PreprintPDF Available

Reduced brain responsiveness to emotional stimuli with escitalopram but not psilocybin therapy for depression

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

Abstract

Psilocybin therapy is an emerging intervention for depression that may be at least as effective as standard first-line treatments i.e., Selective Serotonin Reuptake Inhibitors (SSRIs). Here we assess neural responses to emotional faces (fear, happy, and neutral) using Blood Oxygen-Level Dependent (BOLD) functional Magnetic Resonance Imaging (fMRI) in two groups with major depressive disorder: 1) a ‘psilocybin group’ that received two dosing sessions with 25mg plus six weeks of daily placebo, and 2) an ‘escitalopram group’ that received six weeks of the SSRI escitalopram, plus two dosing sessions with an inactive/placebo dose of 1mg psilocybin. Both groups had an equal amount of psychological support throughout. An emotional face fMRI paradigm was completed at baseline (pre-treatment) and at the six-week post-treatment primary endpoint (three weeks following psilocybin dosing sessions). An analysis examining the interaction between patient group (psilocybin vs. escitalopram) and time-point (pre-vs. post-treatment) showed a robust effect in a distributed network of cortical brain regions. Follow-up analyses showed that post-treatment BOLD responses to emotional faces of all types were significantly reduced in the escitalopram group, with no change, or even a slight increase, in the psilocybin group. Specific analyses of the amygdala showed a reduction of response to fear faces in the escitalopram group, but no effects for the psilocybin group. Despite large improvements in depressive symptoms in the psilocybin group, psilocybin-therapy had only a minor effect on brain responsiveness to emotional stimuli. We suggest that reduced emotional responsiveness may be a biomarker of SSRIs’ antidepressant action that is not shared by psilocybin-therapy.
1
Reduced brain responsiveness to emotional stimuli with
1
escitalopram but not psilocybin therapy for depression
2
3
Matthew B. Wall1,2,3, Lysia Demetriou1,2,4, Bruna Giribaldi5, Leor Roseman5, Natalie Ertl1,2,
4
David Erritzoe5, David J. Nutt5, Robin L. Carhart-Harris5,6.
5
6
1Invicro London, Hammersmith Hospital, UK
7
2Faculty of Medicine, Imperial College London, UK
8
3Clinical Psychopharmacology Unit, University College London, UK
9
4Nuffield Department of Women’s and Reproductive Health, University of Oxford
10
5Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Sciences,
11
Faculty of Medicine, Imperial College London
12
6Psychedelics Division, Neuroscape, University of California San Francisco, USA.
13
14
15
Corresponding Author:
16
Matthew B. Wall
17
matt.wall@invicro.co.uk
18
19
20
Funding and Disclosures
21
This trial was funded by a private donation from the Alexander Mosley Charitable Trust,
22
supplemented by Founders of ICL’s Centre for Psychedelic Research (CPR)
23
(https://www.imperial.ac.uk/psychedelic-research-centre/funding-partners/). Infrastructure
24
support was provided by the NIHR Imperial Biomedical Research Centre and the NIHR
25
Imperial Clinical Research Facility.
26
Authors MBW, LD, and NE’s primary employer is Invicro LLC., a contract research
27
organization which provides services to the pharmaceutical and biotechnology industries.
28
RCH is a scientific advisor to Maya Health, Osmind, Beckley Psytech, Usona Institute,
29
Mindstate, Entheos Labs, and TRYP therapeutics. DJN is a scientific advisor to Awakn Life
30
Sciences, and Algernon pharmaceuticals, and Psyched Wellness. DE is a scientific advisor to
31
Mindstate, Aya Biosciences, and Clerkenwell Health.
32
33
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice.
2
Abstract
34
35
Psilocybin therapy is an emerging intervention for depression that may be at least as
36
effective as standard first-line treatments i.e., Selective Serotonin Reuptake Inhibitors
37
(SSRIs). Here we assess neural responses to emotional faces (fear, happy, and neutral) using
38
Blood Oxygen-Level Dependent (BOLD) functional Magnetic Resonance Imaging (fMRI) in
39
two groups with major depressive disorder: 1) a ‘psilocybin group that received two dosing
40
sessions with 25mg plus six weeks of daily placebo, and 2) an ‘escitalopram group’ that
41
received six weeks of the SSRI escitalopram, plus two dosing sessions with an
42
inactive/placebo dose of 1mg psilocybin. Both groups had an equal amount of psychological
43
support throughout. An emotional face fMRI paradigm was completed at baseline (pre-
44
treatment) and at the six-week post-treatment primary endpoint (three weeks following
45
psilocybin dosing sessions). An analysis examining the interaction between patient group
46
(psilocybin vs. escitalopram) and time-point (pre- vs. post-treatment) showed a robust
47
effect in a distributed network of cortical brain regions. Follow-up analyses showed that
48
post-treatment BOLD responses to emotional faces of all types were significantly reduced in
49
the escitalopram group, with no change, or even a slight increase, in the psilocybin group.
50
Specific analyses of the amygdala showed a reduction of response to fear faces in the
51
escitalopram group, but no effects for the psilocybin group. Despite large improvements in
52
depressive symptoms in the psilocybin group, psilocybin-therapy had only a minor effect on
53
brain responsiveness to emotional stimuli. We suggest that reduced emotional
54
responsiveness may be a biomarker of SSRIs antidepressant action that is not shared by
55
psilocybin-therapy.
56
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
3
Introduction
57
58
Major Depressive Disorder (MDD) is one of the most prevalent and debilitating psychiatric
59
disorders 1. Current treatment guidelines suggest psychotherapy for mild depression, with
60
pharmacotherapy or a combination of the two treatments recommended for moderate-to-
61
severe cases 2. Pharmacotherapy with Selective Serotonin Reuptake Inhibitors (SSRIs) is a
62
popular option. However, SSRIs are only moderately effective, take four to eight weeks to
63
show a meaningful therapeutic response, and have acceptability rates (indexed by
64
treatment discontinuation) comparable with placebo 3, i.e. around 25-30% 4.
65
66
A phenomenon commonly referred to as emotional blunting(i.e., a restricted range or
67
intensity of emotional experience) has been associated with SSRI use, with one survey
68
suggesting a prevalence close to 50% 5. Diminished libido and sexual functioning with SSRI
69
treatment 6,7 and a modest impact on symptoms of anhedonia in depression 8 are also
70
commonly reported. One potential cause of diminished emotional responsiveness via SSRIs
71
may be increased 5-HT activity on inhibitory postsynaptic 5-HT1A receptors in limbic and
72
paralimbic circuitry implicated in affective and hedonic functioning 9,10. Specifically,
73
decreased amygdala responsiveness to emotional stimuli (i.e. a normalization of responses
74
to negative stimuli) has been found in depressed patients soon after beginning a course of
75
SSRIs 11,12, and similar effects have been found in healthy subjects with short-term use of
76
SSRIs 13,14. This emotional/amygdala blunting has been hypothesized to be central to their
77
therapeutic effect 15,16.
78
79
Novel treatments for major depression are generating a great deal of interest in psychiatry
80
17,18. Ketamine has shown potential as a rapid-acting treatment option 19, and promising
81
results are being seen in small clinical trials assessing psilocybin therapy for depression 2022
82
and depressive symptoms 2325. Psilocybin is a naturally occurring ‘classic’ psychedelic which
83
exerts its characteristic subjective effects through direct agonism of the 5-HT2A receptor 26.
84
Acutely a 25mg dose of psilocybin has profound effects on spontaneous brain function 27
85
and longer-term functional brain changes have been reported with this dose of psilocybin in
86
depressed 2830 as well as in healthy individuals 31,32. In clinical use, psilocybin dosing
87
sessions can be combined with psychological therapy and support, before (‘preparation’
88
sessions), during the dosing sessions, and after (‘integration’ sessions)33.
89
90
Emotional face perception paradigms are widely used in human fMRI studies, and
91
particularly in depression research 34. Using this approach in a sample of patients before and
92
one-day after psilocybin therapy for treatment-resistant depression we previously observed
93
an increase in BOLD responsiveness to emotional face stimuli in depressed patients35, which
94
may reflect early changes in responses to treatment or ‘afterglow’ phenomena36, i.e., a sub-
95
acute life in mood. Despite being directionally opposite to the BOLD-reducing action of SSRIs
96
on emotional faces 11,12,37, this finding was predictive of longer-term treatment response 35.
97
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
4
An extension of this work identified changes in amygdala connectivity that were also
98
associated with clinical improvements 38. In contrast to the emotional blunting associated
99
with SSRIs, a sense of increased emotional connection is often reported with effective
100
psychedelic therapy 39. Thus, a differential effect on emotional functioning between SSRIs
101
and psychedelic therapy could explain their contrasting fMRI findings in emotional
102
processing paradigms.
103
104
The aim of the present study was to directly compare the brain effects of the two
105
treatments for the first time, in a double-blinded, randomized-controlled trial. We chose
106
escitalopram as an active comparator for psilocybin therapy because of its high
107
pharmacological selectivity as a 5-HT reuptake inhibitor and good antidepressant
108
performance; in terms of both tolerability and efficacy 3,40. Psilocybin therapy was carried
109
out in a manner that was generally consistent with previous studies. The clinical procedures
110
are detailed below and the clinical findings are fully reported elsewhere 41. The specific
111
emotional facial expression paradigm employed in this study was similar to those used in
112
previous work, e.g. 11,35. The main pre-registered hypothesis for this trial was that the two
113
treatments would have significantly different effects on brain responsiveness to emotional
114
faces (see trial registration under identifier NCT03429075;
115
https://clinicaltrials.gov/ct2/show/NCT03429075). This is therefore the first report of the
116
primary study outcome.
117
118
119
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
5
Methods
120
121
This study was approved by the Brent Research Ethics Committee, with additional approvals
122
from the UK Medicines & Healthcare products Regulatory Agency, the Health Research
123
Authority, and Imperial College London. MRI data collection, drug storage, and dispensing
124
took place at Invicro LLC, Hammersmith Hospital, London. All subjects gave written
125
informed consent, and the trial was conducted under the principles of Good Clinical
126
Practice.
127
128
Design
129
The full clinical study procedure is reported in 41. This was a phase II, double-blinded,
130
randomized, controlled experimental medicine trial. MRI scanning was done prior to any
131
therapeutic intervention (baseline) and six weeks and one day after the first dosing day. For
132
the psilocybin group, they received 2 x 25mg doses of psilocybin, three weeks apart. The
133
post-treatment MR scan occurred three weeks after the second 25mg dose. After the first of
134
the two dosing sessions, patients in the psilocybin group were provided with a bottle of
135
capsules containing microcrystalline cellulose (i.e. inert placebo capsules) and instructed to
136
take one per day for the next three weeks and two per day for the final three weeks until
137
scan visit two, which was the primary endpoint. Subjects in the escitalopram group received
138
a 1mg dose of psilocybin on each of the two dosing visits (also three weeks apart); 1mg has
139
negligible subjective effects and therefore served as a control procedure/placebo. These
140
subjects were provided with encapsulated escitalopram (10mg capsules) and were
141
instructed to take one capsule (10mg escitalopram) daily for the first three weeks, and two
142
capsules (20mg) daily for the final three weeks. Scan visit two occurred on the day of the
143
final capsule ingestion at the approximate time of peak plasma concentration, implying both
144
steady state and acute presence of escitalopram for the escitalopram group.
