ArticlePDF AvailableLiterature Review

Integrating Psychotherapy and Psychopharmacology:Psychedelic-Assisted Psychotherapy and Other Combined Treatments

Taylor & Francis
Expert Review of Clinical Pharmacology
Authors:

Abstract

Introduction Combinations of psychotherapy with antidepressants are gold-standard psychiatric treatments. They operate through complex and interactional mechanisms, not unlike the reemergent paradigm of psychedelic-assisted psychotherapy, which promising research suggests may also be highly effective in even challenging populations. Areas covered We review the therapeutic mechanisms behind both conventional and psychedelic paradigms, including the evolution of this knowledge and the associated explanatory frameworks. We explore how psychedelics have provided insights about psychiatric illnesses and treatments over the past decades. We discuss limitations to early explanatory models while highlighting and comparing the psychological and biological mechanisms underlying many psychiatric treatments. Methods A narrative review was conducted based on a search in Medline/Pubmed up to January 1st, 2020, and iterative retrieval of references from recent reviews and clinical trials. Expert opinion The contextual model of the common factors of psychotherapy provides a powerful perspective on psychotherapy, antidepressants, and psychedelics, as well as 3,4-methylenedioxymethamphetamine (MDMA) and ketamine. It aligns well with key tenets of psychedelic-assisted psychotherapy. Conventional antidepressants and especially psychedelics may improve the efficacy of psychotherapy via neurochemical changes and increased environmental sensitivity. Combined treatments hold significant promise for advancing the knowledge and treatment of many forms of psychopathology.
REVIEW
Integrating psychotherapy and psychopharmacology: psychedelic-assisted
psychotherapy and other combined treatments
Kyle T. Greenway
a
, Nicolas Garel
a
, Lisa Jerome
b
and Allison A. Feduccia
c
a
Department of Psychiatry, McGill University, Montreal, Quebec, Canada;
b
Data Services, MAPS Public Benefit Corporation;
c
Project New Day,
Psychedelic.Support
ABSTRACT
Introduction: Combinations of psychotherapy with antidepressants are gold-standard psychiatric
treatments. They operate through complex and interactional mechanisms, not unlike the reemergent
paradigm of psychedelic-assisted psychotherapy, which promising research suggests may also be
highly effective in even challenging populations.
Areas covered: We review the therapeutic mechanisms behind both conventional and psychedelic
paradigms, including the evolution of this knowledge and the associated explanatory frameworks. We
explore how psychedelics have provided insights about psychiatric illnesses and treatments over the
past decades. We discuss limitations to early explanatory models while highlighting and comparing the
psychological and biological mechanisms underlying many psychiatric treatments.
Methods: A narrative review was conducted based on a search in Medline/Pubmed up to January 1
st
,
2020, and iterative retrieval of references from recent reviews and clinical trials.
Expert opinion: The contextual model of the common factors of psychotherapy provides a powerful
perspective on psychotherapy, antidepressants, and psychedelics, as well as 3,4-methylenedioxy-
methamphetamine (MDMA) and ketamine. It aligns well with key tenets of psychedelic-assisted
psychotherapy. Conventional antidepressants and especially psychedelics may improve the efficacy of
psychotherapy via neurochemical changes and increased environmental sensitivity. Combined treat-
ments hold significant promise for advancing the knowledge and treatment of many forms of
psychopathology.
ARTICLE HISTORY
Received 27 January 2020
Accepted 18 May 2020
KEYWORDS
Psychedelics; mdma; lsd;
psilocybin; psychotherapy;
antidepressants; psychiatry;
psychedelic-assisted
psychotherapy; mdma-
assisted psychotherapy;
ketamine; ketamine-assisted
psychotherapy
1. Introduction
The global prevalence of psychiatric disorders and their socie-
tal impacts are increasing [1,2], and they are now responsible
for approximately 28% of illness morbidity worldwide – half of
which is attributable to anxiety and depression [2]. Major
depressive disorder is the most common psychiatric illness
[3], the leading cause of disability [3], and is associated with
extraordinary individual and societal costs [4]. Although exist-
ing treatments are generally considered effective [5], treat-
ment-resistance is a common and increasing clinical
phenomenon [6,7]. New psychiatric treatments are thus sorely
needed – particularly ones that are efficacious in patients who
have failed the existing treatments.
Developing central nervous system drugs is associated with
higher development costs, longer approval/development
times, and greater rates of costly failures than other drugs
[810]. In spite of these difficulties, the Food and Drug
Administration (FDA) has bestowed Breakthrough Therapy
designations on several emerging treatments for psychiatric
disorders due to their potential for ‘benefit above that of
available therapies for life-threatening illnesses’ [11]. Of those
that are not yet approved, all except for one are specific
combinations of psychoactive molecules with psychotherapy:
psilocybin-assisted psychotherapy for major depressive
disorder as well as treatment-resistant depression (being
developed by separate entities) [12,13], and 3,4-methylene-
dioxymethamphetamine (MDMA)-assisted psychotherapy for
posttraumatic stress disorder (PTSD) [14]. Additionally, esketa-
mine (Spravato™) has recently received FDA-approval for treat-
ment-resistant depression in conjunction with a conventional
antidepressant, after also receiving the Breakthrough Therapy
designation in August 2016 [15]. This is an intranasal formula-
tion of the s-enantiomer of ketamine, which has been
described by at least one expert as ‘arguably the most impor-
tant discovery in half a century’ [16]. With some notable
exceptions, like brexanolone infusions for postpartum depres-
sion and vortioxetine for major depressive disorder [17,18],
these developments appear to represent the only recent
major pharmacologic advances in the realm of mood and
anxiety disorders and the only treatments specifically being
developed in refractory populations [19].
All three molecules – psilocybin, MDMA, and ketamine –
have been described as ‘psychedelics’ [20]. This term is
a neologism coined in 1953 by psychiatrist Humphrey
Osmond from the Greek psyche (mind/soul) and delos
(reveal/manifest), primarily referring to lysergic acid diethyla-
mide (LSD) [21]. There is some confusion around terminology.
In keeping with the primary mechanism of action of LSD,
many authors reserve the term psychedelic for serotonin 2A
CONTACT Kyle T. Greenway kyle.greenway@mail.mcgill.ca McGill University, Department of Psychiatry, 1033 Avenue Des Pins, Montréal, Canada
EXPERT REVIEW OF CLINICAL PHARMACOLOGY
https://doi.org/10.1080/17512433.2020.1772054
© 2020 Informa UK Limited, trading as Taylor & Francis Group
(5HT
2A
) receptor agonists, whereas others describe this class as
classic (or classical) psychedelics [22,23]. In addition to – or
instead of the label psychedelic, MDMA is classed as an
‘entactogen’ or ‘empathogen’ (empathy inducing) and keta-
mine is described as a ‘dissociative’. Their highly distinct prop-
erties are broached below, but they share the capacity to
engender powerful temporary alterations in consciousness
that may hold therapeutic potential [19,20]. When combined
with specific forms of psychotherapy that aim to translate
these non-ordinary experiences into healing, the term psyche-
delic-assisted psychotherapy (PAP) is generally employed [20].
The aim of this article is to review the principles, history, and
results of PAP, focusing on the prototypical psychedelic LSD
and another 5HT
2A
agonist, psilocybin. We begin with an
examination of conventional psychotherapy and psychophar-
macology, specifically the antidepressants, and their combina-
tion. The evolving understanding of how psychotherapy and
various pharmacotherapies interact holds significant promise
for improved conceptualizations and treatment of a wide
range of psychiatric conditions.
2. Methods
We performed two Medline/Pubmed searches: ‘psychedelic*’,
and ‘antidepressant AND psychotherapy AND review AND
(depression OR mood)’ restricted to English language pub-
lished before January 1
st
, 2020. The former yielded 1038
results and the latter 2122. The database Clinicaltrials.gov
was searched for completed trials on psilocybin, LSD, MDMA,
and ketamine. The identified publications were screened for
relevance, and additional articles were identified by iteratively
reviewing references contained within identified articles. In
total, some 239 have been selected for inclusion in this review.
3. Conventional treatments
The combination of psychopharmacological and psychother-
apeutic interventions is now considered the gold standard in
a wide variety of psychiatric conditions because of comple-
mentarity and potentially synergistic benefits [5].
Psychotherapy can address underlying unresolved psychologi-
cal issues and support behavioral modifications, while drugs
modulate neurochemical signaling and other downstream
processes. We elected to focus on how antidepressants
combine with psychotherapy as this combination has been
studied in depth [5,24], and its benefits are better established
than other psychotherapy-medication combinations, such as
benzodiazepines and antipsychotics [25,26]. Furthermore, anti-
depressants are of particular relevance as they are used to
treat a wide variety of conditions beyond depression, includ-
ing a wide range of anxiety disorders [27], neuropathic pain
[28], and irritable bowel syndrome [29].
3.1. Pharmacotherapies
The first drug to be described as an antidepressant was iso-
niazid, a tuberculosis treatment that was serendipitously
observed in the 1950s to improve various symptoms asso-
ciated with depression [30]. Further clinical and pharmacolo-
gical research led to the discoveries of a number of similar
molecules, now classed as monoamine oxidase inhibitors [30],
as well as the formulation of the ‘monoamine hypothesis’
which postulates that depression is a result of the depletion
of serotonin, norepinephrine, and/or dopamine [31]. The dis-
covery and study of psychedelics played a key role in this line
of investigation [32,33], as discussed below, as well as the
resultant overall shift toward a biomedical model of psychia-
try. The monoamine hypothesis was fruitful, yielding a number
of new classes of antidepressants that act primarily on mono-
amines, including the tricyclics, the selective serotonin reup-
take inhibitors (SSRIs), the serotonin-norepinephrine reuptake
inhibitors (SNRIs), and a variety of other molecules [30,34]. The
SSRIs and SNRIs are currently the most commonly prescribed
antidepressants by a significant margin, mostly because of
their superior safety and tolerability [34,35].
A large body of evidence demonstrates that these antide-
pressants are preferred and effective treatments for many
patients [27,34,3638], fit well into a medical model of mental
healthcare [39,40], and are cost-effective treatments for anxi-
ety and depression [4143]. Nevertheless, their short-term and
long-term efficacy are subject to some debate [4448]. Their
disadvantages include both short-term and long-term side
effects [49], which possibly continue for years following drug-
discontinuation [50], and benefits that may not persist after
treatment discontinuation [50,51]. Relatively long medication
trials of weeks or months are needed to evaluate response
[52]. As with many medications, antidepressant treatment is
marked by poor adherence in approximately 50% of patients
[49], partly due to the delayed onset of benefits [53], the need
for daily dosing even after symptoms have improved [49,52],
impairments in motivation and self-care intrinsic to disorders
such as depression [54], and potential side-effects like
impaired sexual functioning [55]. Patients may also prema-
turely discontinue antidepressants due to fears of dependence
[56], rightfully or wrongfully.
A significant increase in antidepressant prescription rates
has been documented over the recent decades with 1 in 8
American adults estimated to have filled a prescription for an
antidepressant in 2013 [35,57]. This rise is partly attributable to
the increasing duration of antidepressant treatment courses
[58], which some attribute to antidepressant discontinuation
syndrome. That is, patients may be apprehensive about stop-
ping antidepressants due to challenging discontinuation
Article highlights
Psychiatric treatments, including psychotherapy and antidepressants,
act both biologically and psychologically.
Combinations of antidepressants with psychotherapy act through
interactional and potentially synergistic mechanisms.
Though operating with greater acuity, similar mechanisms underly
the combinations of psychotherapy with psychedelics, MDMA, or
ketamine.
This acuity presents significant therapeutic opportunities and
challenges.
Research on conventional combinations can inform the optimal use
of these novel combinations, and vice versa.
