
S, et al. (2012): Duloxetine inhibits effects of MDMA (ecstasy) in vitro
and in humans in a randomized placebo-controlled laboratory study.
PLoS One 7:e36476.
65. Liechti ME, Gamma A, Vollenweider FX (2001): Gender differences in
the subjective effects of MDMA. Psychopharmacology (Berl) 154:
161168.
66. Studerus E, Gamma A, Kometer M, Vollenweider FX (2012): Prediction
of psilocybin response in healthy volunteers. PLoS One 7:e30800.
67. Braff DL, Geyer MA (1980): Acute and chronic LSD effects on rat
startle: Data supporting an LSD rat model of schizophrenia. Biol
Psychiatry 15:909916.
68. Vollenweider FX, Csomor PA, Knappe B, Geyer MA, Quednow BB
(2007): The effects of the preferential 5-HT2A agonist psilocybin on
prepulse inhibition of startle in healthy human volunteers depend on
interstimulus interval. Neuropsychopharmacology 32:18761887.
69. Gouzoulis-Mayfrank E, Heekeren K, Thelen B, Lindenblatt H, Kovar
KA, Sass H, et al. (1998): Effects of the hallucinogen psilocybin on
habituation and prepulse inhibition of the startle reex in humans.
Behav Pharmacol 9:561566.
70. Quednow BB, Kometer M, Geyer MA, Vollenweider FX (2012):
Psilocybin-induced decits in automatic and controlled inhibition are
attenuated by ketanserin in healthy human volunteers. Neuropsycho-
pharmacology 37:630640.
71. Heekeren K, Neukirch A, Daumann J, Stoll M, Obradovic M, Kovar KA,
et al. (2007): Prepulse inhibition of the startle reex and its attentional
modulation in the human S-ketamine and N,N-dimethyltryptamine
(DMT) models of psychosis. J Psychopharmacol 21:312320.
72. Gonzalez-Maeso J, Ang RL, Yuen T, Chan P, Weisstaub NV, Lopez-
Gimenez JF, et al. (2008): Identication of a serotonin/glutamate
receptor complex implicated in psychosis. Nature 452:9397.
73. Quednow BB, Schmechtig A, Ettinger U, Petrovsky N, Collier DA,
Vollenweider FX, et al. (2009): Sensorimotor gating depends on
polymorphisms of the serotonin-2A receptor and catechol-O-methyl-
transferase, but not on neuregulin-1 Arg38Gln genotype: A replication
study. Biol Psychiatry 66:614620.
74. Kornetsky C (1957): Relation of physiological and psychological
effects of lysergic acid diethylamide. AMA Arch Neurol Psychiatry 77:
657658.
75. Dimascio A, Greenblatt M, Hyde RW (1957): A study of the effects of
L.S.D.: Physiologic and psychological changes and their interrela-
tions. Am J Psychiatry 114:309317.
76. Sokoloff L, Perlin S, Kornetsky C, Kety SS (1957): The effects of
D-lysergic acid diethylamide on cerebral circulation and over-all
metabolism. Ann N Y Acad Sci 66:468477.
77. Belleville RE, Fraser HF, Isbell H, Wikler A, Logan CR (1956): Studies
on lysergic acid diethylamide (LSD-25): I. Effects in former morphine
addicts and development of tolerance during chronic intoxication.
AMA Arch Neurol Psychiatry 76:468478.
78. Forrer GR, Goldner RD (1951): Experimental physiological studies with
lysergic acid diethylamide (LSD-25). AMA Arch Neurol Psychiatry 65:
581588.
79. Hysek CM, Simmler LD, Ineichen M, Grouzmann E, Hoener MC,
Brenneisen R, et al. (2011): The norepinephrine transporter inhibitor
reboxetine reduces stimulant effects of MDMA (ecstasy) in humans.
Clin Pharmacol Ther 90:246255.
80. Horita A, Dille JM (1954): Pyretogenic effect of lysergic acid dieth-
ylamide. Science 120:11001101.