145
146
Clinical scales
147
The primary pre-registered (https://clinicaltrials.gov/ct2/show/NCT03429075) clinical
148
outcome was the Quick Inventory of Depression Score (QIDS-SR16) 42. Additional measures
149
used were the Beck Depression Inventory (BDI) 43, the Warwick-Edinburgh Mental Well-
150
being Scale (WEMWBS) for assessment of well-being 44, the Snaith-Hamilton Pleasure Scale
151
(SHAPS) 45 for assessment of anhedonia, the Laukes Emotional Intensity Scale (LEIS) 6 for
152
assessment of emotional function, and the PRSexDQ 46 for assessment of changes to sexual
153
function.
154
155
Participants and Recruitment
156
For full details of the recruitment and screening procedures see 41. Participants were
157
recruited using trial networks, social media, and other sources, via a recruitment website
158
(https://www.imperial.ac.uk/psychedelic-research-centre). Volunteers emailed the
159
recruitment coordinator, so all recruited participants were self-referred. There followed a
160
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
6
multi-step screening process involving telephone and video calls, and in-person sessions
161
with a trial psychiatrist.
162
163
Inclusion criteria were: a score of at least 17 (indicating moderate-to-severe MDD) on the
164
HAM-D-17 assessments which were performed on the video call, confirmation of a diagnosis
165
of depression and a satisfactory medical history (obtained from the patient’s general
166
physician), willingness to withdraw completely from psychiatric medication (at least two
167
weeks) and psychotherapy (at least three weeks) before starting the trial, age 18-80 years,
168
and sufficient competence with English. The main exclusion criteria were a history of certain
169
exclusory mental or physical health conditions (both physician-assessed), pregnancy,
170
previous courses of escitalopram, and drug or alcohol dependence.
171
172
Thirty patients were randomized to the psilocybin arm of the study, and 29 were
173
randomized to the escitalopram arm. However, four patients in the escitalopram arm
174
discontinued treatment due to side effects and were therefore excluded in the present
175
analyses. A further five subjects in the escitalopram arm did not complete the second MRI
176
visit because of Covid-19 lockdowns in the UK in March/April 2020. Four subjects in the
177
psilocybin arm also did not complete their second MRI scanning session also because of
178
Covid-19 lockdowns. After the end of the trial, it was revealed that one subject in the
179
psilocybin arm had been using cannabis regularly throughout the trial, so their data was also
180
excluded. Consequently, there were N=21 subjects available for analysis in the escitalopram
181
arm of the study, and N=25 in the psilocybin arm. See figure 1 for the full recruitment flow
182
diagram.
183
184
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
7
185
Figure 1. Recruitment flow diagram for the study.
186
187
Task and image acquisition
188
The fMRI task involved stimuli with three facial expressions: fear, happy, and neutral. These
189
were selected from the Karolinska Directed Emotional Faces set 47 and presented in blocks
190
of five images of the same type, with each image on-screen for 3s. Rest/baseline blocks
191
were also included, which displayed a small fixation cross at the center of the screen. There
192
were eight repeats of each block type presented in a pre-determined pseudo-random order,
193
and 32 blocks in total (eight each of fear, happy, neutral, and rest/baseline). The total task
194
time was eight minutes, plus a 10s buffer period at the end of the task to ensure the last
195
response was fully captured. Two different pseudo-random orders of block presentation
196
were used, and subjects saw one order on the first visit and a different order on the second
197
visit. Patients passively viewed the faces.
198
199
Data was acquired using a Siemens TIM Trio 3 Tesla MRI scanner (Siemens, Erlangen),
200
equipped with a 32-channel phased-array head-coil. Anatomical images were acquired using
201
the recommended parameters for MPRAGE by the ADNI-GO project 48: TE = 2.98ms, TR =
202
2300ms, 160 sagittal slices, 256x256 in-plane FOV, flip angle = 9, 1mm isotropic voxels.
203
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
8
The functional (Echo-Planar-Imaging) acquisition was based on the multiband EPI WIP v012b
204
provided by the University of Minnesota 4951 using a multiband acceleration factor of 2, and
205
a slice acceleration (GRAPPA) factor of 2 (TR=1250ms, TE=30ms, 44 slices, 3mm isotropic
206
voxels, FOV = 192x192mm, flip angle = 70, bandwidth = 2232Hz/pixel, 392 volumes
207
acquired). This was based on sequences previously tested and validated by 52.
208
209
Data analysis
210
Analysis was conducted using FSL version 5 53. Processing of the anatomical data used the
211
fsl_anat script, which performs a number of processes including inhomogeneity correction
212
and brain extraction using BET (Brain Extraction Tool). Pre-processing of the functional data
213
included head-motion correction, smoothing (6 mm), registration to a standard template
214
(MNI152) using a two-step registration using the subjects’ anatomical scans, and high-pass
215
filtering (0.01 Hz).
216
217
For the first-level analyses, a general linear model contained regressors derived from the
218
occurrence of the stimuli in three (fear, happy, and neutral face blocks) separate regressors.
219
These stimulus-related time-series were convolved with a standard Gamma function to
220
model the hemodynamic response. An extended set of 24 head-motion regressors were also
221
included as confounds, which included temporal derivatives and quadratic functions derived
222
from the raw head-motion parameters. Modelling used FSL’s FILM (FMRIB’s Improved
223
Linear Model) for pre-whitening and autocorrelation correction. Contrasts were computed
224
that compared each stimulus condition with the fixation cross baseline condition, as well as
225
a summary contrast that modelled all stimulus conditions relative to the fixation cross
226
baseline.
227
228
To visualize treatment effects, mid-level, fixed-effects, within-subjects analyses were used
229
to generate comparisons between pre- and post-intervention visits for each subject. Group
230
analyses were random effects (FLAME-1) models, with statistical maps thresholded at Z =
231
2.3, and p < 0.05 (cluster corrected). Mean analyses including data from all subjects and all
232
visits were generated to verify the success of the task paradigm in producing an expected
233
pattern of brain activation, and the general acquisition and analysis procedures (see
234
supplementary material for results). Between-subjects group-level analyses then used the
235
results of the mid-level analyses for a comparison between the two treatment groups
236
(psilocybin vs. escitalopram). These comparisons modelled both the difference between
237
groups, and the difference between study visits, and therefore test an interaction effect.
238
Data were extracted from functional clusters/maps in these group-level analyses and
239
plotted to visualize the precise pattern of effects across the task conditions, visits, and drug
240
treatment groups. Additionally, group-level analyses of pre- vs. post-treatment effects were
241
also performed with an anatomical amygdala mask, derived from the Harvard-Oxford sub-
242
cortical atlas, provided with FSL. The rationale for this analysis was based on previous work
243
showing that amygdala connectivity and responsiveness to emotional stimuli can be
244
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
9
specifically affected by psilocybin therapy 35,38 and that it is also a region of interest in
245
depression research 10. Finally, a control analysis was performed that compared the pre-
246
therapy (baseline) scans of the two treatment groups, to examine any potential baseline
247
differences between the two groups.
248
249
Additional exploratory analyses were conducted to examine the relationship between
250
measures of clinical outcomes, and the BOLD activation data, using Pearson’s correlations
251
and moderation analyses to examine the specific relationships between clinical depression
252
outcomes, BOLD activation data, and a subjective measure of emotional function.
253
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
10
Results
254
255
Demographics
256
257
Demographic and selected clinical (baseline, pre-treatment) information for this sample are
258
shown in table 1. There were no significant differences at baseline between the two groups
259
on any demographic or clinical measure except for the QIDS-SR-16 score, where the
260
escitalopram group were somewhat higher.
261
262
Characteristic
Escitalopram
(N=21)
Psilocybin
(N=25)
Difference
Demographic
Age: MeanSD (range)
42.811.7(21-64)
t(44)=-1.06,
p = 0.294
Female sex: no. (%)
9 (36)
2 (1) = 0.287,
p = 0.592
White race: no. (%)
24 (96)
2 (1) = 2.67,
p = 0.102
Employment status: no. (%)
Employed
16 (64)
2 (2) = 3.55,
p = 0.471
Unemployed
7 (28)
Student
2 (8)
University education (%)
19 (76)
2 (1) = 0.12,
p = 0.725
Weekly alcohol use, UK Units:
MeanSD (range)
4.85.8 (0-20)
t(44)=-1.73,
p = 0.090
No previous psilocybin use: no. (%)
17 (68)
2 (1) = 0.063,
p = 0.801
Discontinued psychiatric medication
for trial: no. (%)
10 (40)
2 (1) = 0.00,
p = 1.0
Clinical
Duration of illness, years: MeanSD
(range)
21.311.0(3-44)
t(44)=-1.84,
p = 0.08
Previous psychiatric medications:
MeanSD (range)
2.21.68 (0-6)
t(44)=-0.505,
p = 0.616
Previous use of psychotherapy: no. (%)
23 (92)
2 (1) = 0.033,
p = 0.855
QIDS-SR-16 score at baseline:
MeanSD (range)
13.93.4 (7-19)
t(44)=2.128,
p = 0.039
BDI-1A score at baseline: MeanSD
(range)
28.76.6 (16-41)
t(44)=0.34,
p = 0.735
Table 1. Demographic information and selected baseline clinical scores for the
263
sample. Pre-treatment baseline was 7-10 days before dosing day 1. All inferential
264
statistics in the third column are non-significant (at an alpha value of p < 0.05) except
265
the QIDS-SR-16 baseline score.
266
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
11
267
Clinical data
268
269
The full clinical results from the entire cohort are presented in 41, however, selected clinical
270
data from the restricted cohort of n = 45 that completed the MRI scanning sessions, after
271
exclusions, is presented here (baseline to six week timepoints). For the primary outcome
272
measure (QIDS SR-16) a mixed-effects analysis with one between-groups factor (treatment)
273
and one within-subjects factor (time) showed a significant main effect of treatment group
274
(F[1,44] = 11.76, p = 0.0013) and time (F[6,251] = 26.36, p < 0.0001), but no significant
275
interaction (figure 2A). A similar analysis was used for Beck Depression Inventory (BDI)
276
scores, which also showed significant main effects of time (F[3,113] = 55.89, p < 0.0001),
277
time (F[1,44] = 8.73, p = 0.005), and a significant interaction (F[3,127] = 6.49, p = 0.0004),
278
suggesting a significantly greater decrease in BDI scores in the psilocybin group (figure 2B).