2 K. T. GREENWAY ET AL.
symptoms [59,60]. Further, if such symptoms are mistaken for
a relapse of the original disorder, there may be inappropriate
re-initiation of the medication [61]. Some authors argue that
the rise in prescription is due to excessive biomedicalization of
psychiatric illness over the preceding decades [62].
These drawbacks aside, conventional antidepressants
remain extremely important treatments in psychiatry more
than six decades after their discovery. The monoamine
hypothesis, however, has fared less well [63]. The mechanism
of action of these medications is now understood to be more
complex than the correction of neurotransmitter abnormal-
ities. Although uncertainties remain, recent explanatory mod-
els tend to emphasize the roles of neuroplasticity [63],
inflammation [64], and glutamatergic systems in depression
[31]. Copious evidence that talk therapies are, much like anti-
depressants, effective treatments of a wide variety of psychia-
tric illnesses also challenges the monoamine hypothesis
[27,38,65,66], pointing instead to mechanisms that are shared
amongst various illnesses and treatments.
3.2. Psychotherapy
Evidence-based psychotherapies like Cognitive Behavioral
Therapy (CBT) are generally accepted to be equivalently effi-
cacious for depressive disorders as antidepressants, except for
persistent depressive disorder [67]. Psychotherapy offers
advantages in comparison to medication; harms and side
effects are less common [68], quality of life (as opposed to
symptomatology) may be more improved [69], and many
patients prefer psychotherapy [70]. For some conditions, like
specific phobia, psychotherapy is superior to all other treat-
ments [27]. Unfortunately, the availability of psychotherapy is
often lacking [71], partly because of greater upfront costs and
the (generally false) perception of policy-makers that it is not
cost-effective [71,72]. There are nevertheless notable counter-
examples, such as the highly successful Improving Access to
Psychological Therapies program in the United Kingdom
[73,74], which demonstrates that providing psychotherapy at
a population level is feasible. An additional difficulty with
psychotherapy is that the treatment requires greater patient
commitment than medications, in terms of appointments and
active efforts to engage, which can be challenging for ill
patients who often suffer from, for example, impaired cogni-
tion or low motivation [75]. Benefits of psychotherapy, like
antidepressants, typically appear after several weeks of treat-
ment [66,67,76], but are more likely to persist post-treatment
[77].
Psychotherapy has no single definition but may be under-
stood as an interpersonal intervention that relies on repeated
encounters, a healing relationship, and a particular explana-
tory model, all within a structured therapeutic frame
[65,78,79]. Its origins have been variously traced to Josef
Breuer and Sigmund Freud’s development of psychoanalysis
[80], Philippe Pinel’s moral treatment [81], or, more broadly, to
diverse traditions including Western philosophy [82], Eastern
religions [83], and shamanism [78].
Modalities and techniques of psychotherapy are numerous
and diverse. However, a significant portion of the therapeutic
benefits of psychotherapy may be due to components that are
generally shared amongst various approaches – the so-called
common factors – rather than ingredients that are unique to
a particular therapeutic model [84,85]. Indeed, the ‘dodo bird
verdict’ postulates that all psychotherapeutic approaches are
similarly efficacious in part because the systematic application
of any therapeutic theory by a skilled therapist is beneficial
regardless of that theory’s actual veracity [86]. It was first
described in 1936 and remains an important topic today
[87,88]. Some authors thus speak of the creation and applica-
tion of a ‘therapeutic myth’ between patient and therapist as
a key component of psychotherapy [78,89]. That is, establish-
ing a mutually acceptable rationale that explains an indivi-
dual’s psychopathology and guides its treatment.
One widely-referenced model for the common factors is
the contextual model, which posits three primary mechanisms
of action of psychotherapy: the real relationship, the creation
of expectations through explanation of disorders and treat-
ments, and the enactment of health promoting actions [84,85].
While these factors are shared between schools of psychother-
apy, their implementation can vary dramatically in terms of
how therapists and patients relate to each other, the explana-
tory frameworks (or therapeutic myths) that they employ, and
the tasks that they undertake. For example, CBT partly strives
to correct collaboratively identified maladaptive patterns of
thought and to facilitate engagement in behavioral exposure
and activation [90]. Psychodynamic psychotherapy, on the
other hand, partly aims to change patients’ problematic pat-
terns of relating to their selves and to others via developing
insight about past relationships and their relationship with the
therapist [90]. The contextual model also acknowledges that
a strong therapeutic alliance is a necessity of all pathways, as
is supported by research [85]. Exactly how – and how well –
this alliance is established varies amongst techniques and
individual therapists [85].
While the contextual model explains research findings of
equivalent benefits between various psychotherapies in most
psychiatric conditions, it fails to explain why particular
approaches are superior in treating other conditions. One
classic example is the clear superiority of CBT including
a specific technique known as Exposure Response Prevention
over psychodynamic psychotherapy in the treatment of obses-
sive-compulsive disorder (OCD) [91,92]. Nevertheless, the con-
textual model provides a powerful lens for understanding the
psychological underpinnings of psychiatric care, even beyond
psychotherapy.
3.3. Shared pathways
Combining antidepressants with psychotherapy to treat
depression is often framed in terms of additive benefits via
distinct mechanisms, and indeed response rates are typically
greater than either in isolation [24], especially in terms of
quality of life [67]. However, antidepressant treatment is
more than a biological modulator of neurotransmitters, and
combination treatment is highly interactional. For example,
adding psychotherapy to antidepressant therapy may actually
increase the tolerability of medication, as evidenced by
reduced trial drop-out rates [67,93]. Effective treatment with
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 3
antidepressants may even reduce the use of immature
defense mechanisms and dysfunctional thinking [94,95], key
concepts in psychodynamic therapy and CBT, respectively.
This may be due to decreased symptomatology (which fre-
quently includes cognitive changes [75]) increasing therapy
efficiency [96], amongst other biological or expectancy effects.
The contextual model may also be applied to antidepres-
sants in that a correctable chemical imbalance may be seen as
an (imperfect) explanatory model, or a therapeutic myth,
through which the real relationship between the prescriber
and the patient provides for healing [97]. The famously power-
ful placebo effects in studies on depression support this per-
spective [45], as placebos routinely achieve 50% to 85% of
antidepressant levels of efficacy [98]. Such benefits are mark-
edly reduced when a plausible explanatory model of illness
and treatment is not provided. For example, a three-arm trial
comparing blinded antidepressant or placebo treatment to
supportive care (theoretically delivering the common factors
except for explanation) demonstrated a 46% symptom reduc-
tion with antidepressant medication versus 36% for placebo
and 5% for supportive care [99]. This suggests that, given
a specific therapeutic context, acts like pill-taking (placebo or
antidepressant) can have major benefits.
Conversely, explanatory models can reduce the benefits of
real medications. One study demonstrated that presenting an
antidepressant as an ‘active placebo’ greatly reduced benefits
on anxiety [100]. In a testament to the power of expectation
and the complexity of mind-body interactions, functional
magnetic resonance imaging changes in this study were also
dependent on the provided explanatory model. Further sup-
port for the psychological importance of antidepressant pre-
scription is that their effectiveness is partly mediated by the
reported strength of therapeutic alliance [99,101], much like
psychotherapy [85]. Such findings have led some to argue that
the treatment of depression by antidepressants is primarily
a psychological treatment [102], but alternative explanations
better capture the rich complexity of simultaneous treatment
by psychotherapy and medication.
One possibility is that modulating neurotransmitters with
antidepressants increases sensitivity to the environment
[103,104]. This has been demonstrated in a reanalysis of the
Sequenced Treatment Alternatives to Relieve Depression
(STAR*D) trial – the largest prospective clinical trial on depres-
sion ever [105] – which found that higher doses of the anti-
depressant citalopram were associated with a much greater
positive impact from positive socioeconomic status markers,
like education, employment, and income. This finding con-
trasts with the remarkably flat dose-response curve of antide-
pressants found in many studies [106], which may partly result
from positive and negative environments being amplified at
higher doses and thus canceling out. An analogous result has
been shown in animals (free from expectations); in one study,
rat models of depression treated with fluoxetine improved in
enriched living conditions but worsened in stressful living
conditions on several biological and behavioral outcomes
[107]. In combination with therapy, antidepressant treatment
might similarly increase sensitivity to reparative interactions
with a psychotherapist.
This mechanism could also explain why antidepressants
with very distinct pharmacologic profiles tend to achieve
remarkably similar levels of efficacy. For example, in the
STAR*D study, after failing to respond to one SSRI (citalopram),
response and remission rates did not differ between patients
switched to another SSRI (sertraline), an SNRI (venlafaxine), or
a norepinephrine-dopamine reuptake inhibitor (bupropion)
[105]. On the surface, this would seem to suggest that seroto-
nin, norepinephrine, and dopamine abnormalities were
equally distributed in this large, real-world population.
Alternatively, any monoamine modulation may potentially
improve symptoms via a shared mechanism like environmen-
tal sensitization, rather than the correction of neurotransmitter
imbalances. This may partly account for the disappointing lack
of benefits of antidepressants in patients with medical issues
or addictions [108110], given that their environments may be
relatively more negative. In contrast, antidepressants have
been found to be more efficacious than placebos in the treat-
ment of addiction when accompanied by psychosocial inter-
ventions [111].
Just as the mechanism of action of antidepressants is more
psychological than may appear at first glance, psychotherapy
acts more biologically than might be expected. Introspection,
challenging dysfunctional thoughts, cultivating self-
compassion, practicing adaptive behaviors, and healing inter-
actions with a therapist may all remodel dysfunctional neural
circuits [112]. Indeed, neuroimaging has demonstrated that
both psychotherapy and pharmacotherapy engender neural
reorganization to a similar extent across many conditions
[113]. Plasma markers of increased neuroplasticity and cellular
resilience have also been found with successful psychother-
apy [114], as have epigenetic changes in both children and
adults [115,116]. Psychotherapy’s capacity to modify gene
expression has prompted renowned psychopharmacologist
Stephen Stahl to analogize it as an ‘epigenetic drug’ [117].
These findings demonstrate that any effective treatment of
psychopathology, in all of its complexity, cannot be fully
understood from purely biological or psychological
perspectives.
4. Psychedelics, MDMA, and ketamine
4.1. How early research on psychedelics transformed
psychiatry
One new – or rather, resurgent – approach that thoroughly
integrates multiple paradigms is psychedelic-assisted psy-
chotherapy. The quintessential psychedelic LSD was first
synthesized by Albert Hoffman while working for Sandoz in
1938, and its psychoactive effects were discovered during his
accidental ingestion 5 years later [118]. Its unique psychologi-
cal effects inspired hope for potential therapeutic applications,
which Sandoz pursued by distributing LSD to investigators
and clinicians worldwide, free of charge [119].
Of those who began to grapple with LSD, some viewed its
effects as mimicking psychosis and therefore referred to it as
a psychotomimetic, a term used at the time to describe
another psychedelic, mescaline [120]. Others saw LSD as caus-
ing ‘an upsurge of unconscious material into consciousness’,
4 K. T. GREENWAY ET AL.
producing dream-like states that greatly interested psycho-
analysts [121]. Still others drew parallels between LSD and
delirium tremens, an often-terrifying altered state induced by
alcohol withdrawal [122].
In every case, psychedelics challenged the dominant psy-
chological conceptualizations of mental illness. The psychoto-
mimetic model, for example, conflicted with the view that
psychotic disorders like schizophrenia arose from ‘refrigerator
mothers’ [123]. Instead, researchers began to frame schizo-
phrenia in a more biological light, hypothesizing the etiology
to be pathological metabolism of endogenous molecules
resembling LSD or mescaline [124]. This hypothesis has since
been essentially discarded, but the shift toward a biological
perspective set the stage for the first commercial antipsycho-
tics in 1952 [32,125], which have since superseded psycho-
analytic approaches in the treatment of schizophrenia.
