PreprintPDF Available

Preliminary safety and effectiveness of psilocybin-assisted therapy in adults with fibromyalgia: An open-label, pilot clinical trial

Authors:
Preprints and early-stage research may not have been peer reviewed yet.

Abstract and Figures

Fibromyalgia (FM) is the prototypical nociplastic pain condition, characterized by widespread pain and issues with cognition, mood, and sleep. Currently, there are limited treatment options available that effectively treat FM symptoms. Psilocybin-assisted therapy (PAT) is an emerging combined drug-therapy intervention, but no studies to-date have investigated PAT for FM. Here, we report findings from an open-label, pilot trial of PAT for FM (N=5; NCT05128162). In conjunction with psychotherapy (two preparatory, four integration sessions), participants received two doses of oral psilocybin (15mg and 25mg) delivered two weeks apart. Regarding safety (primary outcome), there were transient elevations of blood pressure or heart rate during dosing which normalized by the end of treatment, with no serious adverse events. Four of five participants reported transient headaches following dosing. Compared to baseline, participants reported clinically meaningful improvements in the following secondary outcomes one month following their second psilocybin dose (reported as Cohen’s d): pain severity (d=-2.1, 95% CI[-3.7 to -0.49]), pain interference (d=-1.8, 95% CI [-3.27 to -0.24]), and sleep disturbance (d=-2.5, 95% CI [-4.21 to -0.75]). Using the Patient Global Impression of Change, one participant reported their symptoms “very much improved,” two reported “much improved,” and two reported “minimally improved.” Compared to baseline, there were improvements in the following exploratory outcomes after the intervention: FM symptoms, anxiety, and fatigue. This small open-label trial preliminarily supports that PAT is well-tolerated by people with FM, establishing a basis for larger randomized controlled trials.
Content may be subject to copyright.
1
Preliminary safety and effectiveness of psilocybin-assisted therapy in adults with
1
fibromyalgia: An open-label, pilot clinical trial
2
3
Jacob S. Aday1,2, Jenna McAfee1, Deirdre A. Conroy3, Avinash Hosanagar3,4, Vijay Tarnal1,2,
4
Cody Weston4, Katharine Scott1, Dana Horowitz5, Steven E. Harte1,2,3, Niloufar Pouyan1,2,3,
5
Nicolas G. Glynos1,2, Anne K. Baker1,2, Jeffrey Guss6, Alan K. Davis7,8,9, Helen J. Burgess3,
6
George A. Mashour1,2,3, Daniel J. Clauw1, Kevin F. Boehnke*1,2
7
8
1. Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI,
9
USA
10
2. Michigan Psychedelic Center, University of Michigan, Ann Arbor, MI, USA
11
3. Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA.
12
4. Psychiatry Department, University of Michigan Medical School, Ann Arbor, MI, USA
13
5. Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
14
6. Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
15
7. Center for Psychedelic Drug Research and Education, College of Social Work, The Ohio
16
State University, Columbus, OH, USA
17
8. Comprehensive Cancer Center, Department of Internal Medicine, Division of Medical
18
Oncology, The Ohio State University, Columbus, OH USA
19
9. Center for Psychedelic and Consciousness Research, Department of Psychiatry and
20
Behavioral Sciences, Johns Hopkins University, Baltimore, MD USA
21
22
*Corresponding author: Kevin Boehnke, 24 Frank Lloyd Wright Drive, Ann Arbor, MI, 48106.
23
Email: kboehnke@med.umich.edu Phone: 734-998-6939
24
25
2
Abstract
26
Fibromyalgia (FM) is the prototypical nociplastic pain condition, characterized by widespread
27
pain and issues with cognition, mood, and sleep. Currently, there are limited treatment options
28
available that effectively treat FM symptoms. Psilocybin-assisted therapy (PAT) is an emerging
29
combined drug-therapy intervention, but no studies to-date have investigated PAT for FM. Here,
30
we report findings from an open-label, pilot trial of PAT for FM (N=5; NCT05128162). In
31
conjunction with psychotherapy (two preparatory, four integration sessions), participants
32
received two doses of oral psilocybin (15mg and 25mg) delivered two weeks apart. Regarding
33
safety (primary outcome), there were transient elevations of blood pressure or heart rate during
34
dosing which normalized by the end of treatment, with no serious adverse events. Four of five
35
participants reported transient headaches following dosing. Compared to baseline, participants
36
reported clinically meaningful improvements in the following secondary outcomes one month
37
following their second psilocybin dose (reported as Cohen’s d): pain severity (d=-2.1, 95% CI[-
38
3.7 to -0.49]), pain interference (d=-1.8, 95% CI [-3.27 to -0.24]), and sleep disturbance (d=-2.5,
39
95% CI [-4.21 to -0.75]). Using the Patient Global Impression of Change, one participant
40
reported their symptoms “very much improved,” two reported “much improved,” and two
41
reported “minimally improved.” Compared to baseline, there were improvements in the
42
following exploratory outcomes after the intervention: FM symptoms, anxiety, and fatigue. This
43
small open-label trial preliminarily supports that PAT is well-tolerated by people with FM,
44
establishing a basis for larger randomized controlled trials.
45
46
3
1. Introduction
47
Fibromyalgia (FM) is a common chronic pain condition that affects 24% of the general
48
population[1]. FM is currently understood as the prototypical nociplastic pain condition, i.e., a
49
disorder of pain regulation via altered centralized pain processing[2]. Common FM symptoms
50
include widespread pain, poor sleep quality, fatigue, and cognitive difficulties[3], and many
51
individuals with FM have a history of trauma and/or comorbid psychiatric conditions[4]. The
52
heterogeneous nature of FM manifests clinically as varied phenotypic expressions, which makes
53
many treatment options, such as pharmacotherapy and physical interventions, only minimally
54
effective[5]. This leads to considerable divergence in patient outcomes and results in significant
55
economic and personal burden[6]. Given the mixed success of existing treatments, there is a
56
pressing need to develop tailored and more effective therapeutic approaches[7].
57
58
One potential treatment for FM is psilocybin-assisted therapy (PAT)[8], a treatment paradigm
59
that combines psychotherapy with administration of psilocybin[9]. Psilocybin is a serotonergic
60
compound found in numerous mushroom species that causes substantial alterations in cognition,
61
mood, affect, and sensory experience[10]. Human studies with psilocybin indicate altered
62
functional connectivity of neuronal networks associated with clinical benefits[11]. Speculatively,
63
the subsequent changes in functional connectivity by psilocybin might also facilitate changes
64
associated with clinical benefits in conditions characterized by nociplastic pain, but this remains
65
untested. Additionally, psychedelic use and PAT have been linked with increased psychological
66
flexibility[12, 13], and it hypothesized that insights gained during PAT therapy sessions may be
67
more likely to “stick” and facilitate lasting psychological and behavioral change, particularly in
68
disorders characterized by psychological rigidity[14-16]. Indeed, recent studies of PAT have
69
demonstrated promising findings for treatment of major and treatment-resistant depression[17-
70
4
20], anxiety[19-22], end-of-life distress[23, 24], obsessive compulsive disorder[25], and
71
substance use disorders[26, 27].
72
73
Preliminary clinical trials and case studies suggest that psilocybin may produce analgesic effects
74
in intractable phantom limb pain[28], cluster headache[29], and migraine[30]. This has been
75
complemented by recent preclinical research demonstrating that single-dose psilocybin reduced
76
mechanical hypersensitivity in rats for at least 28 days[31]. Similarly, surveys show that many
77
individuals using psilocybin and other psychedelics for chronic pain report substantial pain
78
relief[32-34]. In a North American survey study of participants with fibromyalgia[35],
79
perceptions of benefit from psychedelic use were generally neutral (59.4%) or positive (36.8%),
80
with less than 3% reporting negative impacts on overall health or pain symptoms. Notably, of the
81
12 participants in that study who reported using psychedelics with the specific intention of
82
treating chronic pain, 11 reported improved symptoms.
83
84
In summary, converging lines of research support the mechanistic potential of PAT in the
85
treatment of FM. However, clinical studies assessing the safety and tolerability of psilocybin and
86
associated therapy in people with FM are limited. Thus, in the current pilot study, we evaluated
87
the preliminary safety and effectiveness of PAT in the treatment of FM using an open-label
88
design among five individuals.
89
90
2. Methods
91
This open-label clinical trial was conducted at the University of Michigan Chronic Pain and
92
Fatigue Center in Ann Arbor, Michigan. All study procedures were approved by the University
93
5
of Michigan Medical School Institutional Review Board under protocol HUM00208367. The
94
study was prospectively registered on clinicaltrials.gov under identifier NCT05128162. The study
95
protocol is available in supplement 1. In addition to the study procedures outlined below,
96
participants also completed phenotyping visits consisting of sensory testing and magnetic
97
resonance imaging, the results of which will be reported separately.