279
280
The same analysis model applied to well-being (WEMWBS) data showed a significant main
281
effect of time (F[2,92) = 20.93, p < 0.0001), a significant main effect of treatment (F[1,44) =
282
12.11, p = 0.0011) and a significant interaction between the two factors (F[3,127) = 4.97, p =
283
0.0027). These results suggest significantly greater improvement in well-being scores in the
284
psilocybin treatment group (see figure 2C). A two-way ANOVA analysis of scores on the
285
Snaith Hamilton Anhedonia Pleasure Scale (SHAPS) showed no main effect of treatment
286
group (F[1,44) = 2.13, p = 0.15), but a significant effect of pre- vs. post-treatment (F[1,44) =
287
79.89, p < 0.0001), and a significant interaction (F[1,44) = 7.34, p = 0.0096), again suggesting
288
greater improvement in anhedonia in the psilocybin group (figure 2D). The change (pre- vs.
289
post-treatment) in scores on perceived emotional responsiveness or intensity (LEIS) were
290
analysed using an unpaired t-test, and showed a significant difference between the groups
291
(t[44] = 5.27, p < 0.0001), i.e., there was a relative decrease in emotional-intensity in the
292
escitalopram group and a relative increase in the psilocybin group (figure 2E). Finally,
293
change scores on the Psychotropic-Related Sexual Dysfunction Questionnaire (PRSexDQ)
294
were also significantly different between the two treatment groups (t[44] = 3.08, p =
295
0.0036) in this restricted cohort (figure 2F).
296
297
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
12
298
299
Figure 2. Selected clinical data from the fMRI sub-sample analyzed here. Error bars
300
are standard errors. A: Significant interaction between the treatment groups and
301
time on Beck Depression Inventory (BDI) scores, * p = 0.0002. B. Significant main
302
effect of treatment on the Quick Inventory of Depressive Symptoms (QIDS), * p =
303
0.002. C: Significant differences between the treatment groups on the Warwick-
304
Edinburgh Mental Well-being Scale (WEMWBS), * p = 0.0031. D: Significant
305
differences between the treatment groups on the Snaith Hamilton Anhedonia
306
Pleasure Scale (SHAPS), * p = 0.014. E: Significant differences between the treatment
307
groups on change (pre- vs. post-treatment) scores on the Laukes Emotional Intensity
308
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
13
Scale (LEIS), * p < 0.0001). F: Significant differences between the treatment groups
309
on the Psychotropic-Related Sexual Dysfunction Questionnaire (PRSexDQ). * p =
310
0.0036. Note, higher scores on the PRSexDQ reflect greater sexual dysfunction.
311
312
Imaging data
313
314
Subject head-motion was assessed and no subjects were excluded on this basis. There were
315
also no significant differences in head-motion between groups or across study visits; see
316
supplementary material for full details. Results of an analysis modelling the mean task
317
activation of all subjects from each group and all scan visits showed the predicted pattern of
318
task effects, with the emotional faces activating a broad pattern of brain regions including
319
primary and secondary visual cortex, amygdala, thalamus, insula, and superior frontal
320
regions (see supplementary figure S1). This pattern is entirely consistent with previous work
321
(e.g. 54) and therefore validates the experimental and analysis procedures.
322
323
Results from the main analyses of treatment and drug group effects are shown in figure 3.
324
This is a complex effect that models the interaction between the (within-subject) pre- and
325
post-treatment factor and the (between-subjects) treatment group factor. The voxel-wise
326
analyses found a large network of areas in which there was a decreased BOLD response
327
post-treatment in the escitalopram group, relative to the psilocybin group. Significant
328
clusters were evident in the dorsolateral pre-frontal cortex, supramarginal gyrus, mid-insula,
329
and superior temporal lobe.
330
331
Data was extracted from this set of clusters and plotted across all the stimulus conditions
332
(figure 3, panels B-E). A mixed-model 2 (group) x 2 (visit) x 3 (facial expression) ANOVA
333
showed a significant main effect of study visit (F[1,44] = 5.27, p = 0.027). There were also
334
significant interactions between the treatment group and visit factors (F[1,44] = 10.83, p =
335
0.002), and the visit and facial expression factors (F[2,86] = 10.09, p < 0.001).
336
337
Follow-up comparisons revealed that in the escitalopram group there was a significant
338
reduction in responses on the second visit (six weeks) for all three individual facial
339
expressions: fear (t[20] = 2.82, p = 0.011), happy (t[20] = 3.79, p = 0.001), and neutral (t[20]
340
= 2.25, p = 0.036). This effect is also significant when collapsing across all facial expressions
341
(t[29] = 3.16, p = 0.005). All these results survive a Bonferroni-corrected p threshold of
342
0.0125, except the neutral faces result. In the psilocybin group, similar comparisons showed
343
a significant increase in responses on the post-therapy visit for the neutral facial expressions
344
(t[24] = -3.17, p = 0.004).This neutral faces result for the psilocybin group survived the
345
corrected alpha threshold (p = 0.0125). There were no other significant effects.
346
347
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
14
348
Figure 3. Results from the main analysis. Panel A shows results of a voxel-wise (Z>
349
2.3, p < 0.05, corrected for multiple comparisons) analysis of an interaction effect
350
between the two treatment groups and study visit (psilocybin (pre- > post-
351
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
15
treatment) > escitalopram (pre- > post-treatment), showing a relatively greater
352
activation to faces in the post-treatment scan (relative to the pre-treatment baseline
353
scan) for the psilocybin group, compared with the escitalopram group. Panels B-E
354
break down this high-level effect and plot ROI data from this total set of clusters for
355
all task conditions, groups, and study visits. Results show that the effect is driven by
356
a decrease in response to all face types in the escitalopram group in the post-
357
treatment visit, with minimal post-treatment change in the psilocybin group. In fact,
358
for neutral faces there is an increased activation post-treatment with psilocybin. * =
359
p < 0.05, see text for exact values.
360
361
Figure 4 shows the results of the analyses using a bilateral amygdala mask. These analyses
362
compared pre- vs. post-treatment effects for the various task contrasts within each group
363
and found a significant activation cluster in the right amygdala for the escitalopram group,
364
with a smaller cluster in the right amygdala for the psilocybin group, both on the task
365
contrast of fear > neutral faces (figure 3, panels A and C). There were no other significant
366
effects on the other task contrasts conducted in these analyses using the amygdala mask.
367
Extracting and plotting data from the relevant activation clusters (figure 3; panels B and D),
368
and comparing baseline to post-therapy responses found that there was no change in the
369
psilocybin group post-treatment, however, there was a reduction in responses in the
370
escitalopram group, with a significant effect for the fear faces (t[20]=2.33, p=0.031).
371
However, this effect does not survive a multiple-comparisons correction for the four tests
372
conducted here.
373
374
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
16
375
376
Figure 4. Results from analyses using an anatomical amygdala mask for the
377
comparison of pre- vs. post-treatment for the task contrast fear > neutral. The
378
escitalopram group showed relatively large and bilateral activation clusters for this
379
contrast (A) whereas the psilocybin group showed a more dorsal and smaller cluster
380
only in the left amygdala (C). ROI data from these clusters revealed a significant
381
reduction in response to fear faces post-treatment in the escitalopram group, with
382
no effects seen in the psilocybin group. * = p < 0.05.
383
384
Reassuringly, in the control analysis of the pre-treatment (baseline) scans, BOLD activations
385
were not significantly different between the two groups on any task condition or contrast.
386
387
388
Relationships between BOLD and clinical data
389
390
Exploratory analyses were conducted to examine relationships between the ROI data
391
plotted in figure 2 and the selected clinical data presented in figure 1. Correlations were
392
calculated within each patient group comparing the change in clinical scores (i.e., score at
393
baseline, subtracted from the 6-week follow-up) with the change in BOLD activity (visit 1
394
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
17
subtracted from visit 2) from the regions identified in figure 2. There were no significant
395
correlations present in these analyses. Results can be found in the supplementary material.
396
397
Additional exploratory work used two moderation analyses to test whether the change in
398
brain activity (average of all faces) could predict the main clinical outcome (change in QIDS
399
scores from baseline to six weeks), and whether this was moderated by emotional function
400
(change in score on the LEIS). In the psilocybin group, the LEIS alone was significantly
401
predictive of QIDS outcomes (Z = -2.24, p = 0.025), but brain activity alone was not (Z = -
402
0.07, p = 0.944), and neither was the interaction between the predictor and moderator (Z = -
403
0.91, p = 0.363). For the escitalopram group, the change in brain activity was predictive of
404
BDI outcomes (Z = 1.97, p = 0.048), and while the LEIS alone was not (Z = 0.36, p = 0.721);
405
the interaction (i.e., a moderation of the predictive power of brain activity via LEIS) was
406
strongly significant (Z = 3.14, p = 0.002).
407
408
In moderation analyses, the interaction term is the most instructive result, and a significant
409
result denotes that the relationship between the predictor and the dependent variable is
410
stronger or weaker, given varying levels of the moderator. In this case, since all three
411
variables are negative (i.e. relative decreases in brain activity, decreases in depression, and
412
decreases in emotional function) it implies that - in the escitalopram group - the relationship
413
between brain function and the clinical outcome (QIDS) is stronger when patients have
414
greater levels of emotional blunting (i.e. lower scores on the LEIS). LEIS scores were
415
relatively increased in the psilocybin group, meaning that the significant direct effect of the
416
LEIS on clinical outcome (Z = -2.24) implies that a higher level of emotional function post-
417
psilocybin-therapy relates to a greater post-treatment reduction in depression scores (i.e.
418
greater clinical effect) after the psilocybin-therapy. These analyses were also repeated with
419
the BDI as the dependent variable and showed a similar pattern of results; see
420
supplementary material.
421
422
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
18
423
Figure 5. Results of exploratory moderation analyses examining the effects of the
424
change in brain activity on the primary clinical outcome (QIDS) and its moderation by
425
a measure of emotional function (LEIS). In the psilocybin group (top) emotional
426
function has a significant effect on clinical outcomes, but there is no significant effect
427
of brain activity and no interaction effect. In the escitalopram group (bottom) the
428
change in brain activity has a robust effect on clinical outcome, and emotional
429
function also has a strong moderating effect, supporting mechanistic assumptions
430
about the action of the SSRI. Significant effects are denoted by a green arrow and *.
431
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
19
Discussion
432
433
Consistent with our primary hypothesis for this comparative mechanisms trial, we found
434
significant differences in brain responsiveness to emotional stimuli after escitalopram
435
compared with psilocybin for depression. The separate examination of treatment effects
436
revealed that the between-group differences were largely driven by decreases in
437
responsiveness post-treatment in the escitalopram group. Null, or opposite (i.e., slightly
438
increased activation) effects were seen in the psilocybin group, depending on the exact face
439
type i.e., increased activations post-treatment to neutral faces was seen in the whole brain
440
analysis. Results were generally consistent whether examining whole brain activations or
441
focusing specifically on the amygdala. Exploratory moderation analyses suggested that
442
outcomes were better predicted by the change in brain activity, moderated by changes in
443
emotional function, in the escitalopram group, whereas clinical outcomes were better
444
predicted by changes in emotional function in the psilocybin group.