Similarly in the case of depression, LSD’s potency in combina-
tion with its structural similarity to the recently discovered
serotonin lead to the first proposal in 1954 that serotonin
might play a significant role in mental illness [33,126]. Thus
depression was also framed more biologically, a view that was
also consolidated by the appearance of a novel commercial
drug, imipramine, in 1959 and the propagation of the mono-
amine hypothesis discussed above [31].
Prior to the psychedelics, psychiatry had already seen other
efforts at integrating biological and psychological treatments.
For example, ‘narcoanlaysis’ referred to the practice of utilizing
barbiturates to facilitate psychoanalysis [127], and various
other agents were studied as adjuncts of psychotherapy,
such as nitrous oxide, ether, and carbon dioxide [128]. While
these agents rapidly fell out of fashion, research on psyche-
delics continued steadily. More than 1,000 scientific publica-
tions including 40,000 patients were published in this era
[129], and many mainstream figures described psychedelics
as offering new frontiers for psychiatry and neuroscience
[119]. However, until recently, psychiatry’s shift toward biome-
dical models and the political backlash against recreational
use led to this work being largely halted [62].
Before then, LSD inspired remarkably sophisticated reflec-
tion on the relationship between biology and psychology that
other psychoactive drugs did not, and even the earliest works
anticipated many later advances in the psychological under-
standing of conventional antidepressants [130]. For instance,
Sandison and colleagues described in 1954 how the drug
effects were highly influenced by the therapeutic relationship,
the environment, and the patient’s unique psychic structure
[131,132]. These reports and others stand in stark contrast to
more simplistic and reductionist explanatory models from the
same period.
4.2. Psychedelic pharmacology and phenomenology
LSD is a highly absorbable semi-synthetic tryptamine with
significant structural overlap with serotonin [33]. It has strong
affinity for a variety of receptors, binding potently to 5HT
2A
,
5HT
1A
, 5HT
2 C
, and various adrenergic and dopaminergic
receptors, amongst others [120]. Its pharmacology is complex
[33,120], but 5HT
2A
(partial) agonism is central to its effects, as
demonstrated by the nearly complete extinction of psycholo-
gical effects by 5HT
2A
antagonists like ketanserin [133,134].
A typical dose of 100–200 μg of LSD causes profound altera-
tions in cognition, sensory processing, and emotions [33],
usually lasting 8–12 hours and peaking around 2 hours post-
ingestion, roughly corresponding to maximal plasma
levels [120].
Other psychedelics like psilocybin/psilocin, dimethyltrypta-
mine (DMT), and mescaline are generally understood to differ
more pharmacokinetically than pharmacodynamically: agon-
ism of 5HT
2A
is the major feature shared by the family [33].
This commonality is remarkable given that the origins of
various psychedelics can be traced to pre-Columbian
Mesoamerican societies, North American native peoples, or
mid-20
th
century Switzerland [33]. Notable pharmacokinetic
differences include psilocybin’s shorter duration of effects (4–-
6 hours) compared to LSD [135], and the destruction of DMT
by liver monoamine oxidase enzymes, necessitating parenteral
administration (or smoking [136]) or simultaneous administra-
tion of monoamine oxidase inhibitors as are found in botanical
preparations such as ayahuasca [33]. One shared feature of
this class is a rapid buildup of tolerance and cross-tolerance,
often attributed to tachyphylaxis of 5HT
2A
receptors, after only
several consecutive days of dosing [33]. The molecules are
considered to be exceptionally safe when administered with
appropriate precautions though safety concerns include the
triggering of psychotic episodes and potentially lethal interac-
tions with other medications/substances [33,120,137].
The phenomenology of the profoundly altered state pro-
duced by 5HT
2A
agonism is multifaceted and heterogenous
between individuals and studies. Modern studies employing
psychometrically-valid questionnaires, like the Altered states
of Consciousness Scale and the Mystical Experience
Questionnaire [138,139], consistently find dramatic perceptual
changes like sensory distortions and synesthesia [133], feelings
of ego dissolution or oceanic boundlessness [140,141], and
mystical experiences that strongly resemble those arising
naturally [142]. Suggestibility is increased [143], and empatho-
genic effects on mood such as increased trust and openness
are common [144]. Emotional processing is altered, including
reduced recognition of facial expressions of fear [144], and
subjects may report facing challenging emotions and gaining
a newfound sense of resolution [145]. Meaning-making is
altered; e.g., music may evoke more wonder and transcen-
dence [146], and rapidly take on personal meaning [147].
Indeed, the experiences themselves are often described as
highly meaningful [135], even years later [148].
4.2.1. Psychedelics and neural networks
Various efforts aiming to explain these effects, or relating
them to known psychological concepts, focus on observed
changes in communication within and between neural net-
works. Psychedelics typically decrease the former and increase
the latter [120,149], resulting in novel pathways of neural
communication [120]. Emphasis is often placed on alterations
to the Default Mode Network (DMN), which is most active
when individuals are not focused on the immediate environ-
ment [150], but rather engaged in self-referential thought
[151], or mental time travel [152], for example. Its
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 5
disintegration may thus engender a more present-moment
state, as neuroimaging studies of meditators suggest [153].
Links have been made between the DMN and the Freudian
conception of ego given its role in one’s sense of self as well
as its suppressive effects on lower-level subcortical systems
[154]. Though such analogies are inherently imperfect, it has
been shown that the disintegration of the DMN correlates
with participant-reported ratings of ‘ego dissolution’ [140].
The disintegration of other networks, including the cortico-
striato-thalamo-cortical circuit of the salience network
[155,156], have also been hypothesized as important
mechanisms.
Other effects include decreased constraints on cognition
[149], which may account for their phenomenological descrip-
tion as amplifiers of mental processes [157]. Amongst the
changes within and between neural networks, reduced con-
textualization and filtering of sensorial information, and/or
increased communication involving visual/auditory systems,
may produce perceptual disturbances [158,159]. Several over-
arching models have been proposed, for instance describing
these states of less constrained cognition and perception as
‘high entropy’ [149], or as being liberated for bottom-up
information flow [158]. No consensus has yet been reached,
but psychedelic states continue to inspire significant funda-
mental and clinical interest [160].
4.3. MDMA pharmacology and phenomenology
MDMA, has a distinct pharmacological profile from psychede-
lics that induces alterations in normal consciousness consid-
ered by some to be ‘psychedelic’ [138]. MDMA reverses
monoamine re-uptake transporters to effectively increase pre-
synaptic concentrations of norepinephrine, dopamine, and
especially serotonin [161]. Subsequent release of hormones,
including oxytocin, prolactin, cortisol, and vasopressin, also
contribute to the subjective effects [141]. MDMA is known as
an empathogen or entactogen and a ‘heart opening’ sub-
stance [161], with effects typically lasting for 4 hours after
a standard dose of 75–125 mg. Visual hallucinations are not
common, but sensory and perceptual alterations do occur
[138]. MDMA acutely modulates emotional-memory circuits,
including decreasing activity in the amygdala and increasing
connectivity between the hippocampus and amygdala [162].
One of the results appears to be greater prosocial behavior
[163,164]. Before MDMA was classified as a schedule 1 con-
trolled substance in the United States in 1985 due to wide-
spread recreational use [165], it was employed in thousands of
therapy sessions [166]. These early therapists generally already
had experience working with psychedelics like LSD but found
the non-hallucinatory MDMA to be well-suited to particular
applications such as couple’s therapy.
4.4. Ketamine pharmacology and phenomenology
Ketamine’s history and pharmacology differ dramatically
from MDMA and LSD. It is primarily a N-methyl-D-aspartate
receptor (NMDA) antagonist that modulates glutamergic
transmission [167], though it is active at a large variety of
receptors [168]. It’s efficacy against depression is thought to
owe to enhanced neuroplasticity and reversal of stress-
induced neuronal changes [169,170]. Doses between 0.5
and 10.0 mg per kilogram of body weight can induce pro-
found alterations in consciousness characterized by signifi-
cantly decreased movement, bodily sensation, and
interactivity [138]. These properties led to the description
of ketamine as a ‘dissociative anesthetic’, a term coined by
the wife of one of the first pharmacologists to study keta-
mine, Edward Domino [171]. Domino and his colleagues had
initially settled on descriptions such as ‘dreaming’ for keta-
mine’s effects [171], similar to language employed for psy-
chedelics [133], and indeed the states induced by ketamine
overlap phenomenologically with those induced by MDMA
and especially psilocybin [138]. Comparable levels of experi-
ences of unity, spiritual experiences, insightfulness, visual
phenomena, and altered meaning are reported with all
three compounds.
Since its discovery in 1962 [171], ketamine found wide-
spread medical applications including anesthesia and pain
management [172], and thus was never listed as a Schedule
1 substance. Like the psychedelics, Ketamine has been
employed as a psychotomimetic, a use that lead Yale research-
ers to demonstrate in 2000 that a single subanesthetic dose
rapidly improved depressive symptoms within minutes or
hours [173]. Since then, ketamine has been increasingly
employed as a treatment of refractory unipolar and bipolar
depression [174], amongst psychiatric conditions including
anxiety disorders and PTSD [175,176]. Ketamine (and esketa-
mine) is generally employed much like conventional antide-
pressants [177], typically administered by repeated 40-minute
IV 0.5 mg/kg infusions twice to thrice weekly [178], for months
or years [179]. In contrast, ketamine’s first use in psychiatry
resembled psychedelic-assisted psychotherapy in that intense
experiences were intentionally engendered by relatively high
doses, within a psychotherapeutic frame [180]. Ketamine is
thus unique in that it has been – and continues to be – utilized
within both psychedelic and more conventional paradigms
[174,181].
5. Psychedelic-assisted psychotherapy
Soon after its discovery, LSD attracted the attention of psycho-
analysts who were deeply interested in internal mental pro-
cesses and concepts like ego [132]. Bringing unconscious
material into conscious awareness such that it may be ‘worked
through’ is a fundamental aim of insight-oriented psy-
chotherapies, and LSD thus found early uses as
a psychoanalysis catalyst [131,182]. From there, the approach
of PAP evolved beyond its psychoanalytic origins. As early as
1959, an increased emphasis was placed on patient autonomy,
with therapists taking less active roles in the therapeutic
process [130]. The common experiences of ego-death and
feelings of transcendence were explained with concepts
from thinkers like Jung, Maslow and others [130,183].
Contributing to the development of movements like transper-
sonal psychology [184], spirituality was often embraced rather
than seen as a regressive defense as Freud had [185]. The
effects of music within psychotherapy were discovered, and
music became an important component of the therapeutic
6 K. T. GREENWAY ET AL.
approach [130,186]. A different treatment rhythm from stan-
dard psychotherapy emerged, partly due to drug effects per-
sisting for 4–12 hours and tachyphylaxis complicating repeat
dosing [33]. Over time, principals that are relatively common
to modern psychedelic protocols were established.
5.1. Modern protocols
One of the most referenced clinical approaches is that of
Stanislav Grof, and his classic 1980 book LSD Psychotherapy
[157]. He worked extensively with LSD, supervising over 4500
LSD research sessions during the 1950–1960s [157]. Although
his book is not the first or only of its kind, it has been cited in
nearly all modern PAP studies including those employing
psilocybin, ketamine, and MDMA [187189]. For example, the
MDMA Treatment Manual used in clinical trials investigating
MDMA-assisted psychotherapy for treatment of PTSD largely
draws from Grof’s work [188], and other early psychedelic
researchers. The most common tenets and their evolution
will thus be explored.