98
99
2.1 Study Design and Participants
100
Participants in this trial were adults aged 25-64 who did not smoke tobacco and had a diagnosis
101
of FM or who had reported FM symptoms for the past year. At screening, all participants also
102
met the 2016 FM survey criteria for fibromyalgia[36]. Exclusion criteria included being pregnant
103
or nursing, cardiovascular condition in the past year (e.g., coronary artery disease, stroke, angina,
104
uncontrolled hypertension, transient ischemic attack), epilepsy, insulin-dependent diabetes,
105
active autoimmune disease, clinically significant laboratory abnormalities per a complete blood
106
count and metabolic panel, past year or current substance use disorder (other than caffeine), past
107
or current history of having a psychotic disorder or bipolar I or II disorder. All screened subjects’
108
(n=17) medical records were reviewed by study physicians and those found to be eligible after
109
medical record review met with study physicians to ensure they did not meet any exclusionary
110
medical criteria. Participants with scores representing severe depression on the Patient Health
111
Questionnaire 8 were also excluded[37]. Exclusionary medications included monoamine oxidase
112
inhibitors, psychoactive prescription medications (e.g., benzodiazepines, opioids) more than
113
twice per week, prohibited drugs of abuse including illicit opioids, cocaine, methamphetamines,
114
3,4-Methylenedioxymethamphetamine, and any use of hallucinogens in the past 6 months or
115
more than 10 lifetime hallucinogen uses. Other prohibited medications included antihypertensive
116
6
medications, UGT1A9 or 1A10 inhibitors (e.g., regorafenib, rifampicin, deferasiroxor, ginseng)
117
and aldehyde or alcohol dehydrogenase inhibitor (e.g,, disulfiram). Initially, participants taking
118
antidepressant medications of any variety were excluded, but following recent studies showing
119
the safety of concomitant antidepressant use during PAT[38], we amended our protocol to
120
include individuals taking selective serotonin reuptake inhibitors (SSRIs) and selective
121
norepinephrine reuptake inhibitors (SNRIs) as well as less than 300 mg/day of bupropion. A full
122
description of eligibility criteria are included in the study protocol. Participants were recruited
123
through flyers posted in University of Michigan clinics, UMhealthresearch.org, and electronic
124
health record searches with IRB permission between September 2023 and April 2024.
125
126
Figure 1. Overall study design.
127
2.2 Description of therapy
128
Therapy visits occurred at the Sleep and Circadian Research Laboratory and via Zoom (certain
129
therapy visits only). The study therapy manual was generally consistent with the Yale Manual
130
for Psilocybin-Assisted Therapy of Depression[39]. However, the manual was modified for
131
fibromyalgia with input from Fluence, a continuing education organization in the emerging field
132
7
of PAT, as well as consultation with experts in delivering PAT (co-authors AKD and JG). Each
133
participant was paired with a consistent therapist dyad throughout the study, consisting of a lead
134
therapist and co-therapist. The lead therapists were doctoral level psychotherapists (JM and
135
DAC) and the co-therapist had a Masters of Social Work (DH).
136
137
As is standard in PAT research, the therapy component included preparatory, dosing, and
138
integration sessions with the therapist dyad[40]. During the preparatory sessions, the therapists a)
139
developed a trusting therapeutic rapport with participants, b) gathered information about lived
140
experience with FM, c) provided education regarding psychedelic experiences, d) described the
141
therapeutic approach to be used, and e) explored participants’ expectations for treatment.
142
Therapists explained the logistics of the dosing sessions (e.g., dosing day procedures, session
143
duration, music, use of eyeshades), delineated behavioral boundaries of interaction between the
144
participant and the therapists, and discussed safety measures. In dosing sessions, therapists
145
maintained an attentive but non-intrusive presence, encouraging participants to focus inward on
146
their thoughts, emotions, and body sensations, including engaging with difficult content that
147
arose. They also assisted the participant by meeting any immediate needs for comfort or safety.
148
Toward the end of the dosing sessions, participants were invited to talk about their experiences in
149
the session and the emotions that were evoked. Therapists focused on eliciting descriptions of
150
phenomenology, rather than interpreting or guiding this report. The integration phase began the
151
day after the dosing sessions and involved thoroughly reviewing the participant’s experience
152
during the dosing session and, in some cases, reinforcing aspects of the experience to foster
153
changes in thought and behavior. Therapists accomplished this by asking open-ended questions
154
about the session, intended to elicit introspective, interpersonal, spiritual, or noetic insights that
155
8
occurred during the session that the participant may otherwise forget or have difficulty
156
verbalizing. Therapists supported the participants’ narrative expression of their experience and
157
emphasized this over interpretive interventions.
158
159
2.3 Study drug
160
Study drug was provided as psilocybin in 15 mg and 25 mg gel capsules synthesized by the
161
analytical chemistry lab at the Usona Institute. Participants received two doses of psilocybin
162
(15mg followed by 25mg) approximately two weeks apart. Sequential dosing was chosen in
163
alignment with previous studies[18, 41], as well as to allow participants to acclimate to the
164
pharmacodynamic effects of psilocybin in the first dose and thus to prepare for the more intense
165
effects in the second dosing session. Participants could decide to not increase their dose to 25mg
166
at the second dosing session, and one participant (001) chose not to do so. 25mg was selected as
167
the high dose based on research that demonstrates tight dose-response curves across studies of
168
various indications, which are supplemented by findings that body mass index[42] and
169
weight[43] are not significantly related to drug effects.
170
171
2.4 Dosing days
172
For 7 days prior to dosing sessions, participants agreed to abstain from taking nonprescription
173
medications, nutritional supplements, or herbal supplements except when approved by the study
174
team. Similarly, they agreed to abstain from alcoholic beverages and psychoactive drugs 24
175
hours before and after each dosing session, and sildenafil within 72 hours before or after dosing
176
days. On the day of drug administration, participants were asked to eat a low-fat breakfast before
177
reporting to the laboratory. Upon arrival, participants provided urine samples to test for
178
9
exclusionary drugs and pregnancy and completed breath alcohol and COVID-19 tests. Study
179
therapists also underwent COVID-19 testing on dosing days. If all tests were negative,
180
participants met with the study team for a brief interview to discuss if the session was
181
contraindicated. After review of their vital signs and clearance from the study physician,
182
participants then proceeded to a comfortable dosing suite with the same therapists that were
183
present during preparatory sessions. The dosing suite included a living room-like space with a
184
couch for the participant and two armchairs for the therapists as well as an adjacent bedroom
185
where the participants could lie down if they preferred and a bathroom. During these sessions,
186
participants were instructed to lie on the couch or bed, wear eyeshades, and listen to music
187
through headphones or a headband, all of which are meant to enhance and encourage internal
188
attention and reflection. The same playlist, created by researchers at The Ohio State University,
189
was used for every dosing session. Vitals (i.e., heart rate and blood pressure) were intermittently
190
monitored throughout the 8-hour dosing sessions (30, 60, 90, 120, 180, 240, 300, and 360
191
minutes after administration). During the same period as the vital signs measurements, therapists
192
also completed a monitor rating form, which included questions about the presence/intensity of
193
behaviors, signs, and reported symptoms, such as peacefulness, yawning, nausea/vomiting,
194
quantity of speech, anxiety, sleepiness, crying, restlessness, visual changes, euphoria, and
195
feelings of unreality. Study physicians examined the participant if any concerns were raised by
196
study therapists or if there was a concern for elevated vitals. Dosing sessions were video and
197
audio recorded. After the study drug effects subsided, participants completed questionnaires that
198
assessed the subjective experiences of the dosing session (e.g., Mystical Experience
199
Questionnaire, Challenging Experiences Questionnaire, Psychological Insight Questionnaire),
200
and the therapists completed questionnaires assessing participant mood and safety. Participants
201
10
were medically cleared by the study team (including a study physician) and were discharged
202
home with a responsible adult. Participants were asked to process the experience at home by
203
writing a reflection about their experiences during the dosing session and this reflection was
204
discussed during follow-up integration sessions.
205
206
2.5 Measures
207
Primary outcome
208
The primary outcome for this study was safety. This was assessed acutely during dosing sessions
209
via heart rate per minute and blood pressure, as well as globally via adverse event capture from
210
Days 1 (baseline) through 64 (end of treatment). Blood pressure greater than 200 systolic or
211
greater than 110 diastolic for more than 15 minutes was considered to be an adverse event. For
212
heart rate, our goal was to maintain a target heart rate within 20% of baseline. An elevated heart
213
rate was considered clinically significant if it was accompanied by cardiovascular symptoms and
214
an increase in blood pressure (exceeding 20% of baseline) that persisted for over 15 minutes.
215
216
Secondary outcomes
217
Aggregate Worst Pain Score Change
218
We assessed participants’ worst pain intensity daily using a 0-10 (0 = no pain, 10 = worst pain
219
imaginable) numeric rating scale via a daily Qualtrics survey. We compared the average of pain
220
scores from Days 1 to 7 (prior to preparation therapy) to the average of pain scores from the 7-
221
day window (Days 57 to 63) immediately prior to the end of treatment visit (Day 64).
222
Pain Interference
223
Pain interference is the degree to which pain affects important aspects of an individual’s life,
224
11
such as social, cognitive, and physical activities. We assessed pain interference using the 4‑item
225
PROMIS pain interference scale from the PROMIS‐29+2 Profile v2.1 (PROPr)[44, 45]. This
226
uses a 5-point Likert scale to generate raw scores. We then used the HealthMeasures Scoring
227
Service to generate standardized T-scores, which range from 0-100 (median = 50, which
228
represents the general population mean, standard deviation = 10). Higher scores indicate worse
229
pain interference.
230
Sleep disturbance
231
Sleep disturbance includes assessment of sleep quality, perceived ability to fall and stay asleep,
232
satisfaction of sleep, and depth of sleep. We assessed sleep disturbance using the PROMIS Sleep
233
Disturbance Short Form 8b. Scoring was performed the same way as for pain interference, with
234
standardized t-scores ranging from 0-100 (median = 50, which represents the general population
235
mean, standard deviation = 10). Higher scores indicate worse sleep disturbance.
236
Chronic Pain Acceptance
237
The Chronic Pain Acceptance Questionnaire (8 item version) is a validated measure that assesses
238
activity engagement and pain willingness, e.g., recognizing that trying to avoid or control pain
239
may be maladaptive for chronic pain[46]. Participants rate items on a 0 to 6 scale, with 0 being
240
“never true” and 6 being “always true”. Higher scores indicate higher acceptance of chronic pain.