445
446
This study’s findings lend support to the view that psilocybin-therapy and SSRIs have distinct
447
therapeutic mechanisms of action 9. While both drugs act on the 5-HT system, SSRIs
448
increase synaptic concentrations of 5-HT by blocking its reuptake, whereas psilocybin
449
(through its main active metabolite, psilocin) acts as a direct agonist on certain 5-HT
450
receptors, with a key action on the 5-HT2A receptor subtype 26,55. Increased synaptic 5-HT
451
via SSRIs should affect all available 5-HT receptors but there is some evidence to suggest
452
that the activation of inhibitory postsynaptic 5-HT1A receptors, which are heavily expressed
453
in limbic regions 56 and implicated in emotional 57 and sexual functioning 58, play an
454
important role in the action of SSRIs. The four to six week lag in antidepressant effects with
455
SSRIs is thought to be due to a gradual down-regulation of inhibitory 5-HT1A pre-synaptic
456
auto-receptors 59, allowing a slow disinhibition of serotonergic efflux onto postsynaptic
457
targets. Although speculative, activating inhibitory postsynaptic 5-HT1A receptors in limbic
458
regions may dampen their responsiveness and account for the emotional blunting and loss
459
of sexual function described by some patients receiving treatment with SSRIs 7. The
460
moderation analyses provided here suggest that this reduction in brain responsiveness to
461
emotion with SSRIs may be an important factor in their therapeutic action.
462
463
The results of the whole-brain between-groups contrast revealed a pattern of reduced
464
responsiveness under escitalopram in regions including the DLPFC, temporo-parietal
465
junction (TPJ), supramarginal gyrus, and secondary somatosensory cortex. These are largely
466
high-level transmodal regions implicated in a broad range of processes, including social-
467
cognitive functions. They are not necessarily regions most commonly identified in emotional
468
face perception 54, however, a recent meta-analysis 60 highlighted a similar set of regions
469
involved in empathy for physical pain, emotional situations, and emotional faces. A further
470
recent meta-analysis of the neural effects of antidepressants as assessed by emotional
471
response paradigms has also identified effects in a wider set of cortical regions than just the
472
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
20
amygdala 10. Although speculative, the relative preservation of responsiveness in these high-
473
level cortical regions after psilocybin therapy may relate to its positive modulation of socio-
474
emotional functioning 61, an assumption that might also align with the greater
475
responsiveness to neutral faces post psilocybin therapy.
476
477
Psilocybin therapy is emerging as a potential paradigm-shifting62,63 treatment for depression
478
and other commonly comorbid disorders64 ; paradigm-shifting because it is arguably the first
479
truly effective drug-assisted psychotherapy41. This may be because psilocybin enhances
480
affective psychotherapeutic processes in a different way to other compounds that have
481
been combined with psychotherapy 65 including SSRIs66. Psilocybin therapy appears to have
482
an efficacy at least comparable to established treatments20,22,41, with treatment response
483
rates of approximately 70% in three recent trials in MDD21,22,67, and has a favorable side-
484
effect and patient acceptance profile. The present finding of a robust difference in the
485
effects of an SSRI and psilocybin therapy on brain responsiveness to emotional stimuli are
486
consistent with previous assumptions about their differential therapeutic actions9, as well as
487
this trial’s pre-registered primary mechanistic hypothesis
488
(https://clinicaltrials.gov/ct2/show/NCT03429075).
489
490
A generalized emotional blunting is often associated with SSRIs5, whereas greater
491
acceptance of emotions and a multi-faceted sense of reconnection39,68 is commonplace with
492
psilocybin-therapy. The differing pattern of effects in the exploratory moderation analyses
493
presented here also support this perspective. In the psilocybin group, the (relatively
494
increased) emotional function was shown to have a direct positive effect on clinical
495
outcome, such that higher levels of emotional function were associated with greater
496
improvements in clinical symptoms. In the escitalopram group however, the effect of
497
changes in emotional function was a robust moderator of the effect of brain responses on
498
the clinical outcome, but in an opposite direction. The relationship between brain function
499
(decreased responsiveness to faces) and clinical effects (decreased symptom severity) was
500
strongest for those patients who reported the largest muting of the intensity of their
501
emotions (LEIS). As others have recently noted9,16 this emotional blunting associated with
502
SSRIs may be a key factor in their therapeutic efficacy, and the current findings support this
503
perspective.
504
505
The importance of the 5-HT2A receptor for the action of psychedelics is well established26,
506
as demonstrated for example, via a strong and selective affinity-to-potency relationship 69,
507
antagonist pre-treatment blocking the psychedelic effects70, and most recently, subjective
508
psychedelic effects correlating with degree of 5-HT2A receptor occupation in the human
509
brain71. 5-HT2A receptors are highly expressed throughout the human cortex56,72, but
510
particularly in high-level associative/transmodal regions that undergo marked expansion
511
throughout brain development73 and have expanded most evolutionarily74,75. A growing
512
body of evidence has linked 5-HT2A receptor signaling with increases in a variety of markers
513
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
21
of neuroplasticity76,77. The most recent model of psychedelic therapy’s mechanism of action
514
emphasizes a role for enhanced 5-HT2AR-induced cortical plasticity; opening a window for
515
healthy psychological change, guided by concomitant psychotherapy64,78. In contrast to the
516
effects of SSRIs, suppressed emotional or limbic responsiveness is not a feature of the
517
psychedelic therapy model. Indeed, acute emotional release during the trip79 and then
518
progress towards greater emotional acceptance and ‘reconnection’39 are thought to be
519
important components of the psychedelic therapy model. Together with the present results,
520
replicated findings of increased whole brain functional integrity after psilocybin therapy for
521
depression 30, as well as increased dynamic flexibility 29, imply a different antidepressant
522
action for psilocybin relative to SSRIs.
523
524
Regarding this study’s design, it should be noted that the final post-treatment scan occurred
525
hours after the final dose for those in the escitalopram group (coinciding with peak plasma
526
concentration of the drug) but three weeks after the second of two 25mg psilocybin
527
sessions for the psilocybin group. The smaller changes in post-treatment brain functioning in
528
the psilocybin group may be due to the duration since dosing and a different result may
529
have been found had we scanned closer to the last psilocybin dosing session, as we did in
530
previous work, where we showed a relative increases in amygdala responses one-day post-
531
dosing 28. We believe that the present study’s design can be justified however, as the
532
intention was to capture the enduring antidepressant effects of psilocybin-therapy, and
533
these were robust at the 6-week endpoint (3 weeks after the final psilocybin dosing
534
session)41. Our current findings can be seen as consistent with previous work in which
535
healthy volunteers received a single-dose of psilocybin and reduced amygdala responses
536
were observed at a one-week follow-up, with responses returning to baseline after one
537
month 80. Interestingly, a distinct metric (whole-brain network modularity applied to
538
spontaneous brain activity) has been found to reliably index the antidepressant action of
539
psilocybin-therapy, but not escitalopram30. The implication is that (unlike for SSRIs11) the
540
emotional face paradigm may not be an especially sensitive marker of the antidepressant
541
action of psilocybin-therapy (on this time-scale), whereas other brain indices may be more
542
sensitive to its specific action.
543
544
Some previous work has suggested that SSRIs and related antidepressant drugs have a
545
selective action on the processing of negatively valenced emotional stimuli 12. We did not
546
replicate this result in the whole-brain analysis but we did see suggestions of less
547
suppression of amygdala responses to happy and neutral faces with escitalopram relative to
548
its effect on fearful faces. However, consistent with observations from previous work 81,82,
549
faces of a negative valence evoked the largest amygdala responses in this study (figure 3). In
550
alignment with statistical principles, one could suggest that a drug’s modulatory action
551
would be greatest in the domain of functioning that is especially sensitive under baseline
552
conditions, i.e. in this case, the negatively-valenced domain. It seems plausible therefore
553
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
22
that this could account for SSRI’s apparent biased action on negatively-valenced emotional
554
stimuli as seen in previous studies 12.
555
556
In conclusion, consistent with the primary hypothesis for this trial, fMRI analyses revealed a
557
reduced brain responsiveness to emotional face stimuli after six weeks of daily escitalopram
558
but not three weeks after the second of two 25mg dosing sessions with psilocybin-therapy
559
for major depressive disorder. The escitalopram data is consistent with current theories on
560
the therapeutic action of SSRIs 15 and the comparison with psilocybin-therapy highlights the
561
different brain actions of these two different treatment modalities.
562
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
23
References
563
564
1. Malhi GS, Mann JJ. Depression. The Lancet. 2018;392(10161):2299-2312.
565
doi:10.1016/S0140-6736(18)31948-2
566
2. Davidson JRT. Major Depressive Disorder Treatment Guidelines in America and Europe.
567
The Journal of Clinical Psychiatry. 2010;71(suppl E1). doi:10.4088/jcp.9058se1c.04gry
568
3. Cipriani A, Furukawa TA, Salanti G, et al. Comparative Efficacy and Acceptability of 21
569
Antidepressant Drugs for the Acute Treatment of Adults With Major Depressive
570
Disorder: A Systematic Review and Network Meta-Analysis. Focus. 2018;16(4):420-429.
571
doi:10.1176/appi.focus.16407
572
4. Machado M, Iskedjian M, Ruiz I, Einarson TR. Remission, dropouts, and adverse drug
573
reaction rates in major depressive disorder: a meta-analysis of head-to-head trials. Curr
574
Med Res Opin. 2006;22(9):1825-1837. doi:10.1185/030079906X132415
575
5. Goodwin GM, Price J, De Bodinat C, Laredo J. Emotional blunting with antidepressant
576
treatments: A survey among depressed patients. Journal of Affective Disorders.
577
2017;221(June):31-35. doi:10.1016/j.jad.2017.05.048
578
6. Opbroek A, Delgado PL, Laukes C, et al. Emotional blunting associated with SSRI-induced
579
sexual dysfunction. Do SSRIs inhibit emotional responses? International Journal of
580
Neuropsychopharmacology. 2002;5(2):147-151. doi:10.1017/S1461145702002870
581
7. Cascade E, Kalali AH, Kennedy SH. Real-world data on SSRI antidepressant side effects.
582
Psychiatry. 2009;6(2):16. Accessed March 26, 2021. www.iGuard.org
583
8. Cao B, Zhu J, Zuckerman H, et al. Pharmacological interventions targeting anhedonia in
584
patients with major depressive disorder: A systematic review. Progress in Neuro-
585
Psychopharmacology and Biological Psychiatry. 2019;92(December 2018):109-117.
586
doi:10.1016/j.pnpbp.2019.01.002
587
9. Carhart-Harris RL, Nutt DJ. Serotonin and brain function: A tale of two receptors. Journal
588
of Psychopharmacology. 2017;31(9):1091-1120. doi:10.1177/0269881117725915
589
10. Ma Y. Neuropsychological mechanism underlying antidepressant effect: A systematic
590
meta-analysis. Molecular Psychiatry. 2015;20(3):311-319. doi:10.1038/mp.2014.24
591
11. Godlewska BR, Norbury R, Selvaraj S, Cowen PJ, Harmer CJ. Short-term SSRI treatment
592
normalises amygdala hyperactivity in depressed patients. Psychological Medicine.
593
2012;42(12):2609-2617. doi:10.1017/S0033291712000591
594
12. Godlewska BR, Browning M, Norbury R, Cowen PJ, Harmer CJ. Early changes in
595
emotional processing as a marker of clinical response to SSRI treatment in depression.