This protocol centers on carefully engendering powerful
non-ordinary states of consciousness, which are described in
terms such as peak experiences or mystical experiences
[135,190]. Efforts are then made to ‘integrate’ these unusual
experiences into long-term gains. This conceptualization dif-
fers significantly from both conventional pharmacological and
psychotherapeutic approaches. The former involves regularly
administering drugs to control symptoms and/or correct
pathological neurobiological states, while the latter aims to
create change via years or months of corrective experiences
[157]. The PAP protocol consists of three phases: preparation,
treatment, and integration. These roughly align with the con-
textual model’s primary mechanisms of action: establishing
a therapeutic relationship, creating expectations of healing,
and enacting health-promoting activities [84,85].
In the preparation phase, patients are assessed for their
appropriateness for the treatment, including readiness for
potentially intense and challenging experiences. The most
emphasized potential risks are psychiatric including histories
of psychosis or bipolarity [137], or immature psychological
defenses such as those found in borderline personality disor-
der [191]. Potentially dangerous interactions with medications
like antidepressants are identified, and medication adjust-
ments are made as needed to avoid unwanted interactions
such as serotonin syndrome [137]. Forming a therapeutic rela-
tionship, including discussing biographic, medical, personal,
and spiritual histories, is critical in increasing trust and thereby
reducing potential for anxiety or so-called ‘bad-trips’ [137]. In
addition to providing education regarding the specific com-
pound’s psychoactive effects, patients are typically encour-
aged to engage in intention setting [192]. By forming an
explicit rationale for the need and hopes for the treatment
[193], which may also be described as creating the therapeutic
myth or adopting an explanatory framework, patients begin to
set expectations around the experience and outcomes. As
discussed above, this is a critical component for healing in
essentially all psychiatric interventions and is especially so
with psychedelics, ‘amplifiers of mental processes’ [157],
given the increased suggestibility and acutely heightened
environmental sensitivity [130,143,183]. Finally, patients are
typically prepared with ‘flight instructions’ that can be traced
back to 1950s [130], including directions to ‘mentally surren-
der to the experience’ [137], or to ‘trust, let go, be open’ [194].
Experience has shown that cultivating such a mind-set reduces
anxiety and increases the probability of working through
challenging experiences to perhaps achieve therapeutic ‘emo-
tional breakthroughs’ [145,194]. These directions, rare excep-
tions to the typical stance of therapeutic non-directiveness
[20], are given in the spirit of allowing individualized experi-
ences to unfold.
The second phase is the actual treatment sessions of drug
administration, typically lasting 8–12 hours for LSD, and
4–6 hours for psilocybin and MDMA [157]. Treatments typically
involve two therapists or monitors to increase the safety of
patients in vulnerable states, and to permit brief breaks during
long sessions [137]. Theoretically, having both female and
male therapists or monitors present may better evoke diverse
expressions of unconscious patterns of interpersonal relation-
ships in a psychological process known as ‘transference’ [157].
The therapists/monitors are often referred to as guides as
a reflection of the desired stance of non-directiveness [137].
This is often explained with reference to Grof’s concept of the
inner healing intelligence [157], though similar ideas can be
found in very early works [130], which postulates that humans
heal psychologically much as they do physically: via innate
capacities that can be enhanced but not replaced by external
interventions. This framing differs significantly from conven-
tional pharmacotherapy’s emphasis on postulated neurotrans-
mitter pathology, and the emphases found in conventional
psychotherapy models, (e.g. dysfunctional thinking patterns in
CBT and unconscious psychic conflicts in psychoanalysis-
inspired approaches [94,95]), which require more active and/
or authoritative therapeutic stances. The emphasis on patient
autonomy and internal strengths may have partly arisen as
a necessary measure to avoid excessive influence of psy-
chotherapists given the increased suggestibility associated
with psychedelics [143]. However, the hypothesis that
a significant portion of psychiatric treatment efficacy arises
from patients’ innate capacities, independent of treatment-
specific aspects, is supported by the power of the common
factors in both pharmacotherapy and psychotherapy.
5.2. Set and setting
Sociological research on psychedelic use in populations ran-
ging from modern recreational users to traditional indigen-
ous communities suggests a near universal emphasis on the
importance of ‘set and setting’ [195]. This refers to the
patient’s mind-set and the treatment environment, respec-
tively, and is an important concept in clinical studies of
psychedelics for both safety and efficacy [137]. Set and set-
ting might otherwise be described as a patient’s immediate
internal and external environment, and even the earliest
studies recognized that psychedelics tend to amplify
thoughts and feelings about both [130,131]. The former is
optimized during the preparation phase, especially by estab-
lishing a strong therapeutic alliance and an intention for
healing [183,192]. The latter is typically optimized by
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 7
providing a less sterile and more aesthetically pleasing treat-
ment environment, often described as living room-like [137].
The much higher rates of anxiety and negative emotions that
were seen in early psychotomimetic psychedelic studies
attest to the importance of set and setting [196,197].
Influential psychotherapists like Donald Winnicot evoked
similar concepts, such as the creation of therapeutic ‘holding
environments’ [198], and evidence is mounting that even
aesthetic improvements to medical settings can improve
diverse patient outcomes [199201].
5.3. Music
Music therapy is an effective treatment of mood disorders
[202,203], and music forms a core component of PAP in
a unique way, non-verbally supporting and enhancing patient
experiences [204]. Patients typically spend time during the
treatment sessions lying comfortably, wearing blindfolds, and
listening to music via headphones with minimal interaction
with therapists [137]. Qualitative and quantitative studies have
shown an important multidirectional relationships between
music and emotions, mental images, and association of mean-
ing [140,146,197,205]. Three music-specific variables have
been proposed to mediate therapeutic response to psilocybin:
appreciation versus rejection of its style and quality, corre-
spondence versus dissonance to the current emotional state,
and music-evoked openness versus resistance [206]. In some
ways, the presence of music can conflict with the desired non-
directive therapeutic stance as its qualities can induce specific
emotional and psychological states [206]. This can enhance
the holding environment or evoke intense resistance – much
like other directive therapeutic interventions. Resistance is
mitigated by the encouragement to approach negative reac-
tions to specific music with openness and acceptance, and
music is also frequently adapted by therapists during treat-
ment [206].
5.4. Integration
Finally, following treatment, several non-drug sessions of
‘integration’ take place [137,157], usually within a day of the
first treatment and lasting for up to several hours [187]. These
sessions often incorporate elements from various schools of
psychotherapy including CBT, psychodynamics [130], and
existential meaning-making therapies [187], amongst others.
Aspects more unique to PAP include generally encouraging
patients to write down detailed accounts of their (often
intense) experience to facilitate working through [183,187].
As with treatment sessions, therapists are encouraged to
allow for a patient’s own experience and meaning-making
to unfold as it will, without undue influence [130,188].
Patients are typically encouraged to engage in various health-
promoting activities, one of the major common factors of
psychotherapy [84]. Such behavioral interventions likely con-
tribute significantly to the long-lasting benefits obtained with
PAP, evidenced by the magnitude of therapeutic change seen
in therapies that consist entirely of behavioral activa-
tion [207].
6. Modern research, safety and addiction
6.1. Psychedelics
Aside from a few affinities of unknown clinical significance,
including 5HT
1A
or D
2
receptors, psilocybin is remarkably
pharmacologically similar to LSD, and thus it is generally
employed with similar methods [33,120]. One significant dif-
ference is its shorter half-life, which is one of the major rea-
sons that psilocybin is more widely utilized in modern
research than LSD, in that treatment sessions can be con-
ducted with 6-hours of therapist time versus approximately
12 [157,187]. The other major advantage of psilocybin is that it
is generally associated with less public stigma than LSD
[137,187]. Since the first modern study of psilocybin-assisted
psychotherapy in 2006 for OCD [208], promising results have
been published for applications in end of life anxiety
[148,187,209], smoking cessation [210], treatment-resistant
depression [211], as well as to initiate meditation/spiritual
practices [212]. Remarkable hallmarks of these studies include
rapid and large effect sizes, that have persisted during follow-
up periods ranging from 6-months to nearly 5 years. Lasting
effects extend beyond reduced symptomatology include
increased spirituality [212], increased nature-relatedness
[213], and enhanced personality measures of extraversion of
openness [214]. Of note, the patients enrolled in these studies
had often failed to respond to multiple existing psychother-
apeutic and pharmacologic treatments. These results have led
to two pharmaceutical sponsors, Usona Institute and
COMPASS Pathways, receiving FDA Breakthrough Therapy
designation and the go-ahead to proceed to phase 3 trials
for investigating psilocybin as a treatment for major depres-
sive disorder and treatment-resistant depression [12,13].
Modern research on both recreational users and healthy
controls provides additional evidence for the potential bene-
fits of psychedelics, possibly even without concurrent psy-
chotherapy [215,216]. For example, a prospective survey of
654 people who used psychedelics found significant improve-
ments in a measure of psychological well-being [216]. These
benefits were moderated by the traits of the individuals sur-
veyed as well as their attention to set and setting, supporting
the importance of this tenet of PAP. Retrospective surveys
have further shown a correlation between recreational psy-
chedelic use and less criminal recidivism (in contrast to all
other drugs [217]) and less intimate partner violence
[218,219]. Though these studies are unable to prove causality,
controlled studies of psilocybin administered without psy-
chotherapy have also found benefits to mood and well-
being [215]. These findings attest to the general safety of the
psychedelics, and indeed the rate of adverse events in PAP
studies is comparable to standard psychotherapy [220].
Nevertheless, there have been reports of injuries and deaths
resulting from recreational use of psychedelics, which typically
occurred in the setting of ingesting multiple substances and/
or dangerously uncontrolled environments [137]. These risks
appear to be substantially greater in relatively novel psyche-
delic analogs [221].
Overall, LSD and psilocybin appear to be amongst the least
dangerous drugs in recreational users, even in comparison to
8 K. T. GREENWAY ET AL.
those with well-established medical uses like benzodiazepines,
in terms of both harm to users and harms to others [222]. The
potential for addiction is considered to be especially low [33].
This may be in part due to tachyphylaxis of 5HT
2A
receptors,
though more psychological mechanisms may also be at play.
Indeed, some studies have found that recreational use of
psychedelics is associated with less abuse of other substances
[217], and psilocybin and LSD have been employed with
success as a treatment of alcohol and nicotine addictions
[210,223].
6.2. MDMA
Since 2000, MDMA-assisted psychotherapy has been under inves-
tigation as a treatment for chronic treatment-resistant PTSD. Six
phase 2 trials were sponsored by the Multidisciplinary Association
for Psychedelic Studies (MAPS) from 2004–2016, with results indi-
cating substantial improvements in PTSD symptoms and ade-
quate tolerability to the treatment protocol [224]. During MDMA
sessions, participants can more readily process traumatic mem-
ories and engage with the associated challenging emotions, due
to their ‘window of tolerance’ being increased [225]. Greater
introspection, enhanced compassion for oneself and others, and
a larger capacity to feel and talk about traumatic memories have
all been proposed as key psychological mechanisms induced by
MDMA in a therapeutic context [225]. As with classic psychedelics,
integrative non-drug therapy sessions are essential for contextua-
lizing the experience and applying insights to everyday life. Long-
term (12 or more months) reductions in PTSD symptoms and
other positive outcomes have been observed following treatment
[226]. After reviewing data from these trials, the FDA granted
a Breakthrough Therapy designation in 2017, and the first of two
phase 3 trials commenced in November 2018 [14]. An Expanded
Access program for 50 patients with PTSD to undergo MDMA-
assisted psychotherapy will start later in 2020. In addition, MDMA-
assisted psychotherapy has been explored for use in other indica-
tions, including social anxiety in autistic adults and anxiety related
to life-threatening illness [227], and current or planned trials will
evaluate its use for alcohol use disorders and eating disorders.
MDMA is considered to have a moderate abuse liability
amongst recreational users [222]. In research trials conducted in
controlled clinical settings, there have been no reports of drug
craving or substance use disorders (SUDs) arising after treatment
[224]. Therefore, the potential for problematic use is considered
low when administered under supervision with concurrent edu-
cation about risks of ‘Ecstasy’ use in non-medical contexts.