81. Klock JC, Boerner U, Becker CE (1975): Coma, hyperthermia, and
bleeding associated with massive LSD overdose, a report of eight
cases. Clin Toxicol 8:191203.
82. Isbell H (1959): Comparison of the reactions induced by psilocybin
and LSD-25 in man. Psychopharmacologia 1:2938.
83. Gouzoulis-Mayfrank E, Thelen B, Habermeyer E, Kunert HJ, Kovar KA,
Lindenblatt H, et al. (1999): Psychopathological, neuroendocrine and
autonomic effects of 3,4-methylenedioxyethylamphetamine (MDE),
psilocybin and d-methamphetamine in healthy volunteers: Results of
an experimental double blind placebo controlled study. Psychophar-
macology 142:4150.
84. Strassman RJ, Qualls CR (1994): Dose-response study of
N,N-dimethyltryptamine in humans. I. Neuroendocrine, autonomic,
and cardiovascular effects. Arch Gen Psychiatry 51:8597.
85. Watts VJ, Lawler CP, Fox DR, Neve KA, Nichols DE, Mailman RB
(1995): LSD and structural analogs: Pharmacological evaluation at D
1
dopamine receptors. Psychopharmacology (Berl) 118:401409.
86. Giacomelli S, Palmery M, Romanelli L, Cheng CY, Silvestrini B (1998):
Lysergic acid diethylamide (LSD) is a partial agonist of D
2
dopami-
nergic receptors and it potentiates dopamine-mediated prolactin
secretion in lactotrophs in vitro. Life Sci 63:215222.
87. Meltzer HY, Fessler RG, Simonovic M, Doherty J, Fang VS (1977):
Lysergic acid diethylamide: Evidence for stimulation of pituitary
dopamine receptors. Psychopharmacology (Berl) 54:3944.
88. Sommers DK, van Wyk M, Snyman JR (1994): Dexfenuramine-
induced prolactin release as an index of central synaptosomal
5-hydroxytryptamine during treatment with uoxetine. Eur J Clin
Pharmacol 46:441444.
89. Seifritz E, Baumann P, Muller MJ, Annen O, Amey M, Hemmeter U,
et al. (1996): Neuroendocrine effects of a 20-mg citalopram infusion in
healthy males. A placebo-controlled evaluation of citalopram as 5-HT
function probe. Neuropsychopharmacology 14:253263.
90. Callaway JC, McKenna DJ, Grob CS, Brito GS, Raymon LP, Poland
RE, et al. (1999): Pharmacokinetics of Hoasca alkaloids in healthy
humans. J Ethnopharmacol 65:243256.
91. Seibert J, Hysek CM, Penno CA, Schmid Y, Kratschmar DV, Liechti
ME, et al. (2014): Acute effects of 3,4-methylenedioxymethamphet-
amine and methylphenidate on circulating steroid levels in healthy
subjects. Neuroendocrinology 100:1725.
Effects of LSD
10 Biological Psychiatry ]]], 2014; ]:]]]]]] www.sobp.org/journal
Biological
Psychiatry
... Beyond serotonergic effects, LSD influences physiological parameters through the modulation of endocrine and autonomic systems. It has been shown to significantly elevate the plasma levels of cortisol, prolactin, oxytocin, and epinephrine, resulting in increased heart rate, blood pressure, and body temperature [33]. In studies involving doses of 100 µg or greater, moderate increases in the peak heart rate (exceeding 100 bpm) were observed in approximately 20-25% of participants. ...
... -Enhanced emotional empathy, correlating with thalamic activity and changes in the default mode network (DMN). [11,12,33,35,48,49,70] Computational/ modeling studies -LSD-induced brain dynamics -Machine learning and LSD -LSD1 enzyme inhibition (cancer research) -LSD shifted brain dynamics further from equilibrium, increasing response flexibility. ...
... Physiological responses included transient elevations in heart rate, blood pressure, and hormonal levels. Importantly, no severe adverse events were recorded, reinforcing LSD's tolerability when used under clinical supervision [33]. ...