596
Translational Psychiatry. 2016;6(11):1-7. doi:10.1038/tp.2016.130
597
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
24
13. Maron E, Wall M, Norbury R, et al. Effect of short-term escitalopram treatment on
598
neural activation during emotional processing. Journal of Psychopharmacology.
599
2016;30(1):33-39. doi:10.1177/0269881115620462
600
14. McCabe C, Mishor Z, Cowen PJ, Harmer CJ. Diminished Neural Processing of Aversive
601
and Rewarding Stimuli During Selective Serotonin Reuptake Inhibitor Treatment.
602
Biological Psychiatry. 2010;67(5):439-445. doi:10.1016/j.biopsych.2009.11.001
603
15. Harmer CJ, Duman RS, Cowen PJ. How do antidepressants work? New perspectives for
604
refining future treatment approaches. The Lancet Psychiatry. 2017;4(5):409-418.
605
doi:10.1016/S2215-0366(17)30015-9
606
16. Langley C, Armand S, Luo Q, et al. Chronic escitalopram in healthy volunteers has
607
specific effects on reinforcement sensitivity: a double-blind, placebo-controlled semi-
608
randomised study. Neuropsychopharmacol. Published online January 23, 2023:1-7.
609
doi:10.1038/s41386-022-01523-x
610
17. Nutt D, Erritzoe D, Carhart-Harris R. Psychedelic Psychiatry’s Brave New World. Cell.
611
2020;181(1):24-28. doi:10.1016/j.cell.2020.03.020
612
18. Carhart-Harris RL, Goodwin GM. The Therapeutic Potential of Psychedelic Drugs: Past,
613
Present, and Future. Neuropsychopharmacology. 2017;42(11):2105-2113.
614
doi:10.1038/npp.2017.84
615
19. Krystal JH, Abdallah CG, Sanacora G, Charney DS, Duman RS. Ketamine: A Paradigm Shift
616
for Depression Research and Treatment. Neuron. 2019;101(5):774-778.
617
doi:10.1016/j.neuron.2019.02.005
618
20. Carhart-Harris RL, Bolstridge M, Rucker J, et al. Psilocybin with psychological support for
619
treatment-resistant depression: an open-label feasibility study. The Lancet Psychiatry.
620
2016;3(7):619-627. doi:10.1016/S2215-0366(16)30065-7
621
21. Carhart-Harris R, Giribaldi B, Watts R, et al. Trial of Psilocybin versus Escitalopram for
622
Depression. New England Journal of Medicine. 2021;384(15):1402-1411.
623
doi:10.1056/nejmoa2032994
624
22. Davis AK, Barrett FS, May DG, et al. Effects of Psilocybin-Assisted Therapy on Major
625
Depressive Disorder: A Randomized Clinical Trial. JAMA Psychiatry. Published online
626
2020:1-9. doi:10.1001/jamapsychiatry.2020.3285
627
23. Griffiths RR, Johnson MW, Carducci MA, et al. Psilocybin produces substantial and
628
sustained decreases in depression and anxiety in patients with life-threatening cancer: A
629
randomized double-blind trial. Journal of Psychopharmacology. 2016;30(12):1181-1197.
630
doi:10.1177/0269881116675513
631
24. Ross S, Bossis A, Guss J, et al. Rapid and sustained symptom reduction following
632
psilocybin treatment for anxiety and depression in patients with life-threatening cancer:
633
A randomized controlled trial. Journal of Psychopharmacology. 2016;30(12):1165-1180.
634
doi:10.1177/0269881116675512
635
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
25
25. Grob CS, Danforth AL, Chopra GS, et al. Pilot study of psilocybin treatment for anxiety in
636
patients with advanced-stage cancer. Archives of General Psychiatry. 2011;68(1):71-78.
637
doi:10.1001/archgenpsychiatry.2010.116
638
26. Nichols DE. Psychedelics. 2016;(April):264-355.
639
27. Carhart-Harris RL, Erritzoe D, Williams T, et al. Neural correlates of the psychedelic state
640
as determined by fMRI studies with psilocybin. Proceedings of the National Academy of
641
Sciences of the United States of America. 2012;109(6):2138-2143.
642
doi:10.1073/pnas.1119598109
643
28. Carhart-Harris RL, Roseman L, Bolstridge M, et al. Psilocybin for treatment-resistant
644
depression: fMRI-measured brain mechanisms. Scientific Reports. 2017;7(1):13187.
645
doi:10.1038/s41598-017-13282-7
646
29. Doss MK, Považan M, Rosenberg MD, et al. Psilocybin therapy increases cognitive and
647
neural flexibility in patients with major depressive disorder. Translational Psychiatry.
648
2021;11(1):1-10. doi:10.1038/s41398-021-01706-y
649
30. Daws RE, Timmermann C, Giribaldi B, et al. Increased global integration in the brain
650
after psilocybin therapy for depression. Nature Medicine. Published online 2022.
651
doi:10.1038/s41591-022-01744-z
652
31. McCulloch DEW, Madsen MK, Stenbæk DS, et al. Lasting effects of a single psilocybin
653
dose on resting-state functional connectivity in healthy individuals. J Psychopharmacol.
654
2022;36(1):74-84. doi:10.1177/02698811211026454
655
32. Pasquini L, Palhano-Fontes F, Araujo DB. Subacute effects of the psychedelic ayahuasca
656
on the salience and default mode networks. J Psychopharmacol. 2020;34(6):623-635.
657
doi:10.1177/0269881120909409
658
33. Watts R, Luoma JB. The use of the psychological flexibility model to support psychedelic
659
assisted therapy. Journal of Contextual Behavioral Science. 2020;15(April 2019):92-102.
660
doi:10.1016/j.jcbs.2019.12.004
661
34. Groenewold NA, Opmeer EM, de Jonge P, Aleman A, Costafreda SG. Emotional valence
662
modulates brain functional abnormalities in depression: Evidence from a meta-analysis
663
of fMRI studies. Neuroscience & Biobehavioral Reviews. 2013;37(2):152-163.
664
doi:10.1016/j.neubiorev.2012.11.015
665
35. Roseman L, Demetriou L, Wall MB, Nutt DJ, Carhart-Harris RL. Increased amygdala
666
responses to emotional faces after psilocybin for treatment-resistant depression.
667
Neuropharmacology. 2018.
668
36. Majić T, Schmidt TT, Gallinat J. Peak experiences and the afterglow phenomenon: When
669
and how do therapeutic effects of hallucinogens depend on psychedelic experiences?
670
Journal of Psychopharmacology. 2015;29(3):241-253. doi:10.1177/0269881114568040
671
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
26
37. Murphy SE, Norbury R, O’Sullivan U, Cowen PJ, Harmer CJ. Effect of a single dose of
672
citalopram on amygdala response to emotional faces. The British journal of psychiatry :
673
the journal of mental science. 2009;194(6):535-540. doi:10.1192/bjp.bp.108.056093
674
38. Mertens LJ, Wall MB, Roseman L, Demetriou L, Nutt DJ, Carhart-Harris RL. Therapeutic
675
mechanisms of psilocybin: Changes in amygdala and prefrontal functional connectivity
676
during emotional processing after psilocybin for treatment-resistant depression. Journal
677
of Psychopharmacology. Published online 2020:1-14. doi:10.1177/0269881119895520
678
39. Watts R, Day C, Krzanowski J, Nutt D, Carhart-Harris R. Patients’ Accounts of Increased
679
“Connectedness” and “Acceptance” After Psilocybin for Treatment-Resistant
680
Depression. Journal of Humanistic Psychology. 2017;57(5):520-564.
681
doi:10.1177/0022167817709585
682
40. Cipriani A, Santilli C, Furukawa TA, et al. Escitalopram versus other antidepressive agents
683
for depression. Cochrane Database of Systematic Reviews. 2009;(2).
684
doi:10.1002/14651858.CD006532.pub2
685
41. Carhart-Harris R, Giribaldi B, Watts R, et al. Trial of Psilocybin versus Escitalopram for
686
Depression. New England Journal of Medicine. 2021;384(15):1402-1411.
687
doi:10.1056/nejmoa2032994
688
42. Rush AJ, Trivedi MH, Ibrahim HM, et al. The 16-item Quick Inventory of Depressive
689
Symptomatology (QIDS), clinician rating (QIDS-C), and self-report (QIDS-SR): A
690
psychometric evaluation in patients with chronic major depression. Biological
691
Psychiatry. 2003;54(5):573-583. doi:10.1016/S0006-3223(02)01866-8
692
43. Beck AT, Steer RA, Carbin MG. Psychometric properties of the Beck Depression
693
Inventory: Twenty-five years of evaluation. Clinical Psychology Review. 1988;8(1):77-
694
100. doi:10.1016/0272-7358(88)90050-5
695
44. Tennant R, Hiller L, Fishwick R, et al. The Warwick-Edinburgh Mental Well-being Scale
696
(WEMWBS): development and UK validation. Health and Quality of Life Outcomes.
697
2007;5(1):63. doi:10.1186/1477-7525-5-63
698
45. Snaith RP, Hamilton M, Morley S, Humayan A, Hargreaves D, Trigwell P. A scale for the
699
assessment of hedonic tone the Snaith-Hamilton Pleasure Scale. Br J Psychiatry.
700
1995;167(1):99-103. doi:10.1192/bjp.167.1.99
701
46. Montejo AL, García M, Espada M, Rico-Villademoros F, Llorca G, Izquierdo JA.
702
[Psychometric characteristics of the psychotropic-related sexual dysfunction
703
questionnaire. Spanish work group for the study of psychotropic-related sexual
704
dysfunctions]. Actas Esp Psiquiatr. 2000;28(3):141-150.
705
47. Goeleven E, De Raedt R, Leyman L, Verschuere B. The Karolinska Directed Emotional
706
Faces: A validation study. Cognition & Emotion. 2008;22(6):1094-1118.
707
doi:10.1080/02699930701626582
708
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
27
48. Jack CR, Bernstein MA, Fox NC, et al. The Alzheimer’s Disease Neuroimaging Initiative
709
(ADNI): MRI methods. Journal of Magnetic Resonance Imaging. 2008;27(4):685-691.
710
doi:10.1002/jmri.21049
711
49. Cauley SF, Polimeni JR, Bhat H, Wald LL, Setsompop K. Interslice leakage artifact
712
reduction technique for simultaneous multislice acquisitions. Magnetic Resonance in
713
Medicine. 2014;72(1):93-102. doi:10.1002/mrm.24898
714
50. Setsompop K, Gagoski BA, Polimeni JR, Witzel T, Wedeen VJ, Wald LL. Blipped-controlled
715
aliasing in parallel imaging for simultaneous multislice echo planar imaging with reduced
716
g-factor penalty. Magnetic Resonance in Medicine. 2012;67(5):1210-1224.
717
doi:10.1002/mrm.23097
718
51. Xu J, Moeller S, Auerbach EJ, et al. Evaluation of slice accelerations using multiband echo
719
planar imaging at 3T. NeuroImage. 2013;83:991-1001.