6.3. Ketamine
Ketamine is generally employed without application of the
tenets of PAP, such as concurrent psychotherapy or the opti-
mization of set and setting [179]. The altered states it engen-
ders are typically framed for both clinicians and patients as
side-effects to be mitigated [228]. However, ketamine has also
been employed in a PAP-like model to facilitate therapeutic
changes. Indeed, ketamine-assisted psychotherapy has been
successfully used in the treatment of depression/anxiety [181],
PTSD [229], as well as alcohol [230], heroin [231], and cocaine
addiction [232]. These latter results are somewhat surprising
given that ketamine is a substance of abuse [222], with sig-
nificantly more harm and potential for physiological depen-
dence than the psychedelics [233]. Pharmacological
explanations may partly underly these results [234], but can-
not fully explain how ketamine exerts anti-addiction effects
against multiple substances of abuse. Rather, evidence sug-
gest that the experiences induced by ketamine – much like
the psychedelics within a psychotherapeutic frame least
partially mediate its efficacy against addiction [235].
Psychedelic-like experiences may also partially underly keta-
mine’s antidepressant effects even when employed in purely
biomedical contexts [236238].
7. Expert opinion
Psychiatry has evolved considerably over the preceding dec-
ades from primarily biological or psychological models of dis-
ease to more nuanced, integrative frameworks. We have
discussed several key examples, such as how the contextual
model of psychotherapy applies to antidepressants and how
psychotherapy acts biologically. Combinations of psychother-
apy with antidepressants, psychedelics, MDMA, and ketamine
are even more complex, involving bidirectional and potentially
synergistic interactions. These combinations share much; the
contextual model’s three central common factors correspond
well to longstanding PAP approaches, and enhanced environ-
mental sensitivity may underly many diverse combinations.
This increased environmental sensitivity is much more
acute in PAP, giving rise to dramatically altered states that
may result in ‘therapeutic breakthroughs’ or ‘bad-trips’,
thereby presenting both significant opportunities and chal-
lenges. Emerging evidence suggests that PAP can engender
powerful and persistent benefits in highly refractory patients.
These can appear ‘ultra-rapidly’, thereby significantly reducing
the duration of suffering for responders. Likewise, non-
responders can pursue alternative treatments without requir-
ing weeks or months of medication/psychotherapy to evaluate
response. Remitters may thereafter require fewer daily medi-
cations, thereby reducing risks associated with side-effects and
drug-drug interactions. Fewer psychotropic medications may
also yield clearer clinical pictures that are less obfuscated by
uncertainty around compliance and discontinuation symp-
toms. Lastly, amongst psychiatry’s wide variety of ‘therapeutic
myths’, PAP’s explanatory frameworks that highlight patients’
inner healing capacities may increase feelings of autonomy
relative to frameworks that instead emphasize neuropatholo-
gical states.
Challenges include carefully selecting and preparing
patients, who may require medication adjustment or discon-
tinuation – associated with risks including suicide [59] – so as
to avoid potential interactions such as serotonin syndrome
[137]. Altered states require meticulous attention to set and
setting, as well as patient safety, and it is not clear if the high
standards of clinical trials will translate to the clinic.
Therapist-specific factors are known to be an important mod-
erator of clinical results, and current research has largely
been performed by experienced and highly motivated thera-
pists [85,97]. Rightfully or wrongfully, PAP is likely to be
viewed as excessively resource-intensive much like
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 9
conventional psychotherapy. Dissemination of PAP will
require overcoming stigma and making significant changes
to biomedical models, in both conceptual and practical ways.
Accommodating psychotherapy sessions that last for multi-
ple hours, for example, would require different ways of orga-
nizing healthcare.
Whether or not psychedelic treatments become mainstream
remains to be seen. However, the scale of unmet needs in
psychiatry in conjunction with highly promising results, major
ongoing trials, and the FDA’s recent support, suggest that PAP
will find clinical niches. Despite its novelty, the underlying
approach of PAP has much in common with current psychiatric
treatments, and current psychiatric treatments could be
enriched through considering some of its key tenets. For exam-
ple, a clear emphasis on optimizing intentions and expectations
of healing could benefit all existing treatments.
Significant further research will be required to move PAP
into mainstream clinics and hospitals. The various
approaches utilized to date need to be actively compared
with a wider variety of treatments, and may benefit from
utilization of more conventional psychotherapies like CBT.
Medication protocols need to be optimized in terms of dose,
numbers of sessions, and follow-up or maintenance proto-
cols. The key mechanisms of psychotherapeutic change
should be characterized and optimized, potentially by com-
ponent analysis, for specific molecules and certain clinical
populations. Such studies should be demographically
diverse and include [239], but not be limited to, treatment-
resistant subpopulations. Much work also remains to be
done to better understand the neurological mechanisms of
psychedelics, MDMA, and ketamine. Finally, long-term stu-
dies will be necessary to demonstrate that the benefits
obtained from PAP can indeed be maintained, with or with-
out repeat treatments, for various conditions.
Funding
This paper was not funded
Declaration of interest
L. Jerome is an employee of the Multidisciplinary Association for
Psychedelics Studies (MAPS) Public Benefit Corporation, as was A. Feduccia
during the preparation of the manuscript. A. Feduccia is an owner and
employee of Psychedelic.Support and consultant for Project New Day,
a 501(c)(3) not-for-profit organization. The authors have no other relevant
affiliations or financial involvement with any organization or entity with
a financial interest in or financial conflict with the subject matter or materials
discussed in the manuscript apart from those disclosed.
Reviewers disclosures
Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
ORCID
Kyle T. Greenway http://orcid.org/0000-0002-7829-493X
Nicolas Garel http://orcid.org/0000-0002-4031-9943
Lisa Jerome http://orcid.org/0000-0001-6209-8750
Allison A. Feduccia http://orcid.org/0000-0002-6918-0738
References
Papers of special note have been highlighted as either of interest (•) or of
considerable interest (••) to readers.
1. Weinberger AH, Gbedemah M, Martinez AM, et al. Trends in
depression prevalence in the USA from 2005 to 2015: widening
disparities in vulnerable groups. Psychol Med. 2018;48:1308–1315.
2. Vos T, Flaxman AD, Naghavi M, et al. Years lived with disability
(YLDs) for 1160 sequelae of 289 diseases and injuries 1990–2010:
a systematic analysis for the global burden of disease study 2010.
Lancet. 2012;380:2163–2196.
3. Friedrich MJ. Depression is the leading cause of disability around
the world. JAMA. 2017;317:1517.
4. Greenberg PE, Fournier -A-A, Sisitsky T, et al. The economic burden
of adults with major depressive disorder in the United States (2005
and 2010). J Clin Psychiatry. 2015;76:155–162.
5. Huhn M, Tardy M, Spineli LM, et al. Efficacy of pharmacotherapy
and psychotherapy for adult psychiatric disorders: A systematic
overview of meta-analyses. JAMA Psychiatry. 2014;71:706–715.
6. Demyttenaere K, Van Duppen Z. The impact of (the concept of)
treatment-resistant depression: an opinion review. International
Journal of Neuropsychopharmacology. 2019;22(2):85–92.
7. Thomas L, Kessler D, Campbell J, et al. Prevalence of
treatment-resistant depression in primary care: cross-sectional
data. British Journal of General Practice. 2013;63(617):852–858.
8. Lindsley CW. New statistics on the cost of new drug development
and the trouble with CNS drugs. ACS Chemical Neuroscience.
2014;5(12):1142.
9. Gribkoff VK, Kaczmarek LK. The need for new approaches in CNS
drug discovery: why drugs have failed, and what can be done to
improve outcomes. Neuropharmacology. 2017;120:11–19.
10. Nutt D, Goodwin G. ECNP summit on the future of CNS drug
research in Europe 2011: report prepared for ECNP by david nutt
and guy goodwin. European Neuropsychopharmacology. 2011;21
(7):495–499.
11. Corrigan-Curay J, McKee AE, Stein SP. Breakthrough-therapy desig-
nation — an FDA perspective. New England Journal of Medicine.
2018;378(15):1457–1458.
12. FDA grants Breakthrough Therapy Designation to Usona Institute’s
psilocybin program for major depressive disorder [Internet]. 2019
Cited 2019 December 16]. Available from: https://www.business
wire.com/news/home/20191122005452/en/FDA-grants-
Breakthrough-Therapy-Designation-Usona-Institutes
13. Pathways C COMPASS pathways receives FDA breakthrough ther-
apy designation for psilocybin therapy for treatment-resistant
depression [internet]. [Cited 2019 December 16]. Available from:
https://www.prnewswire.com/news-releases/compass-pathways-
receives-fda-breakthrough-therapy-designation-for-psilocybin-
therapy-for-treatment-resistant-depression-834088100.html
14. Feduccia AA, Jerome L, Yazar-Klosinski B, et al. Breakthrough for
trauma treatment: safety and efficacy of MDMA-assisted psy-
chotherapy compared to paroxetine and sertraline. Frontiers in
Psychiatry. 2019;10. DOI:10.3389/fpsyt.2019.00650.
15. Bahr R, Lopez A, Rey JA. Intranasal esketamine (spravatotm) for use
in treatment-resistant depression in conjunction with an oral
antidepressant. P T Peer-Rev. J. Formul. Manag. 2019;44:340–375.
16. Duman RS, Aghajanian GK. Synaptic dysfunction in depression:
potential therapeutic targets. Science. 2012;338(6103):68–72.
17. Scott LJ. Brexanolone: first global approval. Drugs. 2019;79
(7):779–783.
18. Orsolini L, Tomasetti C, Valchera A, et al. Current and future per-
spectives on the major depressive disorder: focus on the new
multimodal antidepressant vortioxetine. CNS & Neurological
Disorders - Drug Targets. 2017;16(1):65–92.
19. Heifets BD, Malenka RC. Disruptive psychopharmacology. JAMA
Psychiatry. 2019;76(8):775–776.
20. Schenberg EE. Psychedelic-Assisted Psychotherapy: A Paradigm
Shift in Psychiatric Research and Development.Front. Pharmacol.
2018;9:733.
10 K. T. GREENWAY ET AL.
21. Lee MA, Shlain B. Acid dreams: the complete social history of LSD:
the CIA, the sixties, and beyond. Rev. Evergreen ed. New York:
Grove Weidenfeld; 1992.
22. Hendricks PS, Crawford MS, Cropsey KL, et al. The relationships of
classic psychedelic use with criminal behavior in the United States
adult population. Journal of Psychopharmacology. 2018;32(1):37–48.
23. Muttoni S, Ardissino M, John C. Classical psychedelics for the
treatment of depression and anxiety: A systematic review. Journal
of Affective Disorders. 2019;258:11–24.
24. Cuijpers P, Sijbrandij M, Koole SL, et al. Adding psychotherapy to
antidepressant medication in depression and anxiety disorders: a
meta-analysis. World Psychiatry. 2014;13(1):56–67.
25. Watanabe N, Churchill R, Furukawa TA. Combination of psychother-
apy and benzodiazepines versus either therapy alone for panic
disorder: a systematic review. BMC Psychiatry. 2007;7(1):18.
26. Morrison AP, Law H, Carter L, et al. Antipsychotic drugs versus
cognitive behavioural therapy versus a combination of both in
people with psychosis: a randomised controlled pilot and feasibility
study. Lancet Psychiatry. 2018;5(5):411–423.
27. Katzman MA, Bleau P, Blier P, et al. Canadian clinical practice guide-
lines for the management of anxiety, posttraumatic stress and
obsessive-compulsive disorders. BMC Psychiatry. 2014;14(Suppl 1):S1.
28. Baltenberger EP, Buterbaugh WM, Martin BS, et al. Review of anti-
depressants in the treatment of neuropathic pain. Mental Health
Clinician. 2015;5(3):123–133.