Article
Full-text available
Lysergic acid diethylamide (LSD) is gaining renewed interest as a potential treatment for anxiety, depression, and alcohol use disorder, with clinical trials reporting significant symptom reductions and long-lasting effects. LSD modulates serotonin (5-HT2A) receptors, which, in turn, influence dysfunctional brain networks involved in emotional processing and cognition. It has also shown promise in psychedelic-assisted psychotherapy, where mystical-type experiences are linked to improved psychological well-being. This review examines LSD’s pharmacokinetics, neurobiological mechanisms, and safety considerations, including cardiovascular risks, emotional vulnerability, and the potential for hallucinogen-persisting perception disorder (HPPD). Challenges such as small sample sizes, variable dosing protocols, and regulatory restrictions limit large-scale trials. Future research should focus on standardization, pharmacogenetic influences, and personalized treatment strategies to ensure its safe and effective integration into clinical practice.
... Psychedelics such as LSD and psilocybin are psychoactive compounds known to induce alterations in perception, consciousness, cognition, and mood. 9,10 Early studies in the 1950-1960s explored the clinical use of psychedelics for treating AUD, revealing promising efficacy and safety profiles. 11 A recent proof-ofconcept study and a randomized placebo-controlled trial (RCT) tested the effects of two doses of psilocybin combined with 12-14 sessions of Cognitive Behavioral Therapy and Motivational Enhancement Therapy in patients with AUD who were aiming to reduce their alcohol use rather than abstain. ...
... In fact, research has shown that psychedelics activate the stress response system (Mason et al., 2023;Reckweg et al., 2022;Szabo, Kovacs, Frecska, & Rajnavolgyi, 2014;Thompson & Szabo, 2020;Uthaug, Lancelotta, Szabo, et al., 2020), affecting various physiological functions. These include changes in blood pressure and heart rate, often characterized by modest increases during the acute phase of intoxication (Dimascio, Greenblatt, & Hyde, 1957;Hasler, Grimberg, Benz, Huber, & Vollenweider, 2004;Schmid et al., 2015). Moreover, psychedelics can impact endocrine function, particularly the release of hormones, such as cortisol (Uthaug, Lancelotta, Szabo, et al., 2020). ...
Article
Full-text available
Background Research has shown that psychedelics may have therapeutic potential in treating mental disorders like depression and anxiety. However, the mechanisms and actions underlying their effects are still not fully understood. Similarly, while the significance of mindset and setting in shaping psychedelic experiences and therapeutic outcomes is well established, information about the influence of the body is comparatively scarce. Aim This paper introduces the concept of bodyset, defined as the state of the body, including both the body and brain. We suggest it as a vital element in preparing for psychedelic experiences and beyond, broadening the traditional ‘set and setting’ framework. Methods Through an extensive literature review, we demonstrate the likely importance of the body in wellbeing, peak performance and peak experiences. Results Comprehensive multidisciplinary research, particularly focusing on various biomarkers, is needed to elucidate the potential role of bodyset in the psychedelic experience and therapy outcomes, and to guide future treatment approaches for mental health disorders. Conclusion Our exploration of the bodyset concept emphasizes the importance of considering not only psychological and environmental factors (mindset & setting), but also the physical state of the body in preparation for psychedelic experiences and psychedelic therapy. This holistic perspective may enhance our comprehension of their effects, therapeutic potential and inform the application of other treatment modalities, such as breathwork, in mental health care.
... Specifically, psychedelics have been shown to acutely modulate self-other processing and to produce acute and enduring prosocial attitudes and behaviors (Duerler et al., 2022;Preller and Vollenweider, 2019;Schmid and Bershad, 2024;Preller et al., 2018). Psychedelics acutely decrease the differentiation between self and other; increase emotional empathy, altruistic behavior, feelings of closeness and trust, and positive affective responses to social feedback; and decrease sensitivity and reactivity to social rejection, social exclusion, and social pain Dolder et al., 2016;Hysek et al., 2014;Kuypers et al., 2014;Pokorny et al., 2017;Schmid et al., 2014;Kamilar-Britt and Bedi, 2015;Preller et al., 2016;Dolder et al., 2018;Schmid et al., 2015;Frye et al., 2014). Participants in a number of studies have also reported enduring improvements in social functioning following psychedelic experiences, including improved relationships and communication and increased empathy, feelings of closeness to others, desire to interact with others and engage in social activities, and altruistic and other prosocial behavior (Griffiths et al., 2018;Griffiths et al., 2008;Griffiths et al., 2006;Griffiths et al., 2011;Noorani et al., 2018;Schmid and Liechti, 2018;Lyubomirsky, 2022;Barone et al., 2019;Kirkpatrick et al., 2014;Elmer et al., 2024). ...