720
doi:10.1016/j.neuroimage.2013.07.055
721
52. Demetriou L, Kowalczyk OS, Tyson G, Bello T, Newbould RD, Wall MB. A comprehensive
722
evaluation of increasing temporal resolution with multiband-accelerated sequences and
723
their effects on statistical outcome measures in fMRI. NeuroImage. 2018;176:404-416.
724
53. Smith SM, Jenkinson M, Woolrich MW, et al. Advances in functional and structural MR
725
image analysis and implementation as FSL. Neuroimage. 2004;23:S208-S219.
726
54. Fusar-Poli P, Placentino A, Carletti F, et al. Functional atlas of emotional faces
727
processing: a voxel-based meta-analysis of 105 functional magnetic resonance imaging
728
studies. Journal of psychiatry & neuroscience : JPN. 2009;34(6):418-432.
729
55. McClure-Begley TD, Roth BL. The promises and perils of psychedelic pharmacology for
730
psychiatry. Nature reviews Drug discovery. Published online 2022. doi:10.1038/s41573-
731
022-00421-7
732
56. Beliveau V, Ganz M, Feng XL, et al. A High-Resolution In Vivo Atlas of the Human Brain’s
733
Serotonin System. Published online 2017. doi:10.1523/JNEUROSCI.2830-16.2016
734
57. Phan KL, Wager T, Taylor SF, Liberzon I. Functional neuroanatomy of emotion: a meta-
735
analysis of emotion activation studies in PET and fMRI. NeuroImage. 2002;16(2):331-
736
348. doi:10.1006/nimg.2002.1087
737
58. Comninos AN, Wall MB, Demetriou L, et al. Kisspeptin modulates sexual and emotional
738
brain processing in humans. Journal of Clinical Investigation. 2017;127(2):709-719.
739
59. Celada P, Puig MV, Amargós-Bosch M, Adell A, Artigas F. The therapeutic role of 5-HT1A
740
and 5-HT2A receptors in depression. In: Journal of Psychiatry and Neuroscience. Vol 29.
741
Canadian Medical Association; 2004:252-265. Accessed March 29, 2021.
742
/pmc/articles/PMC446220/
743
60. Ding R, Ren J, Li S, Zhu X, Zhang K, Luo W. Domain-general and domain-preferential
744
neural correlates underlying empathy towards physical pain, emotional situation and
745
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
28
emotional faces: An ALE meta-analysis. Neuropsychologia. 2020;137:107286.
746
doi:10.1016/j.neuropsychologia.2019.107286
747
61. Preller KH, Pokorny T, Hock A, et al. Effects of serotonin 2A/1A receptor stimulation on
748
social exclusion processing. Proceedings of the National Academy of Sciences of the
749
United States of America. 2016;113(18):5119-5124. doi:10.1073/pnas.1524187113
750
62. Schenberg EE. Psychedelic-assisted psychotherapy: A paradigm shift in psychiatric
751
research and development. Frontiers in Pharmacology. 2018;9(JUL):1-11.
752
doi:10.3389/fphar.2018.00733
753
63. Carhart-Harris RL. Translational Challenges in Psychedelic Medicine. N Engl J Med.
754
2023;388(5):476-477. doi:10.1056/NEJMcibr2213109
755
64. Carhart-Harris RL, Chandaria S, Erritzoe DE, et al. Canalization and plasticity in
756
psychopathology. Neuropharmacology. 2023;226:109398.
757
doi:10.1016/j.neuropharm.2022.109398
758
65. G. Hofmann S, T. Sawyer A, Asnaani A. D-Cycloserine as an Augmentation Strategy for
759
Cognitive Behavioral Therapy for Anxiety Disorders: An Update. Current Pharmaceutical
760
Design. 2012;18(35):5659-5662. doi:10.2174/138161212803530916
761
66. March JS. Fluoxetine, cognitive-behavioral therapy, and their combination for
762
adolescents with depression: Treatment for Adolescents with Depression Study (TADS)
763
randomized controlled trial. Journal of the American Medical Association.
764
2004;292(7):807-820. doi:10.1001/jama.292.7.807
765
67. Sloshower J, Skosnik PD, Safi-Aghdam H, et al. Psilocybin-assisted therapy for major
766
depressive disorder: An exploratory placebo-controlled, fixed-order trial. J
767
Psychopharmacol. Published online March 20, 2023:2698811231154852.
768
doi:10.1177/02698811231154852
769
68. Watts R, Kettner H, Geerts D, et al. The Watts Connectedness Scale: a new scale for
770
measuring a sense of connectedness to self, others, and world. Psychopharmacology.
771
Published online August 8, 2022. doi:10.1007/s00213-022-06187-5
772
69. Glennon RA, Titeler M, McKenney JD. Evidence for 5-HT2 involvement in the mechanism
773
of action of hallucinogenic agents. Life Sciences. 1984;35(25):2505-2511.
774
doi:10.1016/0024-3205(84)90436-3
775
70. Vollenweider FX, Vollenweider-Scherpenhuyzen MFI, Bäbler A, Vogel H, Hell D.
776
Psilocybin induces schizophrenia-like psychosis in humans via a serotonin-2 agonist
777
action. NeuroReport. 1998;9(17):3897-3902. doi:10.1097/00001756-199812010-00024
778
71. Madsen MK, Fisher PM, Burmester D, et al. Psychedelic effects of psilocybin correlate
779
with serotonin 2 A receptor occupancy and plasma psilocin levels.
780
Neuropsychopharmacology. Published online 2019. doi:10.1038/s41386-019-0324-9
781
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
29
72. Erritzoe D, Ashok AH, Searle GE, et al. Serotonin release measured in the human brain: a
782
PET study with [11C]CIMBI-36 and d-amphetamine challenge.
783
Neuropsychopharmacology. 2020;45(5):804-810. doi:10.1038/s41386-019-0567-5
784
73. Hill J, Inder T, Neil J, Dierker D, Harwell J, Van Essen D. Similar patterns of cortical
785
expansion during human development and evolution. Proceedings of the National
786
Academy of Sciences of the United States of America. 2010;107(29):13135-13140.
787
doi:10.1073/pnas.1001229107
788
74. Rilling JK. Comparative primate neuroimaging: Insights into human brain evolution.
789
Trends in Cognitive Sciences. 2014;18(1):46-55. doi:10.1016/j.tics.2013.09.013
790
75. Van Essen DC, Dierker DL. Surface-based and probabilistic atlases of primate cerebral
791
cortex. Neuron. 2007;56(2):209-225. doi:10.1016/j.neuron.2007.10.015
792
76. Ly C, Greb AC, Cameron LP, et al. Psychedelics Promote Structural and Functional Neural
793
Plasticity. Cell Reports. 2018;23(11):3170-3182. doi:10.1016/j.celrep.2018.05.022
794
77. Vaidya VA, Marek GJ, Aghajanian GK, Duman RS. 5-HT(2A) receptor-mediated regulation
795
of brain-derived neurotrophic factor mRNA in the hippocampus and the neocortex.
796
Journal of Neuroscience. 1997;17(8):2785-2795. doi:10.1523/jneurosci.17-08-
797
02785.1997
798
78. Carhart-Harris RL. How do psychedelics work? Current opinion in psychiatry.
799
2019;32(1):16-21. doi:10.1097/YCO.0000000000000467
800
79. Roseman L, Haijen E, Idialu-Ikato K, Kaelen M, Watts R, Carhart-Harris R. Emotional
801
breakthrough and psychedelics: Validation of the Emotional Breakthrough Inventory.
802
Journal of Psychopharmacology. 2019;33(9):1076-1087.
803
doi:10.1177/0269881119855974
804
80. Barrett FS, Doss MK, Sepeda ND, Pekar JJ, Griffiths RR. Emotions and brain function are
805
altered up to one month after a single high dose of psilocybin. Scientific Reports.
806
2020;10(1):1-14. doi:10.1038/s41598-020-59282-y
807
81. Zald DH. The human amygdala and the emotional evaluation of sensory stimuli. Brain
808
research Brain research reviews. 2003;41(1):88-123.
809
82. Todorov A. The role of the amygdala in face perception and evaluation. Motiv Emot.
810
2012;36(1):16-26. doi:10.1007/s11031-011-9238-5
811
812
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
30
Supplementary material
813
814
Head movement
815
816
Overall head-motion characteristics for all subjects were excellent. Assessment of subject
817
head-motion during the scans was performed using the displacement time-series produced
818
during preprocessing of the functional data. These were collated, and means were
819
calculated across the time-series and across the six (three translations, three rotations)
820
displacement parameters. Maximum displacement values were also assessed.
821
822
Only one subject showed a maximum displacement value greater than one voxel dimension
823
(3mm) on a single parameter, for one study visit. Inspection of this subject’s time series
824
showed a single large (~3mm) shift in position part-way through the scan. This subject’s
825
mean displacement (0.088mm) was less than 0.1mm, and was comparable to other subjects
826
in the sample. It was therefore judged to be acceptable, and the subject was included in the
827
analyses. Mean displacement across all subjects, scan sessions, and parameters was -
828
0.00058mm (SD = 0.027mm).
829
830
A 2 (treatment group) by 2 (study visit) mixed-model ANOVA was conducted to examine any
831
systematic differences in head-motion across these factors. There was no main effect of
832
treatment group (F[1,44] = 1.84, p = 0.182) and no main effect of study visit (F[1,44] = 0.11,
833
p = 0.740. There was also no interaction effect (F[1,44] = 0.077, p = 0.782).
834
835
836
837
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
31
Group-level task results
838
839
840
Figure S1. Results from the validation analyses of mean task effects in each group.
841
842
843
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
32
Correlation results
844
845
Correlation of BOLD* data with:
Pearson’s r
p value
Psilocybin group (N=25)
BDI change (baseline to six weeks)
0.132
0.529
WEMWBS change (baseline to 6 weeks)
-0.001
0.997
PRSEXDQ
-0.041
0.846
LEIS change (baseline to 6 weeks)
-0.07
0.762
SHAPS change (baseline to 6 weeks)
-0.001
0.998
QIDS change (baseline to 6 weeks)
0.084
0.718
Escitalopram group (N=21)
BDI change (baseline to six weeks)
0.075
0.748
WEMWBS change (baseline to 6 weeks)
-0.001
0.997
PRSEXDQ
-0.077
0.741
LEIS change (baseline to 6 weeks)
-0.022
0.927
SHAPS change (baseline to 6 weeks)
0.037
0.878
QIDS change (baseline to 6 weeks)
0.187
0.43
*All faces vs. baseline contrast, pre-treatment vs. post-treatment
846
847
Table S1. Correlation analyses between a summary measure of brain activity and key clinical
848
measures. All analyses showed non-significant/null results.
849
850
851
852
Moderation Analyses with BDI as the dependent variable
853
854
The moderation analyses were repeated with BDI change scores (baseline to six-week
855
follow-up) as the dependent variable. For the psilocybin group, this produced the same
856
pattern of results as the analysis with QIDS scores in the main text. There was a significant (Z
857
= -3.02, p = 0.003) effect of the LEIS measure on BDI scores, but no effect of brain activity (Z
858
= -0.29, p = 0.774) and no interaction effect (Z = 1.32, p = 0.187). In the escitalopram group
859
there were no main effects of either brain activity (Z = 1.49, p = 0.137) or LEIS (Z = -0.59, p =
860
0.551) on BDI change scores, but a significant interaction effect (Z = 2.38, p = 0.018).