29. Kułak-Bejda A, Bejda G, Waszkiewicz N. Antidepressants for irritable
bowel syndrome—A systematic review. Pharmacological Reports.
2017;69(6):1366–1379.
30. Yeragani V, Ramachandraih C, Subramanyam N, et al.
Antidepressants: from MAOIs to SSRIs and more. Indian Journal of
Psychiatry. 2011;53(2):180–182.
31. Hillhouse TM, Porter JH. A brief history of the development of
antidepressant drugs: from monoamines to glutamate.
Experimental and Clinical Psychopharmacology. 2015;23(1):1–21.
32. Hewitt K. Rehabilitating LSD history in postwar America: dilworth
Wayne Woolley and the serotonin hypothesis of mental illness.
History of Science. 2016;54(3):307–330.
33. Nichols DE, Barker EL. Psychedelics. Barker EL, editor.
Pharmacological Reviews. 2016;68(2):264–355.
•• A comprehensive review of the pharmacology and history of
psychedelics by a pioneer in the field.
34. Cipriani A, Furukawa TA, Salanti G, et al. Comparative efficacy and
acceptability of 21 antidepressant drugs for the acute treatment of
adults with major depressive disorder: a systematic review and
network meta-analysis. Lancet. 2018;391(10128):1357–1366.
35. Moore TJ, Mattison DR. Adult utilization of psychiatric drugs and
differences by sex, age, and race. JAMA Internal Medicine. 2017;177
(2):274–275.
36. Winter SE, Barber JP. Should treatment for depression be based
more on patient preference? Patient prefer. Patient Preference and
Adherence. 2013;7:1047–1057.
37. Dobscha SK, Corson K, Gerrity MS. Depression treatment prefer-
ences of VA primary care patients. Psychosomatics. 2007;48
(6):482–488.
38. Kennedy SH, Lam RW, McIntyre RS, et al. Canadian network for
mood and anxiety treatments (CANMAT) 2016 clinical guidelines
for the management of adults with major depressive disorder. The
Canadian Journal of Psychiatry. 2016;61(9):540–560.
39. Johnson CF, Williams B, MacGillivray SA, et al. ‘Doing the right
thing’: factors influencing GP prescribing of antidepressants and
prescribed doses. BMC Family Practice. 2017;18(1):72.
40. Elliott C. Listening to prozac: a psychiatrist explores antidepressant
drugs and the remaking of the self. BMJ. 1994;308(6945):1724.
41. Ramsberg J, Asseburg C, Henriksson HM. Effectiveness and
cost-effectiveness of antidepressants in primary care: a multiple
treatment comparison meta-analysis and cost-effectiveness
model. Plos One. 2012;7(8):e42003.
42. Rubio-Valera M, Peñarrubia-María MT, Iglesias-González M, et al.
Cost-effectiveness of antidepressants versus active monitoring for
mild-to-moderate major depressive disorder: a multisite
non-randomized-controlled trial in primary care (INFAP study).
The European Journal of Health Economics. 2019;20(5):703–713.
43. Ross EL, Vijan S, Miller EM, et al. The cost-effectiveness of cognitive
behavioral therapy versus second-generation antidepressants for
initial treatment of major depressive disorder in the United States:
a decision analytic model. Annals of Internal Medicine. 2019;171
(11):785.
44. Andrews PW, Thomson JA, Amstadter A, et al. Primum Non Nocere:
an Evolutionary Analysis of Whether Antidepressants Do More
Harm than Good. Front. Psychol. 2012;3:117.
45. Kirsch I. Placebo Effect in the Treatment of Depression and Anxiety.
Front. Psychiatry. 2019;10:407.
46. Hengartner MP. Methodological Flaws. Conflicts of Interest, and
Scientific Fallacies: implications for the Evaluation of
Antidepressants’ Efficacy and Harm. Front. Psychiatry. 2017;8:275.
47. Østergaard SD. Do not blame the SSRIs: blame the Hamilton
Depression Rating Scale.Acta Neuropsychiatr. 2017;30(5):241–243.
48. Moncrieff J, Wessely S, Hardy R. Active placebos versus antidepres-
sants for depression. cochrane common mental disorders group,
editor. Cochrane Database of Systematic Reviews. 2004.
DOI:10.1002/14651858.CD003012.pub2
49. Sansone RA, Sansone LA, Adherence A. Innov. Clin Neurosci.
2012;9:41–46.
50. Fava GA. Rational use of antidepressant drugs. Psychotherapy and
Psychosomatics. 2014;83(4):197–204.
51. Cartwright C, Gibson K, Read J, et al. Long-term antidepressant use:
patient perspectives of benefits and adverse effects. <![cdata
[patient Preference and Adherence]]>. 2016;10:1401–1407.
52. Steinkamp JM, Goldblatt N, Borodovsky JT, et al. Technological
Interventions for medication adherence in adult mental health
and substance use disorders: A systematic review. JMIR Mental
Health. 2019;6(3):e12493.
53. Keller MB, Hirschfeld RMA, Demyttenaere K, et al. Optimizing out-
comes in depression: focus on antidepressant compliance.
International Clinical Psychopharmacology. 2002;17(6):265–271.
54. Masand PS. Tolerability and adherence issues in antidepressant
therapy. Clinical Therapeutics. 2003;25(8):2289–2304.
55. Clayton DO, Shen WW. Psychotropic Drug?Induced sexual function
disorders. Drug Saf. 1998;19(4):299–312.
56. Brown C, Battista D, Bruehlman R, et al. Beliefs About
Antidepressant Medications in Primary Care Patients: relationship
to Self-Reported Adherence. Medical Care. 2005;43(12):1203–1207.
57. Iacobucci G. NHS prescribed record number of antidepressants last
year. BMJ. 2019;364:l1508.
58. Moore M, Yuen HM, Dunn N, et al. Explaining the rise in antide-
pressant prescribing: a descriptive study using the general practice
research database. BMJ. 2009;339(oct15 2):b3999.
59. Fava GA, Belaise C. Discontinuing antidepressant drugs: lesson
from a failed trial and extensive clinical experience.
Psychotherapy and Psychosomatics. 2018;87(5):257–267.
60. Wentink C, Huijbers MJ, Lucassen P, et al. Discontinuation of anti-
depressant medication in primary care supported by monitoring
plus mindfulness-based cognitive therapy versus monitoring alone:
design and protocol of a cluster randomized controlled trial. BMC
Family Practice. 2019;20(1):105.
61. Davies J, Read J. A systematic review into the incidence, severity
and duration of antidepressant withdrawal effects: are guidelines
evidence-based? Addict. Behav. 2019;97:111–121.
62. Johnson MW. Psychiatry might need some psychedelic therapy.
International Review of Psychiatry. 2018;30(4):285–290.
63. Liu B, Liu J, Wang M, et al. From serotonin to neuroplasticity:
evolvement of theories for major depressive disorder. Frontiers in
Cellular Neuroscience. 2017;11:305.
64. Felger JC. Role of inflammation in depression and treatment
implications. Handb Exp Pharmacol. 2019;250:255–286.
65. Campbell LF, Norcross JC, Vasquez MJT, et al. Recognition of psy-
chotherapy effectiveness: the APA resolution. Psychotherapy.
2013;50(1):98–101.
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 11
66. Imel ZE, Malterer MB, McKay KM, et al. A meta-analysis of psy-
chotherapy and medication in unipolar depression and dysthymia.
Journal of Affective Disorders. 2008;110(3):197–206.
67. Craighead WE, Dunlop BW, Craighead WE, Dunlop BW.
Combination Psychotherapy and. Combination Psychotherapy
and antidepressant medication treatment for depression: for
whom, when, and how. . Annual Review of Psychology. 2014;65
(1):267–300.
68. Crawford MJ, Thana L, Farquharson L, et al. Patient experience of
negative effects of psychological treatment: results of a national
survey. British Journal of Psychiatry. 2016;208(3):260–265.
69. Kamenov K, Twomey C, Cabello M, et al. The efficacy of psychother-
apy, pharmacotherapy and their combination on functioning and
quality of life in depression: a meta-analysis. Psychological
Medicine. 2017;47(3):414–425.
70. McHugh RK, Whitton SW, Peckham AD, et al. Patient preference for
psychological vs pharmacologic treatment of psychiatric disorders:
a meta-analytic review. The Journal of Clinical Psychiatry. 2013;74
(6):595–602.
71. Castelnuovo G, Pietrabissa G, Cattivelli R, et al. Not only clinical
efficacy in psychological treatments: clinical psychology must pro-
mote cost-benefit, cost-effectiveness, and cost-utility analysis.
Frontiers in Psychology. 2016;7:563.
72. Emmelkamp PMG, David D, Beckers T, et al. Advancing psychother-
apy and evidence-based psychological interventions: advancing
psychotherapy. Int J Methods Psychiatr Res. 2014;23:58–91.
73. Clark DM, Layard R, Smithies R, et al. Improving access to psycho-
logical therapy: initial evaluation of two UK demonstration sites.
Behav Res Ther. 2009;47:910–920.
74. Layard R, Clark DM. Why more psychological therapy would cost
nothing. Front Psychol. 2015;6:1713.
75. Rock PL, Roiser JP, Riedel WJ, et al. Cognitive impairment in depres-
sion: a systematic review and meta-analysis. Psychol Med.
2014;44:2029–2040.
76. Cuijpers P, Cristea IA, Karyotaki E, et al. How effective are cognitive
behavior therapies for major depression and anxiety disorders? A
meta-analytic update of the evidence. World Psychiatry.
2016;15:245–258.
77. Amick HR, Gartlehner G, Gaynes BN, et al. Comparative benefits
and harms of second generation antidepressants and cognitive
behavioral therapies in initial treatment of major depressive dis-
order: systematic review and meta-analysis. BMJ. 2015;351:h6019.
78. Frank JD. Persuasion and healing: A comparative study of psy-
chotherapy. Thierd ed. Baltimore: Hopkins Univ. Press; 1991.
79. Brent DA, Kolko DJ. Psychotherapy: definitions, mechanisms of
action, and relationship to etiological models. J Abnorm Child
Psychol. 1998;26:17–25.
80. Freud S, Brill AA. The history of the psychoanalytic movement.
Whitefish, Mont.: Kessinger Pub.; 2007.
81. Cautin RL. A century of psychotherapy, 1860–1960. In: editors,
Norcross JC, VandenBos GR, Freedheim DK. Hist. psychother. con-
tin. change. 2nd ed. Washington: American Psychological
Association; 2011.
82. Robertson D. The philosophy of cognitive-behavioural therapy
(CBT): stoic philosophy as rational and cognitive psychotherapy.
London: Karnac; 2010.
83. Clifford T. Tibetan Buddhist medicine and psychiatry: the diamond
healing. York Beach, Me: S. Weiser; 1984.
84. Cuijpers P, Reijnders M, Huibers MJH. The role of common factors
in psychotherapy outcomes. Annu Rev Clin Psychol.
2019;15:207–231.
85. Wampold BE. How important are the common factors in psy-
chotherapy? An update. World Psychiatry. 2015;14:270–277.
•• An extensive review of psychotherapy’s evidence base and its
therapeutic mechanisms, including the contextual model.
86. Budd R, Hughes I. The Dodo Bird Verdict-controversial, inevitable
and important: a commentary on 30 years of meta-analyses: the
dodo bird verdict: a commentary. Clin Psychol Psychother.
2009;16:510–522.
87. Rosenzweig S. Some implicit common factors in diverse methods
of psychotherapy. Am J Orthopsychiatry. 1936;6:412–415.
88. Brown J. Specific techniques vs. common factors? Psychotherapy
integration and its role in ethical practice. Am J Psychother.
2015;69:301–316.
89. Frank JD. Therapeutic factors in psychotherapy. Am J Psychother.
1971;25:350–361.