241
Patient Global Impression of Change (PGIC)
242
We assessed participant impressions of how PAT affected their global functioning using the
243
PGIC, a one-item questionnaire that uses a 7-point Likert scale ranging from 1 (“very much
244
improved”) to 7 (“very much worse”)[47-49].
245
12
246
Exploratory Outcomes
247
Challenging Experiences Questionnaire
248
The Challenging Experiences Questionnaire is a 26-item self-report survey that was used to
249
assess the extent to which participants endorsed having psychologically challenging experiences
250
during their dosing sessions[50]. The survey includes seven factors: grief, fear, death, insanity,
251
isolation, physical distress, and paranoia.
252
Other psychosocial functioning
253
We assessed anxiety, depression, fatigue, participation in social activities, and cognitive abilities,
254
using the scales from the PROMIS‐29+2. Scoring was conducted in the same way as for pain
255
interference and sleep disturbance, with standardized t-scores for each subscale ranging from 0-
256
100 (median = 50, standard deviation = 10). Higher scores indicate worse symptoms for pain
257
severity, pain interference, sleep disturbance, anxiety, depression, fatigue, and cognitive abilities.
258
Lower scores indicate worse symptoms for physical function and participation in social
259
activities. Due to a coding error, the data on participation in social activities was not usable so
260
will not be reported in this manuscript.
261
Narrative of dosing experiences
262
At the end of each dosing session (Visits 5 and 8), study therapists asked participants to write a
263
narrative describing their experience of the sessions prior to their next in-person meeting. This
264
narrative description was discussed in Visits 6 and 9, respectively, and used as content to draw
265
from in the integration sessions. We present summaries of how each participant experienced
266
psilocybin dosing sessions, drawing directly from the content of their narratives.
267
13
268
2.6. Statistical analysis
269
We present descriptive statistics for primary and secondary outcomes. We present measures of
270
effect sizes (baseline vs. end of treatment) as Cohen’s d with 95% confidence intervals for
271
secondary outcomes (sleep disturbance, chronic pain acceptance, pain severity, pain interference)
272
and exploratory outcomes (physical function, anxiety, depression, fatigue, cognitive abilities).
273
For the Patient Global Impression of Change, we present the participant ratings at the end of
274
treatment. Results are presented for each individual participant as well for participants in
275
aggregate. Two researchers (JSA & KFB) individually reviewed participants’ written narratives
276
of the dosing sessions and each generated a list of prominent themes and quotes. They then
277
compared thematic categories and generated a combined set of themes for each dosing session.
278
These themes and quotes were distilled into brief narratives for each participant included below.
279
280
3. Results
281
Overall, 368 individuals expressed interest in the study, 265 were pre-screened, 54 were eligible
282
for screening, 17 were brought in for screening, and 7 were eligible for participation (Figure 2).
283
Two participants were withdrawn by the study team after consenting given concerns about
284
ability to follow through with study procedures (n=1) and concern about their ability to build
285
rapport with study therapists (n=1). Our original recruitment target was n=10 individuals, but we
286
stopped recruitment early due to challenges with recruitment, concerns about the generalizability
287
of our results given the stringent exclusion criteria, and that standards for psychedelic research
288
changed during the conduct of our trial following the US Food and Drug Administration’s
289
publication of guidance for psychedelic clinical trials[51].
290
14
291
Figure 2. CONSORT diagram.
292
293
Of the n=5 participants who completed the protocol, all were female, four identified as
294
White/Caucasian, and one identified as Black/African American (Table 1).
295
15
296
3.1 Safety
297
Overall, the dosing scheme in this protocol was well-tolerated, with no serious adverse events
298
(Table 2). Participant 001 opted to have their second dose be 15mg rather than 25mg, with their
299
rationale being that they thought the 15mg dose was sufficient and they wanted to experience it
300
again. AEs related to the study treatment occurred on or the day after dosing and were deemed
301
Table 1. Participant demographics and baseline clinical characteristics
Participant 001
Participant 002
Participant 003
Participant
004
Age (years)
63
41
46
62
Sex at Birth
Female
Female
Female
Female
Race
Black African Amer
White
White
White
Highest education achieved
Bachelor’s
Bachelor’s
Advanced/Professional
Bachelor’s
Employment status
Retired
Full time
employed
Full time employed
Retired
Previous psychedelic use
No
No
Yes
No
Current cannabis use
(confirmed by urinalysis)
Yes
No
Yes
Yes
Clinical symptoms
Pain severity
5.9
4.0
5.4
5.4
Chronic pain acceptance
31.0
31.0
25.0
32.0
Sleep disturbance
58.3
55.3
62.6
58.3
Pain Interference
66.6
57.1
63.8
63.8
Physical Function
36.7
48.3
40.5
40.5
Anxiety
51.2
55.8
55.8
48.0
Depression
41.0
41.0
51.8
41.0
Fatigue
58.8
53.1
64.6
64.6
Cognitive abilities
44.3
50.5
44.3
44.3
FM survey score
17.0
11.0
19.0
14.0
Table 1. Pain Severity: 0-10 numeric rating scale. Sleep disturbance, pain interference, physical function, anxiety, depression, fatigue,
cognitive abilities, and participation in social activities are reported as T-scores, with 50 being the general population mean. Most
participants had substantially worse sleep, pain interference, physical function, fatigue, cognitive abilities, and participation in social
activities than the population mean.
16
minor or moderate, with the most common being headache (n=4), diarrhea (n=2), stomach ache
302
(n=1), and migraine (n=1). The headaches were of minor or moderate severity, and three
303
participants took over the counter pain medications (acetaminophen, celecoxib) to help relieve
304
headaches. Headaches resolved within two days. Blood pressure and heart rate stayed within the
305
pre-specified ranges during dosing and were not associated with any other cardiovascular
306
symptoms (Figures 3 and 4). Results from the Challenging Experiences Questionnaire are
307
presented in Table 3. Physical distress and grief were the most commonly reported domains of
308
challenging experiences.
309
Table 2. Adverse events prior, during, and after dosing
310
V 2-4 (preparation)
V5-V9
(administration)
V10-V13
(integration)
Adverse Event (AE) a
5
11
8
Related AEb
0 (5)
4 (5)
0 (5)
Serious AE
0 (5)
0 (5)
0 (5)
AE Severity for Related
Mild
0 (5)
4(5)
0 (5)
Moderate
0 (5)
1(5)
0 (5)
Severe
0 (5)
0 (5)
0 (5)
AE Type
Headache
1 (5)
4 (5)
0 (5)
Diarrhea
0 (5)
2 (5)
1 (5)
17
Stomach Ache
0 (5)
1 (5)
0 (5)
Migraine
0 (5)
1 (5)
0 (5)
Leg throbbing
0 (5)
0 (5)
1 (5)
Back Ache
1 (5)
1 (5)
0 (5)
Jaw pain
1 (5)
1 (5)
0 (5)
Fatigue
1 (5)
0 (5)
0 (5)
Sore Throat
0 (5)
0 (5)
1 (5)
Sinus Pressure
0 (5)
0 (5)
1 (5)
Vomiting
0 (5)
0 (5)
1 (5)
Fever
0 (5)
0 (5)
1 (5)
Chills
0 (5)
0 (5)
1 (5)
Body Aches
0 (5)
0 (5)
1 (5)
Tight L side of neck
1 (5)
1 (5)
0 (5)
Medication taken for
Related AE
0 (5)
3 (5)
0(5)
311
Table 2. Adverse Event (AE) a: Participants are counted once for each category regardless of the number of events. Related AEb:
312
An AE was classified as “related” if there was a reasonable possibility that the study drug or procedure caused the event. Severity
313
and relationship to study drug or procedure were determined by the Principal Investigator. Note: Participant 001 repeated the
314
15mg dose at 2nd dosing session. All other participants had the 15mg followed by the 25mg dose.
315
316
Table 3. Challenging Experiences Questionnaire (%)
317
Participant
Fear
(Dose 1,
Dose 2)
Grief
(Dose 1,
Dose 2)
Physical
Distress
(Dose 1,
Dose 2)
Insanity
(Dose 1,
Dose 2)
Isolation
(Dose 1,
Dose 2)
Death
(Dose 1,
Dose 2)
Paranoia
(Dose 1,
Dose 2)
CEQ Total
(Dose 1,
Dose 2)
001
0%, 0%
50%, 27%
0%, 0%
0%, 0%
0%, 0%
0%, 0%
0%, 0%
12%, 6%
18
002
20%, 20%
17%, 13%
88%, 76%
0%, 0%
0%, 0%
0%, 0%
0%, 0%
25%, 22%
003
0%, 16%
60%, 37%
20%, 48%
0%, 20%
47%, 20%
0%, 80%
30%, 0%
25%, 32%
004
0%, 12%
0%, 47%
64%, 60%
0%, 0%
0%, 40%
0%, 0%
0%, 0%
12%, 29%
005
12%, 0%
0%, 0%
28%, 16%
0%, 0%
0%, 0%
0%, 0%
0%, 0%
8%, 18%
Table 3. Note: Participant 001 received 15mg at both dosing sessions.
318
319
Figure 3. Heart rate in beats per minute after dosing. D1: Dose 1 (15mg). D2: Dose 2 (25mg).
320
001 D2 was 15mg rather than 25mg per request of the participant.
321
322
323
Figure 4. Systolic and diastolic blood pressure after doses 1 and 2. D1: Dose 1 (15mg). D2:
324
Dose 2 (25mg). If a participant had blood pressure >200 systolic or >110 diastolic for >15
325
minutes, they would be transferred to the emergency department.