Article
Full-text available
Studies of psychedelic use in naturalistic and clinical settings have suggested safety and mental health benefits for adults with histories of childhood maltreatment. Acute psychological mechanisms that predict therapeutic benefits in this population, however, have yet to be determined. Two common group settings of naturalistic psychedelic use – organized ceremonies and raves or other electronic dance music events – might facilitate therapeutic psychedelic effects because of the unique psychosocial environments they comprise. This prospective, longitudinal study sought to investigate 2 primary questions: first, whether adults with maltreatment histories planning to use psychedelic drugs with therapeutic intent at ceremonies or raves would see enduring psychological benefits after their experiences; and second, whether subjective dimensions of the acute psychedelic experience would be associated with lasting psychological benefits. Eighty-five participants completed self-report measures in the month before, within 2 days after, and approximately 2 months after a planned psychedelic experience with therapeutic intent at a ceremony or rave assessing childhood maltreatment history; trauma symptoms, internalized (trait) shame, and connectedness at baseline and follow-up; and various dimensions of the acute subjective psychedelic experience. Mean scores in posttraumatic stress disorder (PTSD) symptoms, complex PTSD symptoms, trait shame, social connectedness, and general connectedness significantly improved from baseline to 2-month follow-up (ds = 0.73-1.12). Longitudinal changes in outcomes significantly correlated with acute subjective effects of the psychedelic experience. These findings have implications regarding both the potential clinical benefit of psychedelic use among adults with childhood maltreatment histories as well as the psychological mechanisms of therapeutic action of psychedelics.
... Psychedelics such as LSD and psilocybin are psychoactive compounds known to induce alterations in perception, consciousness, cognition, and mood. 9,10 Early studies in the 1950-1960s explored the clinical use of psychedelics for treating AUD, revealing promising efficacy and safety profiles. 11 A recent proof-ofconcept study and a randomized placebo-controlled trial (RCT) tested the effects of two doses of psilocybin combined with 12-14 sessions of Cognitive Behavioral Therapy and Motivational Enhancement Therapy in patients with AUD who were aiming to reduce their alcohol use rather than abstain. ...
Article
Background Despite the promising therapeutic effects of psilocybin, its efficacy in preventing relapse after withdrawal treatment for alcohol use disorder (AUD) remains unknown. This study aims to assess whether a single dose of psilocybin combined with brief psychotherapy could reduce relapse rates and alcohol use in AUD patients. Methods This single-center, double-blind, randomized clinical trial was conducted in Switzerland. We recruited participants with AUD between June 8, 2020, and August 16, 2023 who completed withdrawal treatment within six weeks prior to enrollment. Participants were randomized (1:1) to receive either a single oral dose of psilocybin (25 mg) or placebo (mannitol), combined with brief psychotherapy. The primary outcomes were abstinence and mean alcohol use at 4-week follow-up. Participants completed the timeline followback to assess daily alcohol use. The trial is registered on ClinicalTrials.gov (NCT04141501). Findings We included 37 participants who completed the 4-week follow-up (female:male = 14:23; psilocybin = 18, placebo = 19) in the analysis. There were no significant differences between groups in abstinence duration (p = 0.55, psilocybin mean = 16.80 days, 95% CI: 14.31–19.29; placebo mean = 13.80 days, 95% CI: 10.97–16.63; Cohen’s d = 0.151) or mean alcohol use per day (p = 0.51, psilocybin: median = 0.48 standard alcohol units, range: 0–3.99, placebo: median = 0.54 standard alcohol units, range: 0–5.96; Cohen’s d = 0.11) at 4-week or 6-month follow-up (abstinence: Cohen’s d = 0.10, alcohol use: Cohen’s d = 0.075). Participants in both groups reported reduced craving and temptation to drink alcohol after the dosing visit, with an additional reduction observed in the psilocybin group. Thirteen adverse events occurred in the psilocybin and seven in the placebo group. One serious adverse event occurred in the psilocybin and four in the placebo group, all related to inpatient withdrawal treatments. Interpretation A single dose of psilocybin combined with five psychotherapy sessions may not be sufficient to reduce relapse rates and alcohol use in severely affected AUD patients following withdrawal treatment. However, given the limited sample size of our study, larger trials are needed in the future to confirm these findings. Funding 10.13039/501100001711Swiss National Science Foundation under the framework of Neuron Cofund, Swiss Neuromatrix Foundation, and Heffter Young Investigator Fellowship Award.