90. Goldstone D. Cognitive-behavioural therapy versus psychodynamic
psychotherapy for the treatment of depression: a critical review of
evidence and current issues. South Afr J Psychol. 2017;47:84–96.
91. Foa EB. Cognitive behavioral therapy of obsessive-compulsive
disorder. Dialogues Clin Neurosci. 2010;12:199–207.
92. Keefe JR, McCarthy KS, Dinger U, et al. A meta-analytic review of
psychodynamic therapies for anxiety disorders. Clin Psychol Rev.
2014;34:309–323.
93. Pampallona S, Bollini P, Tibaldi G, et al. Combined pharmacother-
apy and psychological treatment for depression: a systematic
review. Arch Gen Psychiatry. 2004;61:714–719.
94. Mullen LS, Blanco C, Vaughan SC, et al. Defense mechanisms and
personality in depression. Depress Anxiety. 1999;10:168–174.
95. Cristea IA, Huibers MJH, David D, et al. The effects of cognitive
behavior therapy for adult depression on dysfunctional thinking: A
meta-analysis. Clin Psychol Rev. 2015;42:62–71.
96. DeRubeis RJ, Siegle GJ, Hollon SD. Cognitive therapy vs. medica-
tions for depression: treatment outcomes and neural mechanisms.
Nat Rev Neurosci. 2008;9:788–796.
97. Wampold BE, Imel ZE. The great psychotherapy debate: the evi-
dence for what makes psychotherapy work. 2nd ed. New York, NY,
US: Routledge/Taylor & Francis Group; 2015.
98. Kirsch I, Moore TJ, Scoboria A, et al. The emperor’s new drugs: an
analysis of antidepressant medication data submitted to the U.S.
Food and Drug Administration. Prev. Treat. 2002;5(1):5.
99. Leuchter AF, Hunter AM, Tartter M, et al. Role of pill-taking, expec-
tation and therapeutic alliance in the placebo response in clinical
trials for major depression. Br J Psychiatry. 2014;205:443–449.
A unique three-armed randomized trial comparing supportive
care to psychotherapy combined with either antidepressant or
placebo treatment.
100. Faria V, Gingnell M, Hoppe JM, et al. Do you believe it? Verbal
suggestions influence the clinical and neural effects of escitalo-
pram in social anxiety disorder: a randomized trial. EBioMedicine.
2017;24:179–188.
A unique randomized trial investigating the clinical and neural
consequences of explanatory frameworks.
101. Zilcha-Mano S, Roose SP, Barber JP, et al. Therapeutic alliance in
antidepressant treatment: cause or effect of symptomatic levels?
Psychother. Psychosom. 2015;84:177–182.
102. Ankarberg P, Falkenström F. Treatment of depression with antide-
pressants is primarily a psychological treatment. Psychother Theory
Res Pract Train. 2008;45:329–339.
103. Chiarotti F, Viglione A, Giuliani A, et al. Citalopram amplifies the
influence of living conditions on mood in depressed patients
enrolled in the STAR*D study. Transl Psychiatry. 2017;7:tp201735.
104. Viglione A, Chiarotti F, Poggini S, et al. Predicting antidepressant
treatment outcome based on socioeconomic status and citalopram
dose. Pharmacogenomics J. 2019;19:538.
105. Gaynes BN, Warden D, Trivedi MH, et al. What did STAR*D teach us?
Results from a large-scale, practical, clinical trial for patients with
depression. Psychiatr Serv. 2009;60:1439–1445.
106. Furukawa TA, Cipriani A, Cowen PJ, et al. Optimal dose of selective
serotonin reuptake inhibitors, venlafaxine, and mirtazapine in
major depression: a systematic review and dose-response
meta-analysis. Lancet Psychiatry. 2019;6:601–609.
107. Alboni S, van Dijk RM, Poggini S, et al. Fluoxetine effects on
molecular, cellular and behavioral endophenotypes of depression
are driven by the living environment. Mol Psychiatry.
2017;22:552–561.
108. Angermann CE, Gelbrich G, Störk S, et al. Effect of escitalopram on
all-cause mortality and hospitalization in patients with heart failure
12 K. T. GREENWAY ET AL.
and depression: the MOOD-HF randomized clinical trial. JAMA.
2016;315:2683.
109. Palmer SC, Natale P, Ruospo M, et al. Antidepressants for treating
depression in adults with end-stage kidney disease treated with
dialysis.Cochrane Database Syst Rev. 2016;5:CD004541.
110. Charney DA, Heath LM, Zikos E, et al. Poorer drinking outcomes
with citalopram treatment for alcohol dependence: a randomized,
double-blind, placebo-controlled trial. Alcohol Clin Exp Res.
2015;39:1756–1765.
111. Nunes EV, Levin FR. Treatment of depression in patients with
alcohol or other drug dependence: a meta-analysis. JAMA.
2004;291:1887–1896.
112. Prosser A, Helfer B, Leucht S. Biological v. psychosocial treatments:
a myth about pharmacotherapy v. psychotherapy. Br J Psychiatry.
2016;208:309–311.
113. Barsaglini A, Sartori G, Benetti S, et al. The effects of psychotherapy
on brain function: A systematic and critical review. Prog Neurobiol.
2014;114:1–14.
114. Koch JM, Hinze-Selch D, Stingele K, et al. Changes in CREB phos-
phorylation and BDNF plasma levels during psychotherapy of
depression. Psychother Psychosom. 2009;78:187–192.
115. Roberts S, Keers R, Lester KJ, et al. HPA AXIS RELATED GENES AND
RESPONSE TO PSYCHOLOGICAL THERAPIES: GENETICS AND
EPIGENETICS. Depress Anxiety. 2015;32:861–870.
116. Perroud N, Salzmann A, Prada P, et al. Response to psychotherapy
in borderline personality disorder and methylation status of the
BDNF gene. Transl Psychiatry. 2013;3:e207.
117. Stahl SM. Psychotherapy as an epigenetic “drug”: psychiatric ther-
apeutics target symptoms linked to malfunctioning brain circuits
with psychotherapy as well as with drugs. J Clin Pharm Ther.
2012;37:249–253.
118. Doblin RE, Christiansen M, Jerome L, et al. The past and future of
psychedelic science: an introduction to this issue. J Psychoactive
Drugs. 2019;51:93–97.
119. Pollan M. How to change your mind: what the new science of
psychedelics teaches us about consciousness, dying, addiction,
depression, and transcendence. New York: Penguin Press; 2018.
120. Liechti ME. Modern clinical research on LSD.
Neuropsychopharmacology. 2017;42:2114–2127.
121. Freud S. The interpretation of dreams. New York: Barnes & Noble
Books; 1994.
122. Kaplan RM. Humphry osmond: the psychedelic psychiatrist.
Int J Humanit Soc Sci Educ.
123. Waltz M. From ‘Pathological Motherhood’ to Refrigerator Mothers.
In: Waltz M, editor. Autism soc. med. hist. London: Palgrave
Macmillan UK; 2013; p. 73–86.
124. Stockings GT. A clinica study of the mescaline psychosis, with
special reference to the mechanism of the genesis of schizophrenic
and other psychotic states. J Nerv Ment Dis. 1940;92:678.
125. Ban TA. Fifty years chlorpromazine: a historical perspective.
Neuropsychiatr Dis Treat. 2007;3:495–500.
126. Woolley DW, Shaw E, Biochemical A. Pharmacological suggestion
about certain mental disorders. Proc Natl Acad Sci.
1954;40:228–231.
127. Lindemann E. Psychological changes in normal and abnormal indi-
viduals under the influence of sodium amytal. Am J Psychiatry.
1932;88:1083–1091.
128. Meduna LJ. Carbon dioxide therapy: A neurophysiological treat-
ment of nervous disorders. Springfield: Charles C Thomas Publisher;
1950.
129. Grinspoon L, Reconsidered: LSD. Should clinical research be
resumed? The Sciences. 1981;21:20–23.
130. Blewet DB, Chwelos MD Handbook for the therapeutic use of LSD-25
[Internet]. 1959 [Cited 2020 January 1]. Available from: https://www.
erowid.org/psychoactives/guides/handbook_lsd25.shtml
131. Sandison RA, Spencer AM, Whitelaw JDA. The therapeutic value of
lysergic acid diethylamide in mental illness. J Ment Sci.
1954;100:491–507.
132. Sandison RA. Psychological aspects of the lsd treatment of the
neuroses. J Ment Sci. 1954;100:508–515.
133. Kraehenmann R, Pokorny D, Vollenweider L, et al. Dreamlike effects of
LSD on waking imagery in humans depend on serotonin 2A receptor
activation. Psychopharmacology (Berl). 2017;234:2031–2046.
134. Barrett FS, Preller KH, Herdener M, et al. Serotonin 2A receptor
signaling underlies lsd-induced alteration of the neural response to
dynamic changes in music. Cereb Cortex. 2018;28:3939–3950.
135. Griffiths RR, Johnson MW, Richards WA, et al. Psilocybin occasioned
mystical-type experiences: immediate and persisting dose-related
effects. Psychopharmacology (Berl). 2011;218:649–665.
136. Osório F, Sanches RF DL, LR M, et al. Antidepressant effects of
a single dose of ayahuasca in patients with recurrent depression:
a preliminary report. Rev Bras Psiquiatr. 2015;37:13–20.
137. Johnson MW, Richards WA, Griffiths RR. Human hallucinogen
research: guidelines for safety. J Psychopharmacol Oxf Engl.
2008;22:603–620.
•• A succinct overview on the historical and modern applications
of psychedelic-assisted psychotherapy with an emphasis on
safe research practices.
138. Studerus E, Gamma A, Vollenweider FX. Psychometric evaluation of
the altered states of consciousness rating scale (OAV). Bell V, editor.
PLoS ONE. 2010;5:e12412.
A rigorous psychometric analysis that compares and contrasts
the phenomenology of psilocybin, MDMA, and ketamine.
139. Barrett FS, Johnson MW, Griffiths RR. Validation of the revised
mystical experience questionnaire in experimental sessions with
psilocybin. J Psychopharmacol Oxf Engl. 2015;29:1182–1190.
140. Carhart-Harris RL, Muthukumaraswamy S, Roseman L, et al. Neural
correlates of the LSD experience revealed by multimodal
neuroimaging. Proc Natl Acad Sci U S A. 2016;113:4853–4858.
141. Holze F, Vizeli P, Müller F, et al. Distinct acute effects of LSD,
MDMA, and d-amphetamine in healthy subjects.
Neuropsychopharmacology. 2019;45:462.
142. Barrett FS, Griffiths RR, Hallucinogens C. Mystical experiences: phe-
nomenology and neural correlates. Curr Top Behav Neurosci.
2018;36:393–430.
143. Carhart-Harris RL, Kaelen M, Whalley MG, et al. LSD enhances
suggestibility in healthy volunteers. Psychopharmacology (Berl).
2015;232:785–794.
144. Dolder PC, Schmid Y, Müller F, et al. LSD Acutely Impairs Fear
Recognition and Enhances Emotional Empathy and Sociality.
Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol.
2016;41:2638–2646.
145. Roseman L, Haijen E, Idialu-Ikato K, et al. Emotional breakthrough
and psychedelics: validation of the Emotional Breakthrough
Inventory. J Psychopharmacol (Oxf.). 2019;33:1076–1087.
146. Kaelen M, Barrett FS, Roseman L, et al. LSD enhances the emotional
response to music. Psychopharmacology (Berl).
2015;232:3607–3614.
147. Preller KH, Herdener M, Pokorny T, et al. The Fabric of Meaning and
Subjective Effects in LSD-Induced States Depend on Serotonin 2A
Receptor Activation. Curr Biol. 2017;27:451–457.