326
3.2 Preliminary effectiveness
327
40
60
80
100
120
30 60 90 120 180 240 300 360
Beats per minute
Time after dosing (min)
001 D1 002 D1 003 D1 004 D1 005 D1
001 D2 002 D2 003 D2 004 D2 005 D2
19
Some participants reported clinically significant improvements across numerous symptom
328
domains. Figure 5 shows changes by individual as well as on average for each symptom domain.
329
For secondary outcomes, three of five participants reported >2 point decrease in pain severity,
330
four reported >4 point decreases in pain interference, and 4 reported >6 point decreases in sleep
331
disturbance. Three participants reported enhanced chronic pain acceptance while two reported
332
slight decreases in chronic pain acceptance. On average, there was a 2.3±1.3 point decrease in
333
pain severity (d=-2.1, 95% CI [-3.7 to -0.49]), 7±4.2 point decrease in sleep disturbance (d=-2.5,
334
95% CI [-4.21 to -0.75]), 9.4±4.2 point decrease in pain interference (d=-1.8, 95% CI [-3.27 to -
335
0.24]), and 2±2.8 point increase in chronic pain acceptance (d=0.54, 95% CI [-74 to 1.79]). Via
336
the PGIC, one participant (001) reported that their symptoms were “very much improved,” two
337
reported “much improved” (002, 004), and two reported “minimally improved” (003, 005). On
338
average, FM survey scores improved by 4.2±6.1 points (d=-0.78, 95% CI [-2.06 to 0.53]).
339
340
Figure 5. CPAQ: Chronic Pain Acceptance Questionnaire. Pain Severity reported as change in
341
aggregate pain score from the 7 days prior to the intervention to the end of the intervention.
342
Sleep disturbance, pain interference, physical function, anxiety, depression, fatigue, participation
343
in social activities, and cognitive abilities are all reported as T-scores per PROMIS scoring.
344
Negative change scores indicate improvement for pain severity, pain interference, sleep
345
disturbance, FM score, anxiety, depression, and fatigue. Positive change scores indicate
346
improvement for CPAQ, physical function, participation in social activities, and cognitive
347
abilities.
348
349
20
For exploratory outcomes (Figure 5), four of five participants reported decreases in anxiety,
350
physical function, and fatigue, and three reported improvements in cognitive abilities. On
351
average, there was a 5.4±8.8 point increase in physical function (d=2.1, 95% CI [-0.29 to 2.40]),
352
4.6±4.5 point increase in cognitive abilities (d=1.39, 95% CI [-0.05 to 2.77]), a 5.9±3.7 point
353
decrease in fatigue (d=-1.1, 95% CI [-2.46 to 0.25]), and a 5.6±4.1 point decrease in anxiety
354
(d=-1.4, 95% CI [-2.75 to 0.06]). There were no substantial changes in depression scores
355
(d=0.03, 95% CI [-1.27 to 1.21]).
356
357
3.3 Qualitative narratives
358
Participant 001 reported strong feelings of connectedness with her family as well as dreamlike
359
experiences during the first dosing session (15mg) that included “time travel to ancient Egypt.”
360
She also practiced yoga during this dosing session. During the second dosing session (also
361
15mg), this sense of connectedness extended to messaging with a forest and stars, with the forest
362
saying, “We’re in trouble. Humans are destroying us. We are all connected.” and the stars
363
pointing to the “origin of original injury,” which was a car accident when the participant was 13
364
years old. Following this, the participant noted that these interactions gave her insight on how to
365
“repair” her body so she “started moving & doing yoga.”
366
367
In contrast to 001, Participant 002 did not have a pleasant experience for either dose, and was
368
“disappointed it was not a more enjoyable time”. She “felt almost as if I was not a good
369
participant simply for having to use the bathroom” or removing headphones and the eyemask,
370
tying into a realization that she had high expectations of herself and others that were not often
371
met. Participant 002 also reported moderate pain during both sessions, including back pain
372
21
during the first dosing sessions and that she “felt muscle spasms and parts of my body throb”
373
during the second. Despite these negative experiences, she noted that the “medicine had numbed
374
me” so the pain felt more manageable than it otherwise did. Although she experienced “no grand
375
visions or revelations” and a headache like the “worst hangover” after the session, she believed
376
“the medicine does help so its [sic] worth a day of being uncomfortable”.
377
378
The first dosing session for Participant 003 began with “slight visual distortions” and reflections
379
on “lost loved ones, lost family members, things, places, people I don’t think of often.” Early in
380
the session, the participant was “surprised I’ve no pain to report” but experienced sweating
381
throughout much of the day. At the end of session, she reported to be “awake, present, tired,
382
limited pain, but altogether in a good place.” Themes of connection and a focus on loved ones
383
continued in the second dosing session. The participant noted “I met my grandparents and great
384
aunt again and gained such a sense of pride and love, I felt my grandmother’s touch, as clear as
385
can be.” This second dosing session had stronger visual components, including visiting a “shark
386
habitat,” traveling “back to the caves, tunnels, and trees,” and a brief encounter with an “Asian
387
red dragon” while “in the sky.” Another prominent theme seemed to be a generalized sense of
388
gratitude: “Gratitude keeps surfacing – grateful for the journey, the time and space to rest, even
389
gratitude for this often challenging body, since it means I’m still alive.”
390
391
Participant 004 noted powerful imagery in the first experience that involved moving spatially
392
between different locations, such as moving from a lake into a forest, being “lifted to the sky to
393
dance”, and then being in a cave where she could see the Northern Lights through a crevice. She
394
was “physically comfortable” and was able to “push the music down my body to relieve the
395
22
pain”. By the end of the experience, she was “warm, cozy and almost pain free”. The second
396
experience also involved spatial movement between locations, such as moving from a hammock
397
on a porch, to sitting at a kitchen table with family, then moving to a temple. Her reflections
398
touched on constructing a narrative around her challenges in a way that suited her, noting
399
“trauma has a negative connotation but for me it’s been useful and even beautiful”. She also
400
“acquired… the skill of ‘toughness’”, reflecting that “I hold it, I have used it well” despite not
401
wanting to have acquired it in the first place.
402
403
Participant 005 began by noting that “I had little expectations of what would come from this as
404
far as long-term progress reduction of pain [sic]”. Their first dosing session was characterized by
405
increased mental imagery, mindfulness (i.e., present moment awareness), and a sense of
406
creativity. The participant was particularly engaged with the visual effects of the drug, often
407
finding symbolism and beauty in their visions. They reported a lack of focus the day after dosing,
408
feeling sadness and rejection “around certain issues”, and also sadness because she did not feel
409
like doing the write-up and was worried it would not be good enough. The participant’s second
410
dosing session included a strong focus on introspection and connection to family, ancestors, and
411
nature. They reported insights on familial relationships and on how to let her pain go and cope.
412
They came to the conclusion that much of their pain stemmed from ancestral trauma and that it
413
“felt really good knowing the pain is not mine. It’s my ancestors but they’re [sic] there’s
414
something that I can do about it for me and them.”
415
416
4. Discussion
417
23
This open-label investigation of PAT for fibromyalgia demonstrated that PAT was well-
418
tolerated, with no serious adverse events associated with the study treatment. Mild and moderate
419
AEs, such as headaches or diarrhea on the day of or after dosing, resolved shortly thereafter.
420
There were no persistent psychological AEs by the end of treatment. Most participants also
421
reported improvements in pain severity, pain interference, and sleep disturbance, with small
422
improvements in chronic pain acceptance in three participants and a decrease in chronic pain
423
acceptance in two. The improvements in secondary outcomes of pain severity, pain interference,
424
sleep disturbance, and PGIC all align with what are considered clinically meaningful changes for
425
these domains[49, 52-54].
426
427
Although there have been limited clinical trials of PAT for chronic pain conditions, our findings
428
align with other preliminary work supporting analgesic effects of psilocybin. For instance, a
429
small exploratory study in n=16 participants with cluster headaches found that cluster attack
430
frequency was significantly reduced in the three weeks following administration of psilocybin
431
compared with placebo (d = 0.69)[29]. Similarly, a proof-of-concept, double blinded,
432
randomized, placebo controlled trial of migraine (n=10) reported that the reduction in weekly
433
migraine days from baseline was significantly greater after psilocybin than placebo[30]. Our
434
findings also align with results from naturalistic studies of people using psilocybin for chronic
435
pain[55]. Survey studies have reported that a number of people using psilocybin and other
436
psychedelics for a range of chronic pain conditions often report decreased pain symptoms
437
following psychedelic use, and that in some cases psychedelics are more effective than their
438
other pain medications[32, 56].
439
440
24
In addition to improvements in pain symptoms, participants also reported positive changes in
441
mood, social cognition, sleep, and global functioning. These findings complement findings from
442
the past decade indicating transdiagnostic potential of PAT for a variety of clinical
443
conditions[57] as well as broad improvements in wellbeing observed when psychedelics are
444
studied in healthy adults[58]. For example, numerous studies have noted robust decreases in
445
depression[17, 41] and anxiety following PAT[59]. Although most participants in the current
446
study reported decreases in anxiety, only one had a decrease in depression. This could possibly
447
be driven by floor effects, given that the sample had below average depression scores at baseline
448
because we excluded participants with severe depression. Similar to the current study,
449
improvements in sleep have been observed in studies with psilocybin[60] and other
450
psychedelics[61]. Lastly, all participants reported at least some improvements in global
451
functioning (e.g., one reported “very much improved”, two reported “much improved”, and two
452
reported “minimally improved”). In summary, our preliminary findings motivate future research
453
into the effects of PAT on the multimodal underpinnings of FM, including pain, mood, and
454
sleep.