... Switzerland is widely recognized as a pioneer in psychedelic research and therapy. Since 2014 until end of 2024, the Federal Office of Public Health (FOPH) has granted over 1795 exemptions for the limited medical use of LSD, MDMA, and psilocybin ( [2,3,57], personal communication FOPH, January 2025). This progressive stance has made the issue of specialized training particularly urgent and relevant in Switzerland. ...
Article
Full-text available
The growing interest in psychedelic-assisted therapy (PAT) for treating psychiatric disorders such as treatment-resistant depression, PTSD, and anxiety has led to an increasing demand for specialized training. In Switzerland, MDMA, psilocybin, and LSD are applied in the framework of limited medical use as exceptional treatment options since 2014. The Swiss Medical Association for Psychedelic Therapy (SÄPT) has been a key player in addressing the need for education, offering a comprehensive, three-year training program for physicians and psychologists. This curriculum integrates theoretical knowledge with hands-on experience, emphasizing the therapeutic relationship, ethical considerations, and the management of altered states of consciousness induced by psychedelics. This article gives an overview of the structure and framework of the training and addresses topics covered by the program through theoretical teaching and retreats focusing on practical learning. However, the demand for these programs far exceeds supply. This gap is expected to widen as psychedelics potentially become regulated prescription medications. In response, several organizations have expanded their educational offerings, including further education trainings, workshops, conferences, and symposia. Overall, there is a need for more comprehensive and accessible training programs to meet the growing demand. The evolving landscape of psychedelic research, regulatory changes, and diverse patient populations require flexible and adaptive training models. As the field progresses, it is essential to establish certification standards and ensure the continued quality of training programs to ensure the safe and effective use of PAT in clinical trials and practice.
Article
The therapeutic use of psychedelics for mental health issues holds considerable promise. However, systematic assessment of adverse events associated with these substances has received relatively little attention. Here, we discuss several considerations concerning the assessment of adverse events in psychedelic-assisted therapies. We discuss the preference for using the term “adverse effects” over “side effects”, as well as the ongoing debate regarding which substances are classified as psychedelic. We also provide recommendations on when and how to assess adverse effects, for example the importance to study them in any kind of therapy involving psychedelics, and using comprehensive monitoring of a wide range of physical parameters in combination with behavioral outcomes and the individual’s experience, at baseline and throughout the study. Also, sex-specific differences should be considered. Furthermore, we highlight several significant studies that have addressed these aspects. In summary, psychedelics offer great promise as a potential treatment (add-on) option in psychiatry, but more rigorous assessment of adverse effects is needed to promote safe use and implementation in clinical practice.