861
. CC-BY-NC-ND 4.0 International licenseIt is made available under a
is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. (which was not certified by peer review) The copyright holder for this preprint this version posted June 3, 2023. ; https://doi.org/10.1101/2023.05.29.23290667doi: medRxiv preprint
... 7,8 See also some recent neuroimaging evidence of the neurobiological action of ET in this trial. 67 Although we interpret no difference between conditions in antidepressant response, we note recent evidence of potential psychometric problems with our primary outcome measure (QIDS-SR-16) and how the QIDS-SR-16 data assessed at the 6 week end-point differed from three other measures (BDI, MADRS, and HAM-D) as well as a post-hoc generated depression factor using factors from all four measures. 27 These problems and psychometric differences may have contributed to a null between-condition difference at 6 weeks and during follow-up. ...
Article
Background Psilocybin therapy (PT) produces rapid and persistent antidepressant effects in major depressive disorder (MDD). However, the long-term effects of PT have never been compared with gold-standard treatments for MDD such as pharmacotherapy or psychotherapy alone or in combination. Methods This is a 6-month follow-up study of a phase 2, double-blind, randomised, controlled trial involving patients with moderate-to-severe MDD. Participants were recruited from a hospital in the UK. Male or female patients with major depressive disorder (DSM-IV), moderate to severe depression (HAM-D ≥17), no MRI or SSRI contraindications, confirmed diagnosis by a GP or mental healthcare professional, aged 18–80, and competent in English were eligible. Patients were randomly assigned (1:1) to receive either two 25 mg doses of the psychedelic drug psilocybin administered orally combined with psychological support (‘psilocybin therapy’ or PT) and book-ended by further support or a 6-week course of the selective serotonin reuptake inhibitor (SSRI) escitalopram (administered daily at 10 mg for three weeks and 20 mg for the subsequent three weeks) plus matched psychological support (‘escitalopram treatment’ or ET). The primary outcome measure was change from baseline in the score on the 16-item Quick Inventory of Depressive Symptomatology–Self-Report (QIDS-SR-16) at week 6, which has been reported previously. Herein, we present results at the 6-month follow-up time point. Measures of social functioning, connectedness, and meaning in life constituted the study's secondary outcomes during follow-up. Safety in the follow-up period was not assessed. This trial is registered at ClinicalTrials.gov, NCT03429075. Findings Between January 15th, 2019 and March 20th, 2020, 59 patients were enrolled and 30 (11 females [37%] and 19 males [63%]) were assigned to the psilocybin group and 29 (9 females [31%] and 20 males [69%]) to the escitalopram group. 25 participants in the PT group and 21 in the ET group completed the 6-month follow-up. At the 6-month follow-up, both PT and ET conditions yielded sustained improvements in depressive symptom severity. The mean between-condition difference in QIDS-SR-16 scores at 6-months was 1.51 (95% CI: −1.35, 4.38; p = 0.311). Secondary outcomes demonstrated that PT had greater mean between-condition differences in functioning (WSAS: −7.46; 95% CI: −12.4, −2.47; p < 0.001), psychological connectedness (WCS: 11.02; 95% CI: 1.25, 20.83; p = 0.033), and meaning in life (MLQ: 4.86; 95% CI: 0.67, 9.05; p = 0.021) compared to ET. Interpretation Six-week intensive treatments with either psilocybin or escitalopram (with psychological support) for MDD were associated with long-term improvements in depressive symptom severity. The greater degree of improvement in the PT arm at follow-up on psychosocial functioning, meaning in life, and psychological connectedness suggests warrant future research. However, these results are descriptive and should be interpreted with caution. Key limitations of the study include its suboptimal power to detect small but meaningful differences between treatments, missing data, the potential use of additional interventions during the follow-up period, and reliance on self-reported treatment assessments. These factors may affect the interpretation of the study findings and should be considered when evaluating the results.
... It is tempting to speculate that this top-down organization may also be linked to dampened responsivity in stress circuitry under these drugs 66 , allowing easier top-down control. However, to test this speculation would require a new, carefully designed study, for which some progress has already been made 67 . Even so, further work is required to better scrutinize this hypothesis. ...
Article
Full-text available
Effective interventions for neuropsychiatric disorders may work by rebalancing the brain’s functional hierarchical organization. Here we directly investigated the effects of two different serotonergic pharmacological interventions on functional brain hierarchy in major depressive disorder in a two-arm double-blind phase II randomized controlled trial comparing psilocybin therapy (22 patients) with escitalopram (20 patients). Patients with major depressive disorder received either 2 × 25 mg of oral psilocybin, three weeks apart, plus six weeks of daily placebo (‘psilocybin arm’) or 2 × 1 mg of oral psilocybin, three weeks apart, plus six weeks of daily escitalopram (10–20 mg; ‘escitalopram arm’). Resting-state functional magnetic resonance imaging scans were acquired at baseline and three weeks after the second psilocybin dose ( NCT03429075 ). The brain mechanisms were captured by generative effective connectivity, estimated from whole-brain modeling of resting state for each session and patient. Hierarchy was determined for each of these sessions using measures of directedness and trophic levels on the effective connectivity, which captures cycle structure, stability and percolation. The results showed that the two pharmacological interventions created significantly different hierarchical reconfigurations of whole-brain dynamics with differential, opposite statistical effect responses. Furthermore, the use of machine learning revealed significant differential reorganization of brain hierarchy before and after the two treatments. Machine learning was also able to predict treatment response with an accuracy of 0.85 ± 0.04. Overall, the results demonstrate that psilocybin and escitalopram work in different ways for rebalancing brain dynamics in depression. This suggests the hypothesis that neuropsychiatric disorders could be closely linked to the breakdown in regions orchestrating brain dynamics from the top of the hierarchy.
Article
Full-text available
Serotonergic psychedelics hold promise as a treatment modality for various psychiatric disorders and are currently applied in psychedelic-assisted psychotherapy. We investigated the learning effects of the serotonin receptor agonist psilocybin in a probabilistic cue-reward task with emotional cues in the form of neutral or fearful faces, presented either consciously or subconsciously. This study represents the first investigation into reinforcement learning with psilocybin. Across different dosages, psilocybin preserved learning effects and was statistically noninferior compared to placebo, while suggesting a higher exploratory behavior. Notably, the 20 mg group exhibited significantly better learning rates against the placebo group. Psilocybin induced inferior results with subconscious cues compared to placebo, and better results with conscious neutral cues in some conditions. These findings suggest that modulating serotonin signaling in the brain with psilocybin sufficiently preservers reinforcement learning.
Article
Full-text available
Several studies of the effects on cognition of selective serotonin reuptake inhibitors (SSRI), administered either acutely or sub-chronically in healthy volunteers, have found changes in learning and reinforcement outcomes. In contrast, to our knowledge, there have been no studies of chronic effects of escitalopram on cognition in healthy volunteers. This is important in view of its clinical use in major depressive disorder (MDD) and obsessive-compulsive disorder (OCD). Consequently, we aimed to investigate the chronic effect of the SSRI, escitalopram, on measures of ‘cold’ cognition (including inhibition, cognitive flexibility, memory) and ‘hot cognition’ including decision-making and particularly reinforcement learning. The study, conducted at the University of Copenhagen between May 2020 and October 2021, used a double-blind placebo-controlled design with 66 healthy volunteers, semi-randomised to receive either 20 mg of escitalopram (n = 32) or placebo (n = 34), balanced for age, sex and intelligence quotient (IQ) for at least 21 days. Questionnaires, neuropsychological tests and serum escitalopram measures were taken. We analysed group differences on the cognitive measures using linear regression models as well as innovative hierarchical Bayesian modelling of the Probabilistic Reversal Learning (PRL) task. The novel and important finding was that escitalopram reduced reinforcement sensitivity compared to placebo on both the Sequential Model-Based/Model-Free task and the PRL task. We found no other significant group differences on ‘cold’ or ‘hot’ cognition. These findings demonstrate that serotonin reuptake inhibition is involved in reinforcement learning in healthy individuals. Lower reinforcement sensitivity in response to chronic SSRI administration may reflect the ‘blunting’ effect often reported by patients with MDD treated with SSRIs. Trial Registration: NCT04239339.
Article
Full-text available
Rationale A general feeling of disconnection has been associated with mental and emotional suffering. Improvements to a sense of connectedness to self, others and the wider world have been reported by participants in clinical trials of psychedelic therapy. Such accounts have led us to a definition of the psychological construct of ‘connectedness’ as ‘a state of feeling connected to self, others and the wider world’. Existing tools for measuring connectedness have focused on particular aspects of connectedness, such as ‘social connectedness’ or ‘nature connectedness’, which we hypothesise to be different expressions of a common factor of connectedness. Here, we sought to develop a new scale to measure connectedness as a construct with these multiple domains. We hypothesised that (1) our scale would measure three separable subscale factors pertaining to a felt connection to ‘self’, ‘others’ and ‘world’ and (2) improvements in total and subscale WCS scores would correlate with improved mental health outcomes post psychedelic use. Objectives To validate and test the ‘Watts Connectedness Scale’ (WCS). Methods Psychometric validation of the WCS was carried out using data from three independent studies. Firstly, we pooled data from two prospective observational online survey studies. The WCS was completed before and after a planned psychedelic experience. The total sample of completers from the online surveys was N = 1226. Exploratory and confirmatory factor analysis were performed, and construct and criterion validity were tested. A third dataset was derived from a double-blind randomised controlled trial (RCT) comparing psilocybin-assisted therapy (n = 27) with 6 weeks of daily escitalopram (n = 25) for major depressive disorder (MDD), where the WCS was completed at baseline and at a 6-week primary endpoint. Results As hypothesised, factor analysis of all WCS items revealed three main factors with good internal consistency. WCS showed good construct validity. Significant post-psychedelic increases were observed for total connectedness scores (η2 = 0.339, p < 0.0001), as well as on each of its subscales (p < 0.0001). Acute measures of ‘mystical experience’, ‘emotional breakthrough’, and ‘communitas’ correlated positively with post-psychedelic changes in connectedness (r = 0.42, r = 0.38, r = 0.42, respectively, p < 0.0001). In the RCT, psilocybin therapy was associated with greater increases in WCS scores compared with the escitalopram arm (ηp2 = 0.133, p = 0.009). Conclusions The WCS is a new 3-dimensional index of felt connectedness that may sensitively measure therapeutically relevant psychological changes post-psychedelic use. We believe that the operational definition of connectedness captured by the WCS may have broad relevance in mental health research.