455
456
There were also numerous themes that emerged from participants’ reports of the dosing sessions
457
that are consistent with those reported in previous research. First, most participants reported
458
experiencing dreamlike states and vivid mental imagery that at times were considered to be
459
highly symbolic and meaningful. This aligns with the well-established perceptual effects of the
460
drugs[62] as well as recent findings that the visual effects of psilocybin and other psychedelics
461
may play a role in long-term outcomes[63-65]. A second theme that was common among
462
participants was a sense of connection to loved ones and ancestors, with several participants
463
25
additionally remarking on connections to nature and the universe more broadly. Although rarely
464
reported in clinical trials, experiences of connecting with ancestors have been anecdotally noted
465
in the psychedelic community[66] and are common in indigenous psychedelic ceremonies. In a
466
trial of PAT for alcohol dependence, one participant had struggled with alcohol use since her
467
mother’s death approximately two decades prior[67]. During the dosing session, she “reported to
468
her guides that she saw her deceased mother present in the room with her standing to her left.”
469
Such experiences could potentially be opportunities to process previous traumas and estranged
470
relationships, but further research is needed to delineate the nuances and ethics of such work.
471
Many participants also remarked on a generalized sense of increased gratitude during their
472
dosing sessions, which is consistent with previous research noting enduring increases in the
473
subjective emotion of gratitude following psychedelic experiences[60, 68, 69]. Finally, many of
474
these themes align with a qualitative study of people who had self-medicated with psychedelics,
475
which found that reported pain scores improved acutely and after taking psychedelics, with
476
positive reframing and somatic presence playing important roles in improvement[33].
477
478
Limitations
479
Although promising, these findings are limited by a number of important factors which must be
480
addressed with future research. First, our study design did not use a model of PAT that was
481
clinically translational given the large time commitment associated with therapy (e.g., two
482
therapists present during dosing), so our safety results may not translate to larger studies with
483
less psychotherapeutic support or clinical settings. Second, our results are limited by a small
484
sample size that likely does not generalize to typical clinical populations of fibromyalgia given
485
the rigorous screening process that excluded a very high proportion of individuals who were
486
26
seeking this therapy. The small sample size also means that the findings should be considered as
487
highly preliminary. Third, we had no control group and did not blind study participants or study
488
staff to the treatments administered, which may have affected outcomes given the strong societal
489
narratives around positive treatment outcomes associated with PAT[70]. Lastly, further research
490
is needed to validate and explore the role of psychotherapy in the safety and potential efficacy of
491
this treatment[71]. The preliminary findings from our small, unblinded study will soon be added
492
to by three other ongoing clinical trials of PAT for FM being conducted elsewhere (Table 4).
493
Table 4. Other clinical trials of psilocybin-assisted therapy for fibromyalgia
Study title
Sponsor
Study design
N
Primary outcome
Psilocybin in Patients
with Fibromyalgia:
EEG-measured Brain
Biomarkers of Action
NCT05548075
Imperial
College
London
Mechanistic, within-
subjects design. 2 doses of
up to 25mg psilocybin
with psychotherapeutic
support
20
1. Lempel-Ziv
complexity
2. The Brief
Experiential
Avoidance
Questionnaire
Psilocybin-facilitated
treatment for chronic
pain
NCT05068791
University of
Alabama at
Birmingham
Parallel group design,
double-blind, placebo
controlled.
0.36mg psilocybin/kg
body weight (active)
2.5mg
dextromethorphan/kg
body weight (placebo)
30
1. Change in daily
self-reported pain
severity (0-100
visual analog scale)
The impact of
Psilocybin on Pain in
Fibromyalgia Patients
(PsiloFM)
NCT06368492
Maastricht
University
Double-blind,
randomized, placebo-
controlled. All
participants receive doses
of placebo, 5mg
psilocybin, 10mg
psilocybin
35
1. Ischemic Pain
perception
2. Pressure-evoked
pain perception
3. Self-reported
pain (0-10 visual
analog scale)
Table 4. Currently, there are three additional ongoing trials evaluating the safety and efficacy of PAT in the
494
treatment of FM.
495
27
Conclusions
496
In this small, open-label clinical trial, we show that PAT was safe and well-tolerated among
497
people with FM, and that individuals generally reported positive impacts on global symptoms
498
and across many FM-related symptom domains. Some participants reported clinically
499
meaningful improvements in pain severity, pain interference, anxiety, and sleep disturbance,
500
with small improvements in chronic pain acceptance. Nonetheless, given the study limitations,
501
larger, controlled studies with a more clinically translational design are necessary to understand
502
whether this therapy is safe and effective in the treatment of FM.
503
Preregistration Statement: The study was not posted to a preregistration website.
504
Data Availability Statement: Given the personalized and sensitive nature of the collected data,
505
the dataset is not publicly available.
506
Acknowledgements
507
This project was supported by funds from Tryp Therapeutics, the Michigan Psychedelic Center,
508
and Department of Anesthesiology of the University of Michigan Medical School.
509
Disclosures
510
JSA’s effort on this publication was partially supported by the National Institute of Arthritis and
511
Musculoskeletal and Skin Diseases of the National Institutes of Health under T32AR007080-
512
44S1. JSA has received speaking fees for lectures at The Women's Center of Southeastern
513
Michigan, Massachusetts General Hospital, and Bicycle Day SF. KFB has received grant
514
funding from the National Institute on Drug Abuse and the National Institutes of Arthritis,
515
Musculoskeletal, and Skin Diseases of the National Institutes of Health, as well as from the State
516
of Michigan Veteran Marijuana Research Program. KFB has received speaking fees for lectures
517
from the Medical Cannabis Research Advocacy Alliance, Provide Holy Cross Medical Center,
518
the Southern Pain Society, and the Michigan Center of Clinical Systems Improvement. KFB
519
received an honorarium for developing a podcast on fibromyalgia with Viatris Inc. JRG is a
520
consultant and teacher with Fluence, International and receives no grant funding. Dr. Davis is
521
supported by the Johns Hopkins Center for Psychedelic and Consciousness Research, funded by
522
private philanthropic funding from Tim Ferriss, Matt Mullenweg, Craig Nerenberg, Blake
523
Mycoskie, and the Steven and Alexandra Cohen Foundation. Dr. Davis is also supported by the
524
Center for Psychedelic Drug Research and Education, funded by anonymous private donors, and
525
the Comprehensive Cancer Center at Ohio State University. Dr. Davis is also a board member at
526
Source Research Foundation. AH is a sub investigator in a COMPASS pathways Psilocybin
527
research study ongoing at the University of Michigan. VT serves as the site Principal
528
Investigator at the University of Michigan for a project supported by funding from Takeda
529
Development Center Americas, Inc. SEH is supported by grants from NIH and Arbor Medical
530
28
Innovations, is a member of Arbor Medical Innovations , and consults for Memorial Sloan
531
Kettering Cancer Center, Dana Farber Cancer Institute, Indiana University, and Wayne State
532
University. The content is solely the responsibility of the authors and does not necessarily
533
represent the official views of the National Institutes of Health. Dr. Clauw has received
534
consulting fees from AbbVie Inc, Heron Therapeutics Inc, Aptinyx Inc, Neumentum Inc,
535
Regeneron Pharmaceuticals Inc, Swing Therapeutics Inc, Virios Inc, Allergan Sales LLC, Eli
536
Lily and Company, H. Lundbeck A/S, Pfizer Inc, Samumed LLC, and Tonix Pharmaceuticals
537
Inc. Dr. Clauw has received payment for expert testimony from Fasken Martinueau DuMoulin
538
LLP, Marks & Clerk Law LLP, Pfizer Inc, Nix Patterson LLP, Zuber Lawler & Del Duca LLP as
539
well as Kellogg, Hansen, Todd, Figel & Frederick PLLC.
540
541
542
543
29
References
544
1. D'Onghia, M., et al., The economic burden of fibromyalgia: A systematic literature
545
review. Seminars in Arthritis and Rheumatism, 2022. 56: p. 152060.
546
2. Kaplan, C.M., et al., Deciphering nociplastic pain: clinical features, risk factors and
547
potential mechanisms. Nature Reviews Neurology, 2024: p. 1-17.
548
3. Clauw, D.J., Fibromyalgia: A Clinical Review. JAMA, 2014. 311(15): p. 1547-1555.
549
4. Harte, S.E., R.E. Harris, and D.J. Clauw, The neurobiology of central sensitization.
550
Journal of Applied Biobehavioral Research, 2018. 23(2): p. e12137.
551
5. Hassett, A.L. and R.N. Gevirtz, Nonpharmacologic Treatment for Fibromyalgia:
552
Patient Education, Cognitive-Behavioral Therapy, Relaxation Techniques, and
553
Complementary and Alternative Medicine. Rheumatic diseases clinics of North
554
America, 2009. 35(2): p. 393-407.
555
6. Mascarenhas, R.O., et al., Association of Therapies With Reduced Pain and
556
Improved Quality of Life in Patients With Fibromyalgia: A Systematic Review and
557
Meta-analysis. JAMA internal medicine, 2021. 181(1): p. 104-112.
558
7. Theoharides, T.C., et al., Fibromyalgia Syndrome in Need of Effective Treatments.
559
Journal of Pharmacology and Experimental Therapeutics, 2015. 355(2): p. 255-263.
560
8. Bornemann, J., et al., Study protocol for "Psilocybin in patients with fibromyalgia:
561
brain biomarkers of action". Frontiers in Psychiatry, 2024. 15: p. 1320780.