Article
Background Migraine and cluster headache are two primary headache disorders for which conventional treatments are limited. Classic psychedelic substances such as lysergic acid diethylamide (LSD) and psilocybin are potentially promising new treatment candidates for these conditions. Aims The aim of the present study was to investigate the possible relationship between the lifetime use of classic psychedelics and frequent bad headaches in a large British cohort sample. Methods Using data ( N = 11,419) collected in 1999–2000 as part of the 1958 British National Child Development Study, this cross-sectional study used multiple logistic regression, controlling for a range of potential confounders, to test the hypothesis that lifetime use of classic psychedelics would be associated with lower odds of having frequent bad headaches. Results Lifetime use of classic psychedelics was associated with 25% lower odds of having frequent bad headaches (adjusted odds ratio = 0.75, 95% CI: 0.59–0.95, p = 0.016). Conclusions The results of the present study add to the literature suggesting classic psychedelics as a possible future prophylactic treatment option for primary headache disorders.
Article
Full-text available
Rationale: Lysergic acid diethylamide (LSD) has a history of use as a psychotherapeutic aid in the treatment of mood disorders and addiction, and it was also explored as an enhancer of mind control. Objectives: The present study sought to test the effect of LSD on suggestibility in a modern research study. Methods: Ten healthy volunteers were administered with intravenous (i.v.) LSD (40-80 μg) in a within-subject placebo-controlled design. Suggestibility and cued mental imagery were assessed using the Creative Imagination Scale (CIS) and a mental imagery test (MIT). CIS and MIT items were split into two versions (A and B), balanced for 'efficacy' (i.e. A ≈ B) and counterbalanced across conditions (i.e. 50 % completed version 'A' under LSD). The MIT and CIS were issued 110 and 140 min, respectively, post-infusion, corresponding with the peak drug effects. Results: Volunteers gave significantly higher ratings for the CIS (p = 0.018), but not the MIT (p = 0.11), after LSD than placebo. The magnitude of suggestibility enhancement under LSD was positively correlated with trait conscientiousness measured at baseline (p = 0.0005). Conclusions: These results imply that the influence of suggestion is enhanced by LSD. Enhanced suggestibility under LSD may have implications for its use as an adjunct to psychotherapy, where suggestibility plays a major role. That cued imagery was unaffected by LSD implies that suggestions must be of a sufficient duration and level of detail to be enhanced by the drug. The results also imply that individuals with high trait conscientiousness are especially sensitive to the suggestibility-enhancing effects of LSD.
Article
Full-text available
Social cognition is important in everyday-life social interactions. The social cognitive effects of 3,4-methylenedioxymethamphetamine (MDMA, 'ecstasy') and methylphenidate (both used for neuroenhancement and as party drugs) are largely unknown. We investigated the acute effects of MDMA (75 mg), methylphenidate (40 mg) and placebo using the Facial Emotion Recognition Task, Multifaceted Empathy Test, Movie for the Assessment of Social Cognition, Social Value Orientation Test and the Moral Judgment Task in a cross-over study in 30 healthy subjects. Additionally, subjective, autonomic, pharmacokinetic, endocrine and adverse drug effects were measured. MDMA enhanced emotional empathy for positive emotionally charged situations in the MET and tended to reduce the recognition of sad faces in the Facial Emotion Recognition Task. MDMA had no effects on cognitive empathy in the Multifaceted Empathy Test or social cognitive inferences in the Movie for the Assessment of Social Cognition. MDMA produced subjective 'empathogenic' effects, such as drug liking, closeness to others, openness and trust. In contrast, methylphenidate lacked such subjective effects and did not alter emotional processing, empathy or mental perspective-taking. MDMA but not methylphenidate increased the plasma levels of oxytocin and prolactin. None of the drugs influenced moral judgment. Effects on emotion recognition and emotional empathy were evident at a low dose of MDMA and likely contribute to the popularity of the drug.