Article
Full-text available
Psilocybin therapy shows antidepressant potential, but its therapeutic actions are not well understood. We assessed the subacute impact of psilocybin on brain function in two clinical trials of depression. The first was an open-label trial of orally administered psilocybin (10 mg and 25 mg, 7 d apart) in patients with treatment-resistant depression. Functional magnetic resonance imaging (fMRI) was recorded at baseline and 1 d after the 25-mg dose. Beck’s depression inventory was the primary outcome measure (MR/J00460X/1). The second trial was a double-blind phase II randomized controlled trial comparing psilocybin therapy with escitalopram. Patients with major depressive disorder received either 2 × 25 mg oral psilocybin, 3 weeks apart, plus 6 weeks of daily placebo (‘psilocybin arm’) or 2 × 1 mg oral psilocybin, 3 weeks apart, plus 6 weeks of daily escitalopram (10–20 mg) (‘escitalopram arm’). fMRI was recorded at baseline and 3 weeks after the second psilocybin dose (NCT03429075). In both trials, the antidepressant response to psilocybin was rapid, sustained and correlated with decreases in fMRI brain network modularity, implying that psilocybin’s antidepressant action may depend on a global increase in brain network integration. Network cartography analyses indicated that 5-HT2A receptor-rich higher-order functional networks became more functionally interconnected and flexible after psilocybin treatment. The antidepressant response to escitalopram was milder and no changes in brain network organization were observed. Consistent efficacy-related brain changes, correlating with robust antidepressant effects across two studies, suggest an antidepressant mechanism for psilocybin therapy: global increases in brain network integration. The antidepressant response to psilocybin in individuals with treatment-resistant depression is distinct from escitalopram and depends on a global increase in brain network integration.
Article
Full-text available
Psilocybin has shown promise for the treatment of mood disorders, which are often accompanied by cognitive dysfunction including cognitive rigidity. Recent studies have proposed neuropsychoplastogenic effects as mechanisms underlying the enduring therapeutic effects of psilocybin. In an open-label study of 24 patients with major depressive disorder, we tested the enduring effects of psilocybin therapy on cognitive flexibility (perseverative errors on a set-shifting task), neural flexibility (dynamics of functional connectivity or dFC via functional magnetic resonance imaging), and neurometabolite concentrations (via magnetic resonance spectroscopy) in brain regions supporting cognitive flexibility and implicated in acute psilocybin effects (e.g., the anterior cingulate cortex, or ACC). Psilocybin therapy increased cognitive flexibility for at least 4 weeks post-treatment, though these improvements were not correlated with the previously reported antidepressant effects. One week after psilocybin therapy, glutamate and N-acetylaspartate concentrations were decreased in the ACC, and dFC was increased between the ACC and the posterior cingulate cortex (PCC). Surprisingly, greater increases in dFC between the ACC and PCC were associated with less improvement in cognitive flexibility after psilocybin therapy. Connectome-based predictive modeling demonstrated that baseline dFC emanating from the ACC predicted improvements in cognitive flexibility. In these models, greater baseline dFC was associated with better baseline cognitive flexibility but less improvement in cognitive flexibility. These findings suggest a nuanced relationship between cognitive and neural flexibility. Whereas some enduring increases in neural dynamics may allow for shifting out of a maladaptively rigid state, larger persisting increases in neural dynamics may be of less benefit to psilocybin therapy.
Article
Full-text available
Background Psilocybin is a psychedelic drug that has shown lasting positive effects on clinical symptoms and self-reported well-being following a single dose. There has been little research into the long-term effects of psilocybin on brain connectivity in humans. Aim Evaluate changes in resting-state functional connectivity (RSFC) at 1 week and 3 months after one psilocybin dose in 10 healthy psychedelic-naïve volunteers and explore associations between change in RSFC and related measures. Methods Participants received 0.2–0.3 mg/kg psilocybin in a controlled setting. Participants completed resting-state functional magnetic resonance imaging (fMRI) scans at baseline, 1-week and 3-month post-administration and [11C]Cimbi-36 PET scans at baseline and 1 week. We examined changes in within-network, between-network and region-to-region RSFC. We explored associations between changes in RSFC and psilocybin-induced phenomenology as well as changes in psychological measures and neocortex serotonin 2A receptor binding. Results Psilocybin was well tolerated and produced positive changes in well-being. At 1 week only, executive control network (ECN) RSFC was significantly decreased (Cohen’s d = −1.73, pFWE = 0.010). We observed no other significant changes in RSFC at 1 week or 3 months, nor changes in region-to-region RSFC. Exploratory analyses indicated that decreased ECN RSFC at 1 week predicted increased mindfulness at 3 months ( r = −0.65). Conclusions These findings in a small cohort indicate that psilocybin affects ECN function within the psychedelic ‘afterglow’ period. Our findings implicate ECN modulation as mediating psilocybin-induced, long-lasting increases in mindfulness. Although our findings implicate a neural pathway mediating lasting psilocybin effects, it is notable that changes in neuroimaging measures at 3 months, when personality changes are observed, remain to be identified.
Article
Full-text available
Psychedelic assisted therapy comprises three stages: Preparation, Psychedelic Session, and Integration. Preparation is key for maximising the potential of a beneficial psychedelic experience and integration is important for prolonging improvements. The psychological flexibility model (PFM) appears to be a promising one to guide psychedelic preparation and integration. This paper proposes a model that utilises the PFM as informed by a previously published qualitative study of patient accounts of change processes in psilocybin therapy that identified themes of acceptance and connection as associated with positive outcomes. This new model, the ACE (Accept, Connect, Embody) model presents the six psychological flexibility processes, renamed and rearranged in an acceptance triad (defusion, present moment focus, willingness) and a connection triad (self as context, values, committed action). This paper describes the ACE model and how it is being used in an ongoing trial of psilocybin treatment for major depression. It also describes qualitative evidence supportive of the idea that psychological flexibility may be key to characterizing the processes of change involved in psilocybin assisted therapy for depression. These and other results suggest that psilocybin may be specifically increasing psychological flexibility and point to the possibility that psychotherapy approaches incorporating the PFM may serve as a means to deepen and extend the benefits of psilocybin treatment, thus bridging the experiential gap between a potent inner experience and an outer life better lived.
Article
Background: Several early phase studies have demonstrated that psilocybin-assisted therapy has rapid-acting and persisting antidepressant effects from just one or two doses. However, methodological limitations (e.g., placebo-control, blinding) limit interpretability of the existing literature. Methods: In an exploratory placebo-controlled, within-subject, fixed-order study, individuals with moderate to severe major depressive disorder were administered placebo (n = 19) followed by psilocybin (0.3 mg/kg) (n = 15) 4 weeks later. Dosing sessions were embedded within an manualized course of psychotherapy. Enhanced blinding procedures were used. Depression, anxiety, and quality of life were measured over a 16-week study period. Results: Depression and anxiety significantly improved following both placebo and psilocybin with no significant difference in the degree of change between the two conditions. However, antidepressant effect sizes were larger after psilocybin (d' = 1.02-2.27) than after placebo (d' = 0.65-0.99) and there were high rates of response (66.7%) and remission (46.7%) following psilocybin administration. Antidepressant effects following psilocybin persisted, on average, for 2 months and there were persisting improvements in mood-related quality of life domains. The strength of mystical-type experience during psilocybin dosing was not correlated with subsequent antidepressant effects. Conclusions: The results of this exploratory study highlight the complex interplay between expectancy, therapy effects, and drug/placebo effects in psychedelic-assisted psychotherapy studies. Nonetheless, the acute and persisting clinical improvements observed following psilocybin support further study of its potential in the treatment of major depression. Future studies should more explicitly mitigate and measure expectancy effects and assess the impact of repeated dosing and different forms of psychotherapeutic support.
Article
This theoretical article revives a classical bridging construct, canalization, to describe a new model of a general factor of psychopathology. To achieve this, we have distinguished between two types of plasticity, an early one that we call 'TEMP' for 'Temperature or Entropy Mediated Plasticity', and another, we call 'canalization', which is close to Hebbian plasticity. These two forms of plasticity can be most easily distinguished by their relationship to 'precision' or inverse variance; TEMP relates to increased model variance or decreased precision, whereas the opposite is true for canalization. TEMP also subsumes increased learning rate, (Ising) temperature and entropy. Dictionary definitions of 'plasticity' describe it as the property of being easily shaped or molded; TEMP is the better match for this. Importantly, we propose that 'pathological' phenotypes develop via mechanisms of canalization or increased model precision, as a defensive response to adversity and associated distress or dysphoria. Our model states that canalization entrenches in psychopathology, narrowing the phenotypic state-space as the agent develops expertise in their pathology. We suggest that TEMP - combined with gently guiding psychological support - can counter canalization. We address questions of whether and when canalization is adaptive versus maladaptive, furnish our model with references to basic and human neuroscience, and offer concrete experiments and measures to test its main hypotheses and implications.
Article
Background Psilocybin may have antidepressant properties, but direct comparisons between psilocybin and established treatments for depression are lacking. Methods In a phase 2, double-blind, randomized, controlled trial involving patients with long-standing, moderate-to-severe major depressive disorder, we compared psilocybin with escitalopram, a selective serotonin-reuptake inhibitor, over a 6-week period. Patients were assigned in a 1:1 ratio to receive two separate doses of 25 mg of psilocybin 3 weeks apart plus 6 weeks of daily placebo (psilocybin group) or two separate doses of 1 mg of psilocybin 3 weeks apart plus 6 weeks of daily oral escitalopram (escitalopram group); all the patients received psychological support. The primary outcome was the change from baseline in the score on the 16-item Quick Inventory of Depressive Symptomatology–Self-Report (QIDS-SR-16; scores range from 0 to 27, with higher scores indicating greater depression) at week 6. There were 16 secondary outcomes, including QIDS-SR-16 response (defined as a reduction in score of >50%) and QIDS-SR-16 remission (defined as a score of ≤5) at week 6. Results A total of 59 patients were enrolled; 30 were assigned to the psilocybin group and 29 to the escitalopram group. The mean scores on the QIDS-SR-16 at baseline were 14.5 in the psilocybin group and 16.4 in the escitalopram group. The mean (±SE) changes in the scores from baseline to week 6 were −8.0±1.0 points in the psilocybin group and −6.0±1.0 in the escitalopram group, for a between-group difference of 2.0 points (95% confidence interval [CI], −5.0 to 0.9) (P=0.17). A QIDS-SR-16 response occurred in 70% of the patients in the psilocybin group and in 48% of those in the escitalopram group, for a between-group difference of 22 percentage points (95% CI, −3 to 48); QIDS-SR-16 remission occurred in 57% and 28%, respectively, for a between-group difference of 28 percentage points (95% CI, 2 to 54). Other secondary outcomes generally favored psilocybin over escitalopram, but the analyses were not corrected for multiple comparisons. The incidence of adverse events was similar in the trial groups. Conclusions On the basis of the change in depression scores on the QIDS-SR-16 at week 6, this trial did not show a significant difference in antidepressant effects between psilocybin and escitalopram in a selected group of patients. Secondary outcomes generally favored psilocybin over escitalopram, but the analyses of these outcomes lacked correction for multiple comparisons. Larger and longer trials are required to compare psilocybin with established antidepressants. (Funded by the Alexander Mosley Charitable Trust and Imperial College London’s Centre for Psychedelic Research; ClinicalTrials.gov number, NCT03429075.) VISUAL ABSTRACT Psilocybin versus Escitalopram for Depression