562
9. Aday, J.S., et al., Long-term effects of psychedelic drugs: A systematic review.
563
Neuroscience & Biobehavioral Reviews, 2020. 113: p. 179-189.
564
10. Holze, F., et al., Role of the 5-HT2A Receptor in Acute Effects of LSD on Empathy
565
and Circulating Oxytocin. Frontiers in Pharmacology, 2021. 12.
566
11. Mertens, L.J., et al., Therapeutic mechanisms of psilocybin: Changes in amygdala
567
and prefrontal functional connectivity during emotional processing after psilocybin
568
for treatment-resistant depression. Journal of Psychopharmacology, 2020. 34(2): p.
569
167-180.
570
12. Close, J.B., et al., Psychedelics and psychological flexibility Results of a
571
prospective web-survey using the Acceptance and Action Questionnaire II. Journal
572
of Contextual Behavioral Science, 2020. 16: p. 37-44.
573
13. Doss, M.K., et al., Psilocybin therapy increases cognitive and neural flexibility in
574
patients with major depressive disorder. Translational Psychiatry, 2021. 11(1): p. 1-
575
10.
576
14. Aday, J.S., E.K. Bloesch, and C.C. Davoli, Can Psychedelic Drugs Attenuate Age-
577
Related Changes in Cognition and Affect? Journal of Cognitive Enhancement, 2019.
578
15. Davis, A.K., et al., Development of the Psychological Insight Questionnaire among a
579
sample of people who have consumed psilocybin or LSD. Journal of
580
Psychopharmacology, 2021. 35(4): p. 437-446.
581
16. Peill, J.M., et al., Validation of the Psychological Insight Scale: A new scale to assess
582
psychological insight following a psychedelic experience. Journal of
583
Psychopharmacology, 2022. 36(1): p. 31-45.
584
17. Goodwin, G.M., et al., Single-Dose Psilocybin for a Treatment-Resistant Episode of
585
Major Depression. New England Journal of Medicine, 2022. 387(18): p. 1637-1648.
586
30
18. Davis, A.K., et al., Effects of Psilocybin-Assisted Therapy on Major Depressive
587
Disorder: A Randomized Clinical Trial. JAMA Psychiatry, 2021. 78(5): p. 481-489.
588
19. Ross, S., et al., Rapid and sustained symptom reduction following psilocybin
589
treatment for anxiety and depression in patients with life-threatening cancer: a
590
randomized controlled trial. Journal of Psychopharmacology, 2016. 30(12): p. 1165-
591
1180.
592
20. Griffiths, R.R., et al., Psilocybin produces substantial and sustained decreases in
593
depression and anxiety in patients with life-threatening cancer: A randomized
594
double-blind trial. Journal of Psychopharmacology, 2016. 30(12): p. 1181-1197.
595
21. Goodwin, G.M., et al., Single-dose psilocybin for a treatment-resistant episode of
596
major depression: Impact on patient-reported depression severity, anxiety,
597
function, and quality of life. Journal of Affective Disorders, 2023. 327: p. 120-127.
598
22. Grob, C.S., et al., Pilot Study of Psilocybin Treatment for Anxiety in Patients With
599
Advanced-Stage Cancer. Archives of General Psychiatry, 2011. 68(1): p. 71.
600
23. Agin-Liebes, G.I., et al., Long-term follow-up of psilocybin-assisted psychotherapy
601
for psychiatric and existential distress in patients with life-threatening cancer.
602
Journal of Psychopharmacology (Oxford, England), 2020: p. 269881119897615.
603
24. Shelton, R.C. and P.S. Hendricks, Psilocybin and palliative end-of-life care. Journal
604
of Psychopharmacology, 2016. 30(12): p. 1207-1208.
605
25. Moreno, F.A., et al., Safety, Tolerability, and Efficacy of Psilocybin in 9 Patients With
606
Obsessive-Compulsive Disorder. The Journal of Clinical Psychiatry, 2006. 67(11): p.
607
1735-1740.
608
26. Johnson, M.W., et al., Pilot study of the 5-HT <sub>2A</sub> R agonist psilocybin in
609
the treatment of tobacco addiction. Journal of Psychopharmacology, 2014. 28(11):
610
p. 983-992.
611
27. Bogenschutz, M.P., et al., Percentage of Heavy Drinking Days Following Psilocybin-
612
Assisted Psychotherapy vs Placebo in the Treatment of Adult Patients With Alcohol
613
Use Disorder: A Randomized Clinical Trial. JAMA Psychiatry, 2022. 79(10): p. 953-
614
962.
615
28. Ramachandran, V., et al., Relief from intractable phantom pain by combining
616
psilocybin and mirror visual-feedback (MVF). Neurocase, 2018. 24(2): p. 105-110.
617
29. Schindler, E.A.D., et al., Exploratory investigation of a patient-informed low-dose
618
psilocybin pulse regimen in the suppression of cluster headache: Results from a
619
randomized, double-blind, placebo-controlled trial. Headache: The Journal of Head
620
and Face Pain, 2022. 62(10): p. 1383-1394.
621
30. Schindler, E.A.D., et al., Exploratory Controlled Study of the Migraine-Suppressing
622
Effects of Psilocybin. Neurotherapeutics, 2020.
623
31. Kolbman, N., et al., Intravenous psilocybin attenuates mechanical hypersensitivity
624
in a rat model of chronic pain. Current biology: CB, 2023. 33(24): p. R1282-R1283.
625
32. Bonnelle, V., et al., Analgesic potential of macrodoses and microdoses of classical
626
psychedelics in chronic pain sufferers: a population survey. British Journal of Pain,
627
2022: p. 1-13.
628
31
33. Bornemann, J., et al., Self-Medication for Chronic Pain Using Classic Psychedelics:
629
A Qualitative Investigation to Inform Future Research. Frontiers in Psychiatry, 2021.
630
12.
631
34. Zia, F.Z., et al., Are psychedelic medicines the reset for chronic pain? Preliminary
632
findings and research needs. Neuropharmacology, 2023. 233: p. 109528.
633
35. Glynos, N.G., et al., Knowledge, Perceptions, and Use of Psychedelics among
634
Individuals with Fibromyalgia. Journal of Psychoactive Drugs, 2022. 0(0): p. 1-12.
635
36. Wolfe, F., et al., 2016 Revisions to the 2010/2011 fibromyalgia diagnostic criteria.
636
Seminars in Arthritis and Rheumatism, 2016. 46(3): p. 319-329.
637
37. Kroenke, K., et al., The Patient Health Questionnaire Somatic, Anxiety, and
638
Depressive Symptom Scales: a systematic review. General Hospital Psychiatry,
639
2010. 32(4): p. 345-359.
640
38. Goodwin, G.M., et al., Psilocybin for treatment resistant depression in patients
641
taking a concomitant SSRI medication. Neuropsychopharmacology, 2023. 48(10): p.
642
1492-1499.
643
39. Guss, J., R. Krause, and J. Sloshower, The Yale Manual for Psilocybin-Assisted
644
Therapy of Depression (using Acceptance and Commitment Therapy as a
645
Therapeutic Frame). 2020, OSF.
646
40. Aday, J., et al., Psychedelic-assisted psychotherapy: Where is the psychotherapy
647
research? 2024.
648
41. Carhart-Harris, R.L., et al., Psilocybin with psychological support for treatment-
649
resistant depression: an open-label feasibility study. The Lancet Psychiatry, 2016.
650
3(7): p. 619-627.
651
42. Spriggs, M.J., et al., Body mass index (BMI) does not predict responses to
652
psilocybin. Journal of Psychopharmacology, 2022: p. 02698811221131994.
653
43. Garcia-Romeu, A., et al., Optimal dosing for psilocybin pharmacotherapy:
654
Considering weight-adjusted and fixed dosing approaches. Journal of
655
Psychopharmacology, 2021: p. 0269881121991822.
656
44. Hays, R.D., et al., PROMIS®-29 v2.0 profile physical and mental health summary
657
scores. Quality of Life Research, 2018. 27(7): p. 1885-1891.
658
45. Promis. Obtain & Administer Measures. Available from:
659
https://www.healthmeasures.net/explore-measurement-systems/promis/obtain-
660
administer-measures.
661
46. Fish, R.A., et al., Validation of the Chronic Pain Acceptance Questionnaire (CPAQ)
662
in an Internet sample and development and preliminary validation of the CPAQ-8.
663
PAIN, 2010. 149(3): p. 435-443.
664
47. Dworkin, R.H., et al., Core outcome measures for chronic pain clinical trials:
665
IMMPACT recommendations. PAIN, 2005. 113(1): p. 9.
666
48. Dworkin, R.H., et al., Interpreting the Clinical Importance of Treatment Outcomes in
667
Chronic Pain Clinical Trials: IMMPACT Recommendations. The Journal of Pain,
668
2008. 9(2): p. 105-121.
669
49. Farrar, J.T., et al., Clinical importance of changes in chronic pain intensity measured
670
on an 11-point numerical pain rating scale. PAIN, 2001. 94(2): p. 149-158.
671
32
50. Barrett, F.S., et al., The Challenging Experience Questionnaire: Characterization of
672
challenging experiences with psilocybin mushrooms. Journal of
673
Psychopharmacology, 2016. 30(12): p. 1279-1295.
674
51. Us, F.D.A., Psychedelic Drugs: Considerations for Clinical Investigations. 2023.
675
52. Promis, Meaningful Change for PROMIS®. healthmeasures.net.
676
53. Chen, C.X., et al., Estimating minimally important differences for the PROMIS pain
677
interference scales: results from 3 randomized clinical trials. PAIN, 2018. 159(4): p.
678
775.