Article
Full-text available
3,4-Methylenedioxymethamphetamine (MDMA, 'ecstasy') and methylphenidate are widely used psychoactive substances. MDMA primarily enhances serotonergic neurotransmission, and methylphenidate increases dopamine but has no serotonergic effects. Both drugs also increase norepinephrine, resulting in sympathomimetic properties. Here we studied the effects of MDMA and methylphenidate on 24-h plasma steroid profiles. Sixteen healthy subjects (eight men, eight women) were treated with single doses of MDMA (125 mg), methylphenidate (60 mg), MDMA + methylphenidate, and placebo on four separate days using a cross-over study design. Cortisol, cortisone, corticosterone, 11-dehydrocorticosterone, aldosterone, 11-deoxycorticosterone, dehydroepiandrosterone (DHEA), dehydroepiandrosterone sulfate (DHEAS), androstendione, and testosterone were repeatedly measured up to 24-h using liquid-chromatography tandem mass-spectroscopy. MDMA significantly increased the plasma concentrations of cortisol, corticosterone, 11-dehydrocorticosterone, and 11-deoxycorticosterone and also tended to moderately increase aldosterone levels compared with placebo. MDMA also increased the sum of cortisol + cortisone and the cortisol/cortisone ratio, consistent with an increase in glucocorticoid production. MDMA did not alter the levels of cortisone, DHEA, DHEAS, androstendione, or testosterone. Methylphenidate did not affect any of the steroid concentrations, and it did not change the effects of MDMA on circulating steroids. In summary, the serotonin releaser MDMA has acute effects on circulating steroids. These effects are not observed after stimulation of the dopamine and norepinephrine systems with methylphenidate. The present findings support the view that serotonin rather than dopamine and norepinephrine mediates the acute pharmacologically-induced stimulation of the hypothalamic-pituitary-adrenal axis in the absence of other stressors. © 2014 S. Karger AG, Basel.
Article
Full-text available
A double-blind, randomized, active placebo-controlled pilot study was conducted to examine safety and efficacy of lysergic acid diethylamide (LSD)-assisted psychotherapy in 12 patients with anxiety associated with life-threatening diseases. Treatment included drug-free psychotherapy sessions supplemented by two LSD-assisted psychotherapy sessions 2 to 3 weeks apart. The participants received either 200 μg of LSD (n = 8) or 20 μg of LSD with an open-label crossover to 200 μg of LSD after the initial blinded treatment was unmasked (n = 4). At the 2-month follow-up, positive trends were found via the State-Trait Anxiety Inventory (STAI) in reductions in trait anxiety (p = 0.033) with an effect size of 1.1, and state anxiety was significantly reduced (p = 0.021) with an effect size of 1.2, with no acute or chronic adverse effects persisting beyond 1 day after treatment or treatment-related serious adverse events. STAI reductions were sustained for 12 months. These results indicate that when administered safely in a methodologically rigorous medically supervised psychotherapeutic setting, LSD can reduce anxiety, suggesting that larger controlled studies are warranted.This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives 3.0 License, where it is permissible to download and share the work provided it is properly cited. The work cannot be changed in any way or used commercially.
Article
Full-text available
Methylphenidate and 3,4-methylenedioxymethamphetamine (MDMA, 'ecstasy') are widely misused psychoactive drugs. Methylphenidate increases brain dopamine and norepinephrine levels by blocking the presynaptic reuptake transporters. MDMA releases serotonin, dopamine and norepinephrine through the same transporters. Pharmacodynamic interactions of methylphenidate and MDMA are likely. This study compared the pharmacodynamic and pharmacokinetic effects of methylphenidate and MDMA administered alone or in combination in healthy subjects using a double-blind, placebo-controlled, crossover design. Methylphenidate did not enhance the psychotropic effects of MDMA, although it produced psychostimulant effects on its own. The haemodynamic and adverse effects of co-administration of methylphenidate and MDMA were significantly higher compared with MDMA or methylphenidate alone. Methylphenidate did not change the pharmacokinetics of MDMA and vice versa. Methylphenidate and MDMA shared some subjective amphetamine-type effects; however, 125 mg of MDMA increased positive mood more than 60 mg of methylphenidate, and methylphenidate enhanced activity and concentration more than MDMA. Methylphenidate and MDMA differentially altered facial emotion recognition. Methylphenidate enhanced the recognition of sad and fearful faces, whereas MDMA reduced the recognition of negative emotions. Additionally, the present study found acute pharmacodynamic tolerance to MDMA but not methylphenidate. In conclusion, the combined use of methylphenidate and MDMA does not produce more psychoactive effects compared with either drug alone, but potentially enhances cardiovascular and adverse effects. The findings may be of clinical importance for assessing the risks of combined psychostimulant misuse. Trial registration identification number: NCT01465685 (http://clinicaltrials.gov/ct2/show/NCT01465685).