679
54. Terwee, C.B., et al., Minimal important change (MIC): a conceptual clarification and
680
systematic review of MIC estimates of PROMIS measures. Quality of Life Research:
681
An International Journal of Quality of Life Aspects of Treatment, Care and
682
Rehabilitation, 2021. 30(10): p. 2729-2754.
683
55. Lyes, M., et al., Microdosing psilocybin for chronic pain: a case series. Pain, 2022.
684
56. Andersson, M., M. Persson, and A. Kjellgren, Psychoactive substances as a last
685
resorta qualitative study of self-treatment of migraine and cluster headaches.
686
Harm Reduction Journal, 2017. 14(1): p. 60.
687
57. Raison, C.L., et al., Single-Dose Psilocybin Treatment for Major Depressive
688
Disorder: A Randomized Clinical Trial. JAMA, 2023. 330(9): p. 843-853.
689
58. Griffiths, R., et al., Mystical-type experiences occasioned by psilocybin mediate the
690
attribution of personal meaning and spiritual significance 14 months later. Journal
691
of Psychopharmacology, 2008. 22(6): p. 621-632.
692
59. Holze, F., et al., LSD-assisted therapy in patients with anxiety: open-label
693
prospective 12-month follow-up. The British Journal of Psychiatry, 2024: p. 1-9.
694
60. DellaCrosse, M., et al., “A sense of the bigger picture:” A qualitative analysis of
695
follow-up interviews with people with bipolar disorder who self-reported psilocybin
696
use. PLOS ONE, 2022. 17(12): p. e0279073.
697
61. Sessa, B., et al., Debunking the myth of ‘Blue Mondays’: No evidence of affect drop
698
after taking clinical MDMA. Journal of Psychopharmacology, 2022. 36(3): p. 360-
699
367.
700
62. Aday, J.S., et al., Psychedelic drugs and perception: a narrative review of the first era
701
of research. Reviews in the Neurosciences, 2021. 32(5): p. 559-571.
702
63. Aaronson, S.T., et al., Single-Dose Synthetic Psilocybin With Psychotherapy for
703
Treatment-Resistant Bipolar Type II Major Depressive Episodes: A Nonrandomized
704
Controlled Trial. JAMA Psychiatry, 2023.
705
64. Aday, J.S., et al., Increases in Aesthetic Experience Following Ayahuasca Use: A
706
Prospective, Naturalistic Study. Journal of Humanistic Psychology, 2024: p.
707
00221678241230609.
708
65. Klučková, T., et al., The phenomenology of psilocybin’s experience mediates
709
subsequent persistent psychological effects independently of sex, previous
710
experience or setting. 2024, medRxiv.
711
66. Rundel, M., Psychedelic Psychoanalysis: Transformations of the Self.
712
Psychoanalytic Dialogues, 2022. 32(5): p. 469-483.
713
67. Podrebarac, S.K., et al., Spiritual experiences in psychedelic-assisted
714
psychotherapy: Case reports of communion with the divine, the departed, and
715
33
saints in research using psilocybin for the treatment of alcohol dependence.
716
Spirituality in Clinical Practice, 2021. 8(3): p. 177-187.
717
68. Aday, J.S., et al., Effects of Ayahuasca on Gratitude and Relationships with Nature: A
718
Prospective, Naturalistic Study. Journal of Psychoactive Drugs, 2024: p. 1-10.
719
69. Griffiths, R.R., et al., Psilocybin-occasioned mystical-type experience in
720
combination with meditation and other spiritual practices produces enduring
721
positive changes in psychological functioning and in trait measures of prosocial
722
attitudes and behaviors. Journal of Psychopharmacology, 2018. 32(1): p. 49-69.
723
70. Aday, J.S., et al., Great Expectations: recommendations for improving the
724
methodological rigor of psychedelic clinical trials. Psychopharmacology, 2022.
725
239(6): p. 1989-2010.
726
71. Aday, J.S., R.L. Carhart-Harris, and J.D. Woolley, Emerging Challenges for
727
Psychedelic Therapy. JAMA Psychiatry, 2023. 80(6): p. 533-534.
728
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Background Anxiety disorders are a major public health burden with limited treatment options. Aims We investigated the long-term safety and efficacy of lysergic acid diethylamide (LSD)-assisted therapy in patients with anxiety with or without life-threatening illness. Method This study was an a priori -planned long-term follow-up of an investigator-initiated, two-centre trial that used a double-blind, placebo-controlled, two-period, random-order, crossover design with two sessions with either oral LSD (200 μg) or placebo per period. Participants ( n = 39) were followed up 1 year after the end-of-study visit to assess symptoms of anxiety, depression and long-term effects of psychedelics using Spielberger's State-Trait Anxiety Inventory–Global (STAI-G), the Beck Depression Inventory (BDI), the Persisting Effects Questionnaire and measures of personality traits using the NEO - Five-Factor Inventory. Results Participants reported a sustained reduction of STAI-G scores compared with baseline (least square means (95% CI) = −21.6 (−32.7, −10.4), d = 1.04, P < 0.001, for those who received LSD in the first period (94 weeks after the last LSD treatment) and −16.5 (−26.2, −6.8), d = 1.02, P < 0.05, for those who received LSD in the second period (68 weeks after the last LSD treatment)). Similar effects were observed for comorbid depression with change from baseline BDI scores of −8.1 (−13.2, −3.1), d = 0.71, P < 0.01, and −8.9 (−12.9, −4.9), d = 1.21, P < 0.01, for the LSD-first and placebo-first groups, respectively. Personality trait neuroticism decreased ( P < 0.0001) and trait extraversion increased ( P < 0.01) compared with study inclusion. Individuals attributed positive long-term effects to the psychedelic experience. Conclusions Patients reported sustained long-term effects of LSD-assisted therapy for anxiety.
Article
Full-text available
Background Chronic pain is a leading cause of disability worldwide. Fibromyalgia is a particularly debilitating form of widespread chronic pain. Fibromyalgia remains poorly understood, and treatment options are limited or moderately effective at best. Here, we present a protocol for a mechanistic study investigating the effects of psychedelic-assisted-therapy in a fibromyalgia population. The principal focus of this trial is the central mechanism(s) of psilocybin-therapy i.e., in the brain and on associated mental schemata, primarily captured by electroencephalography (EEG) recordings of the acute psychedelic state, plus pre and post Magnetic Resonance Imaging (MRI). Methods Twenty participants with fibromyalgia will complete 8 study visits over 8 weeks. This will include two dosing sessions where participants will receive psilocybin at least once, with doses varying up to 25mg. Our primary outcomes are 1) Lempel-Ziv complexity (LZc) recorded acutely using EEG, and the 2) the (Brief Experiential Avoidance Questionnaire (BEAQ) measured at baseline and primary endpoint. Secondary outcomes will aim to capture broad aspects of the pain experience and related features through neuroimaging, self-report measures, behavioural paradigms, and qualitative interviews. Pain Symptomatology will be measured using the Brief Pain Inventory Interference Subscale (BPI-IS), physical and mental health-related function will be measured using the 36-Item Short Form Health Survey (SF-36). Further neurobiological investigations will include functional MRI (fMRI) and diffusion tensor imaging (changes from baseline to primary endpoint), and acute changes in pre- vs post-acute spontaneous brain activity – plus event-related potential functional plasticity markers, captured via EEG. Discussion The results of this study will provide valuable insight into the brain mechanisms involved in the action of psilocybin-therapy for fibromyalgia with potential implications for the therapeutic action of psychedelic-therapy more broadly. It will also deliver essential data to inform the design of a potential subsequent RCT.
Article
Full-text available
Rationale Psychedelic-assisted psychotherapy (PAP) has emerged as a potential treatment for a variety of mental health conditions, including substance use disorders and depression. Current models of PAP emphasize the importance of psychotherapeutic support before, during, and after ingestion of a psychedelic to maximize safety and clinical benefit. Despite this ubiquitous assumption, there has been surprisingly little empirical investigation of the “psychotherapy” in PAP, leaving critical questions about the necessary and sufficient components of PAP unanswered. Objectives As clinical trials for psychedelic compounds continue the transition from safety- and feasibility-testing to evaluating efficacy, the role of the accompanying psychotherapy must be better understood to enhance scientific understanding of the mechanisms underlying therapeutic change, optimize clinical outcomes, and inform cost-effectiveness. Results The present paper first reviews the current status of psychotherapy in the PAP literature, starting with recent debates regarding “psychotherapy” versus “psychological support” and then overviewing published clinical trial psychotherapy models and putative models informed by theory. We then delineate lessons that PAP researchers can leverage from traditional psychotherapy research regarding standardizing treatments (e.g., publish treatment manuals, establish eligibility criteria for providers), identifying mechanisms of change (e.g., measure established mechanisms in psychotherapy), and optimizing clinical trial designs (e.g., consider dismantling studies, comparative efficacy trials, and cross-lagged panel designs). Throughout this review, the need for increased research into the psychotherapeutic components of treatment in PAP is underscored. Conclusions PAP is a distinct, integrative, and transdisciplinary intervention. Future research designs should consider transdisciplinary research methodologies to identify best practices and inform federal guidelines for PAP administration.
Article
Full-text available
Psychedelic drugs are currently being investigated for their potential to facilitate a variety of long-lasting psychological changes. One area that has yet to be systematically investigated in psychedelic research, however, regards changes in aesthetic experience (i.e., one’s attitudes, perceptions, and expression of art). This is surprising given the wealth of anecdotes directly noting increased appreciation of aesthetic experiences after psychedelic use and that psychedelics have facilitated their own distinctive aesthetic (i.e., “psychedelic art”). To address this gap in the literature, participants in the current study ( N = 54) completed a validated and multifaceted measure of aesthetic experience 1-week before, 1-week after, and 1-month after attending an ayahuasca retreat. We found that compared with baseline, participants exhibited increased levels of aesthetic experience at both the 1-week and 1-month follow-ups. Contrary to our hypotheses, measures of acute drug effects (e.g., mystical-type experiences, awe, and ego dissolution) did not predict changes in aesthetic experience. Although the study was limited by an open-label design, the results support anecdotal reports noting changes in aesthetic experience after psychedelic use. Further research is needed to address limitations related to the study design as well as to identify predictors and mechanisms of changes in aesthetic experience following psychedelic use.
Article
Full-text available
Qualitative studies and anecdotal reports suggest that experiences with ayahuasca, a psychedelic brew found in Central and South America, may be followed by individuals enduringly feeling more grateful and connected to nature. Yet, to date, these changes have been understudied. Here, participants (N = 54) completed validated surveys related to gratitude, nature relatedness, and nature appreciation one-week before, one-week after, and one-month after attending an aya-huasca retreat center. Compared to baseline, there was a significant increase in gratitude, nature relatedness, and nature appreciation at the one-week and one-month follow-ups. Ratings of mystical-type experiences and awe, but not ego dissolution, during participants' ayahuasca sessions were weakly-to-moderately correlated with these increases. The number of ayahuasca ceremonies attended at the retreat was not related to change in outcomes, underscoring the importance of the quality rather than the quantity of the experiences in post-acute change. Lastly, participant age was negatively related to the occurrence of mystical-type experiences and awe, supporting literature indicating blunted psychedelic effects with increased age. In the context of study limitations, the results suggest that mystical-type experiences and awe occasioned by ayahuasca may be linked to prosocial changes in gratitude and relationships with nature that may be beneficial to mental health. ARTICLE HISTORY
Article
Full-text available
Importance: Psilocybin shows promise as a treatment for major depressive disorder (MDD). Objective: To evaluate the magnitude, timing, and durability of antidepressant effects and safety of a single dose of psilocybin in patients with MDD. Design, setting, and participants: In this phase 2 trial conducted between December 2019 and June 2022 at 11 research sites in the US, participants were randomized in a 1:1 ratio to receive a single dose of psilocybin vs niacin placebo administered with psychological support. Participants were adults aged 21 to 65 years with a Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition diagnosis of MDD of at least 60 days' duration and moderate or greater symptom severity. Exclusion criteria included history of psychosis or mania, active substance use disorder, and active suicidal ideation with intent. Participants taking psychotropic agents who otherwise met inclusion/exclusion criteria were eligible following medication taper. Primary and secondary outcomes and adverse events (AEs) were assessed at baseline (conducted within 7 days before dosing) and at 2, 8, 15, 29, and 43 days after dosing. Interventions: Interventions were a 25-mg dose of synthetic psilocybin or a 100-mg dose of niacin in identical-appearing capsules, each administered with psychological support. Main outcomes and measures: The primary outcome was change in central rater-assessed Montgomery-Asberg Depression Rating Scale (MADRS) score (range, 0-60; higher scores indicate more severe depression) from baseline to day 43. The key secondary outcome measure was change in MADRS score from baseline to day 8. Other secondary outcomes were change in Sheehan Disability Scale score from baseline to day 43 and MADRS-defined sustained response and remission. Participants, study site personnel, study sponsor, outcome assessors (raters), and statisticians were blinded to treatment assignment. Results: A total of 104 participants (mean [SD] age, 41.1 [11.3] years; 52 [50%] women) were randomized (51 to the psilocybin group and 53 to the niacin group). Psilocybin treatment was associated with significantly reduced MADRS scores compared with niacin from baseline to day 43 (mean difference,-12.3 [95% CI, -17.5 to -7.2]; P <.001) and from baseline to day 8 (mean difference, -12.0 [95% CI, -16.6 to -7.4]; P < .001). Psilocybin treatment was also associated with significantly reduced Sheehan Disability Scale scores compared with niacin (mean difference, -2.31 [95% CI, 3.50-1.11]; P < .001) from baseline to day 43. More participants receiving psilocybin had sustained response (but not remission) than those receiving niacin. There were no serious treatment-emergent AEs; however, psilocybin treatment was associated with a higher rate of overall AEs and a higher rate of severe AEs. Conclusions and relevance: Psilocybin treatment was associated with a clinically significant sustained reduction in depressive symptoms and functional disability, without serious adverse events. These findings add to increasing evidence that psilocybin-when administered with psychological support-may hold promise as a novel intervention for MDD. Trial registration: ClinicalTrials.gov Identifier: NCT03866174.
Article
Full-text available
Psilocybin is being investigated as a treatment in adults with treatment-resistant depression (TRD). Withdrawal from serotonergic antidepressant drugs is a common prerequisite for taking part in trials of psilocybin due to the possibility of ongoing antidepressant drugs altering the psychedelic effect. This phase II, exploratory, international, fixed-dose, open-label study explored the safety, tolerability, and efficacy of a synthetic form of psilocybin (investigational drug COMP360) adjunct to a selective serotonin reuptake inhibitor in participants with TRD. Participants received a single 25 mg dose of psilocybin alongside psychological support and were followed-up for 3 weeks. The primary efficacy end point was change in the Montgomery-Åsberg Depression Rating Scale (MADRS) total score from Baseline at Week 3. Secondary end points were safety, including treatment-emergent adverse events (TEAEs), the proportion of responders and remitters at Week 3, and the change from Baseline to Week 3 in Clinical Global Impression–Severity (CGI-S) score. Nineteen participants were dosed and the mean Baseline MADRS total score was 31.7 (SD = 5.77). Twelve (63.2%) participants had a TEAE, most of which were mild and resolved on the day of onset. There were no serious TEAEs or indication of increased suicidal ideation or behavior. At Week 3, mean change from Baseline in MADRS total score was −14.9 (95% CI, −20.7 to −9.2), and −1.3 (SD = 1.29) in the CGI-S. Both response and remission were evident in 8 (42.1%) participants. Larger, comparator-controlled trials are necessary to understand if this paradigm can optimize treatment-outcome where antidepressant drug withdrawal would be problematic.
Article
Nociplastic pain is a mechanistic term used to describe pain that arises or is sustained by altered nociception, despite the absence of tissue damage. Although nociplastic pain has distinct pathophysiology from nociceptive and neuropathic pain, these pain mechanisms often coincide within individuals, which contributes to the intractability of chronic pain. Key symptoms of nociplastic pain include pain in multiple body regions, fatigue, sleep disturbances, cognitive dysfunction, depression and anxiety. Individuals with nociplastic pain are often diffusely tender - indicative of hyperalgesia and/or allodynia - and are often more sensitive than others to non-painful sensory stimuli such as lights, odours and noises. This Review summarizes the risk factors, clinical presentation and treatment of nociplastic pain, and describes how alterations in brain function and structure, immune processing and peripheral factors might contribute to the nociplastic pain phenotype. This article concludes with a discussion of two proposed subtypes of nociplastic pain that reflect distinct neurobiological features and treatment responsivity.
Article
Importance Bipolar II disorder (BDII) is a debilitating condition frequently associated with difficult-to-treat depressive episodes. Psilocybin has evidence for rapid-acting antidepressant effects but has not been investigated in bipolar depression. Objective To establish the safety and efficacy of psilocybin in patients with BDII in a current depressive episode. Design, Setting, and Participants This was a 12-week, open-label nonrandomized controlled trial conducted at Sheppard Pratt Hospital. Participants aged 18 to 65 years with BDII, a current depressive episode longer than 3 months, and documented insufficient benefit with at least 2 pharmacologic treatments during the current episode were invited to participate. Of 70 approached, 19 met inclusion criteria and were enrolled. The trial was conducted between April 14, 2021, and January 5, 2023. Interventions A single dose of synthetic psilocybin, 25 mg, was administered. Psychotropic medications were discontinued at least 2 weeks prior to dosing. Therapists met with patients for 3 sessions during pretreatment, during the 8-hour dosing day, and for 3 integration sessions posttreatment. Main Outcomes and Measures The primary outcome measure was change in Montgomery-Åsberg Depression Rating scale (MADRS) at 3 weeks posttreatment. Secondary measures included MADRS scores 12 weeks posttreatment, the self-rated Quick Inventory of Depression Symptoms-Self Rating (QIDS-SR), and the self-rated Quality of Life Enjoyment and Satisfaction Questionnaire-Short Form (Q-LES-Q-SF), each completed at baseline and all subsequent visits. Safety measures included the Columbia Suicide Severity Rating Scale (CSSRS) and the Young Mania Rating Scale (YMRS) completed at each visit. Results Of the 15 participants in this study (6 male and 9 female; mean [SD] age, 37.8 [11.6] years), all had lower scores at week 3, with a mean (SD) change of −24.00 (9.23) points on the MADRS, (Cohen d = 4.08; 95% CI, −29.11 to −18.89; P < .001). Repeat measures analysis of variance showed lower MADRS scores at all tested posttreatment time points, including the end point (Cohen d = 3.39; 95% CI, −33.19 to −16.95; adjusted P < .001). At week 3, 12 participants met the response criterion (50% decrease in MADRS), and 11 met remission criterion (MADRS score ≤10). At the study end point, 12 patients met both response and remission criteria. QIDS-SR scores and Q-LES-Q-SF scores demonstrated similar improvements. YMRS and CSSRS scores did not change significantly at posttreatment compared to baseline. Conclusions and Relevance The findings in this open-label nonrandomized controlled trial suggest efficacy and safety of psilocybin with psychotherapy in BDII depression and supports further study of psychedelics in this population.