Article
Full-text available
3,4-Methylenedioxymethamphetamine (MDMA, "ecstasy") releases serotonin and norepinephrine. MDMA is reported to produce empathogenic and prosocial feelings. It is unknown whether MDMA in fact alters empathic concern and prosocial behavior. We investigated the acute effects of MDMA using the Multifaceted Empathy Test (MET), dynamic Face Emotion Recognition Task (FERT), and Social Value Orientation (SVO) test. We also assessed effects of MDMA on plasma levels of hormones involved in social behavior using a placebo-controlled, double-blind, random-order, cross-over design in 32 healthy volunteers (16 women). MDMA enhanced explicit and implicit emotional empathy in the MET and increased prosocial behavior in the SVO test in men. MDMA did not alter cognitive empathy in the MET but impaired the identification of negative emotions, including fearful, angry, and sad faces, in the FERT, particularly in women. MDMA increased plasma levels of cortisol and prolactin, which are markers of serotonergic and noradrenergic activity, and of oxytocin, which has been associated with prosocial behavior. In summary, MDMA sex-specifically altered the recognition of emotions, emotional empathy, and prosociality. These effects likely enhance sociability when MDMA is used recreationally and may be useful when MDMA is administered in conjunction with psychotherapy in patients with social dysfunction or posttraumatic stress disorder.
Article
Full-text available
One of the oldest models of schizophrenia is based on the effects of serotonergic hallucinogens such as mescaline, psilocybin, and (+)-lysergic acid diethylamide (LSD), which act through the serotonin 5-HT2A receptor. These compounds produce a 'model psychosis' in normal individuals that resembles at least some of the positive symptoms of schizophrenia. Based on these similarities, and because evidence has emerged that the serotonergic system plays a role in the pathogenesis of schizophrenia in some patients, animal models relevant to schizophrenia have been developed based on hallucinogen effects. Here we review the behavioural effects of hallucinogens in four of those models, the receptor and neurochemical mechanisms for the effects and their translational relevance. Despite the difficulty of modelling hallucinogen effects in nonverbal species, animal models of schizophrenia based on hallucinogens have yielded important insights into the linkage between 5-HT and schizophrenia and have helped to identify receptor targets and interactions that could be exploited in the development of new therapeutic agents.
Article
Schizophrenic patients exhibit impairments in both sensorimotor gating and habituation in a number of paradigms. Through human and animal model research, these fundamental cognitive deficits have well-described neurobiologic bases and offer insights into the neuroanatomic and neurotransmitter abnormalities that characterize patients with schizophrenic spectrum disorders. In this context, the startle response is particularly interesting, because it is a cross-species response to strong stimuli that is plastic or alterable using experimental and neurobiologic manipulations. Thirty-nine medicated schizophrenic patients and 37 normal control subjects were studied in a new electromyography based startle response paradigm in which both prepulse inhibition (an operational measure of sensorimotor gating) and habituation (the normal decrease in response magnitude to repeated stimuli over time) can be separated and assessed in one test session. The results indicate that schizophrenic patients have extensive deficits in both intramodal and cross-modal sensorimotor gating and a trend to show acoustic startle habituation deficits. The deficit in prepulse inhibition of startle amplitude exhibited by schizophrenic patients was evident when an acoustic prepulse stimulus preceded either an acoustic or a tactile startle stimulus. No deficit was observed in the prepulse-induced facilitation of startle latencies, indicating that the failure of gating was not due to a failure of stimulus detection. These findings suggest centrally mediated deficits in sensorimotor gating in schizophrenic patients.
Article
Psychedelic drugs fell from grace in the 1960s. Now, scientists are rediscovering them as potential treatments for a range of illnesses.