148. Griffiths RR, Johnson MW, Carducci MA, et al. Psilocybin produces
substantial and sustained decreases in depression and anxiety in
patients with life-threatening cancer: A randomized double-blind
trial. J Psychopharmacol (Oxf.). 2016;30:1181–1197.
149. Carhart-Harris RL, Leech R, Hellyer PJ, et al. The entropic brain:
a theory of conscious states informed by neuroimaging research
with psychedelic drugs. Front Hum Neurosci. 2014;8:20.
150. Buckner RL, Andrews-Hanna JR, Schacter DL. The brain’s default
network: anatomy, function, and relevance to disease. Year Cogn.
Neurosci. 2008. Malden: Blackwell Publishing; 2008.
151. Andrews-Hanna JR The brain’s default network and its adaptive
role in internal mentation - jessica r. andrews-hanna, 2012. the
neuroscientist. 2011
152. Karapanagiotidis T, Bernhardt BC, Jefferies E, et al. Tracking
thoughts: exploring the neural architecture of mental time travel
during mind-wandering. NeuroImage. 2017;147:272–281.
153. Millière R, Carhart-Harris RL, Roseman L, et al. Psychedelics,
Meditation, and Self-Consciousness. Front. Psychol. 2018;9:1475.
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 13
154. Carhart-Harris RL, Friston KJ. The default-mode, ego-functions and
free-energy: a neurobiological account of Freudian ideas. Brain.
2010;133:1265–1283.
155. Peters SK, Dunlop K, Downar J. Cortico-striatal-thalamic loop cir-
cuits of the salience network: a central pathway in psychiatric
disease and treatment. Front Syst Neurosci. 2016;10:104.
156. Preller KH, Razi A, Zeidman P, et al. Effective connectivity changes
in LSD-induced altered states of consciousness in humans. Proc
Natl Acad Sci U S A. 2019;116:2743–2748.
157. Grof S, Hofmann A, Weil A LSD psychotherapy. ben lomond, calif.:
multidisciplinary association for psychedelic studies; 2008.
Among the most widely read and cited references for psyche-
delic-assisted psychotherapy.
158. Carhart-Harris RL, Friston KJREBUS. and the anarchic brain: toward
a unified model of the brain action of psychedelics. Pharmacol Rev.
2019;71:316–344.
159. Müller F, Dolder PC, Schmidt A, et al. Altered network hub con-
nectivity after acute LSD administration. NeuroImage Clin.
2018;18:694–701.
160. Geyer M, Vollenweider F. Serotonin research: contributions to
understanding psychoses. Trends Pharmacol Sci. 2008;29:445–453.
161. de la Torre R, Farré M, Roset PN, et al. Human Pharmacology of
MDMA: pharmacokinetics, metabolism, and disposition. Ther Drug
Monit. 2004;26:137–144.
162. Carhart-Harris RL, Wall MB, Erritzoe D, et al. The effect of acutely admi-
nistered MDMA on subjective and BOLD-fMRI responses to favourite
and worst autobiographical memories. Int J Neuropsychopharmacol.
2014;17:527–540.
163. Ballesta S, Reymond G, Pozzobon M, et al. Effects of MDMA
Injections on the behavior of socially-housed long-tailed macaques
(macaca fascicularis). PLoS ONE. 2016;11:e0147136.
164. Kirkpatrick M, Delton AW, Robertson TE, et al. Prosocial effects of
MDMA: A measure of generosity. J Psychopharmacol Oxf Engl.
2015;29:661–668.
165. Greer G, Tolbert R. Subjective reports of the effects of MDMA in
a clinical setting. J Psychoactive Drugs. 1986;18:319–327.
166. Passie T. The early use of MDMA (‘Ecstasy’) in psychotherapy (-
1977–1985). Drug Sci Policy Law. 2018;4:2050324518767442.
167. Lur G, Fariborzi M, Higley MJ. Ketamine disrupts neuromodulatory
control of glutamatergic synaptic transmission Adams MM, editor.
PLOS ONE. 2019;14:e0213721.
168. Sleigh J, Harvey M, Voss L, et al. Ketamine – more mechanisms of
action than just NMDA blockade. Trends Anaesth Crit Care.
2014;4:76–81.
169. Moda-Sava RN, Murdock MH, Parekh PK, et al. Sustained rescue of
prefrontal circuit dysfunction by antidepressant-induced spine
formation. Science. 2019;364:eaat8078.
170. Haile CN, Murrough JW, Iosifescu DV, et al. Plasma brain derived neuro-
trophic factor (BDNF) and response to ketamine in treatment-resistant
depression. Int J Neuropsychopharmacol. 2014;17:331–336.
171. Domino EF. Taming the ketamine tiger. 1965 Anesthesiology.
2010;113:678.
172. . Hurth KP, Jaworski A, Thomas KB. et al. The reemergence of ketamine
for treatment in critically ill adults: crit. Care Med. 2020; 1.
173. Berman RM, Cappiello A, Anand A, et al. Antidepressant effects of
ketamine in depressed patients. Biol Psychiatry. 2000;47:351–354.
174. Kryst J, Kawalec P, Mitoraj AM, et al. Efficacy of single and repeated
administration of ketamine in unipolar and bipolar depression: a meta-
analysis of randomized clinical trials. Pharmacol. Rep. 2020.
DOI:10.1007/s43440-020-00097-z
175. . Glue P, Neehoff S, Sabadel A. et al. Effects of ketamine in patients
with treatment-refractory generalized anxiety and social anxiety
disorders: exploratory double-blind psychoactive-controlled repli-
cation study. J Psychopharmacol (Oxf.). 2020;34(3):267.
176. Hartberg J, Garrett-Walcott S, De Gioannis A. Impact of oral keta-
mine augmentation on hospital admissions in treatment-resistant
depression and PTSD: a retrospective study. Psychopharmacology
(Berl). 2018;235:393–398.
177. Wajs E, Aluisio L, Holder R, et al. Esketamine nasal spray plus oral
antidepressant in patients with treatment-resistant depression:
assessment of long-term safety in a phase 3, open-label study
(SUSTAIN-2). J Clin Psychiatry. 2020;81.
178. Singh JB, Fedgchin M, Daly EJ, et al. A double-blind, randomized,
placebo-controlled, dose-frequency study of intravenous ketamine in
patients with treatment-resistant depression. Am J Psychiatry.
2016;173:816–826.
179. Phillips JL, Norris S, Talbot J, et al. Single, repeated, and mainte-
nance ketamine infusions for treatment-resistant depression:
a randomized controlled trial. Am J Psychiatry. 2019;176:401–409.
180. Kolp E, Friedman HL. University of florida, et al. ketamine psyche-
delic psychotherapy: focus on its pharmacology, phenomenology,
and clinical applications. Int J Transpers Stud. 2014;33:84–140.
181. Dore J, Turnipseed B, Dwyer S, et al. Ketamine assisted psychother-
apy (KAP): patient demographics, clinical data and outcomes in
three large practices administering ketamine with psychotherapy.
J Psychoactive Drugs. 2019;51:189–198.
182. Busch AK, Johnson WCLSD 25 as an aid in psychotherapy; preli-
minary report of a new drug. Dis. Nerv. Syst. 1950;11:241–243.
183. Grof S, Goodman LE, Richards WA, et al. LSD-assisted psychother-
apy in patients with terminal cancer. Int Pharmacopsychiatry.
1973;8:129–144.
184. Elmer LD, MacDonald DA, Friedman HL. Transpersonal psychology,
physical health, and mental health: theory, research, and practice.
Humanist Psychol. 2003;31:159–181.
185. Kasprow MC, Scotton BW. A review of transpersonal theory and its
application to the practice of psychotherapy. J Psychother P Res.
1999;8:12.
186. Bonny HL, Pahnke WN. The use of music in psychedelic (LSD)
psychotherapy. J Music Ther. 1972;9:64–87.
187. Ross S, Bossis A, Guss J, et al. Rapid and sustained symptom
reduction following psilocybin treatment for anxiety and depres-
sion in patients with life-threatening cancer: a randomized con-
trolled trial. J Psychopharmacol (Oxf.). 2016;30:1165–1180.
188. Mithoefer MC A manual for MDMA-assisted psychotherapy in the
treatment of posttraumatic stress disorder. 8.1. 1115 Mission Street
Santa Cruz, CA 95060: MAPS Public Benefit Corporation; 2017.
189. Kolp E, Young MS. Florida mental health institute of university of
south florida, et al. ketamine-enhanced psychotherapy: preliminary
clinical observations on its effects in treating death anxiety.
Int J Transpers Stud. 2007;26:1–17.
190. Klavetter RE, Mogar RE. Peak experiences: investigation of their
relationship to psychedelic therapy and self-actualization.
J Humanist Psychol. 1967;7:171–177.
191. Taking Psychedelics BI, Seriously J. Palliat. Med. 2018;21:417–421.
192. Watts R, Luoma JB. The use of the psychological flexibility model to
support psychedelic assisted therapy. J Context Behav Sci.
2020;15:92–102.
193. Leary T, Metzner R, Dass R, et al. The psychedelic experience:
a manual based on the Tibetan book of the dead. New York:
Citadel Press; 2007.
194. Richards WA. Sacred knowledge: psychedelics and religious experi-
ences. New York (NY): Columbia University Press; 2015.
195. Hartogsohn I. Constructing drug effects: A history of set and
setting. Drug Sci Policy Law. 2017;3:2050324516683325.
196. Strassman R. DMT: the spirit molecule: a doctor’s revolutionary
research into the biology of near-death and mystical experiences.
Rochester, Vt: Park Street Press; 2001.
197. Carhart-Harris RL, Roseman L, Haijen E, et al. Psychedelics and the
essential importance of context. J Psychopharmacol (Oxf.).
2018;32:725–731.
198. Winnicott DW. Transitional objects and transitional phenomena;
a study of the first not-me possession. Int J Psychoanal.
1953;34:89–97.
199. Gross R, Sasson Y, Zarhy M, et al. Healing environment in psychia-
tric hospital design. Gen Hosp Psychiatry. 1998;20:108–114.
200. Dijkstra K, Pieterse M, Pruyn A. Physical environmental stimuli that
turn healthcare facilities into healing environments through psy-
chologically mediated effects: systematic review. J Adv Nurs.
2006;56:166–181.
14 K. T. GREENWAY ET AL.
201. PS S, van Der, Dusseldorp E, FM K, et al. Impact of the physical
environment of psychiatric wards on the use of seclusion. Br
J Psychiatry. 2013;202:142–149.
202. Zhao K, Bai ZG, Bo A, et al. A systematic review and meta-analysis
of music therapy for the older adults with depression. Int J Geriatr
Psychiatry. 2016;31:1188–1198.
203. Leubner D, Hinterberger T. Reviewing the effectiveness of music
interventions in treating depression. Front Psychol. 2017;8:1109.
204. Hoffer A. D-Lysergic acid diethylamide (LSD): A review of its present
status. Clin Pharmacol Ther. 1965;6:183–255.
205. Watts R, Day C, Krzanowski J, et al. Patients’ Accounts of Increased
“Connectedness” and “Acceptance” After Psilocybin for Treatment-
Resistant Depression. J Humanist Psychol. 2017;57:520–564.
206. Kaelen M, Giribaldi B, Raine J, et al. The hidden therapist: evidence
for a central role of music in psychedelic therapy.
Psychopharmacology (Berl). 2018;235:505–519.
207. Richards DA, Ekers D, McMillan D, et al. Cost and outcome of
behavioural activation versus cognitive behavioural therapy for
depression (COBRA): a randomised, controlled, non-inferiority
trial. Lancet. 2016;388:871–880.
208. Moreno FA, Wiegand CB, Taitano EK, et al. Safety, tolerability, and
efficacy of psilocybin in 9 patients with obsessive-compulsive
disorder. J Clin Psychiatry. 2006;67:1735–1740.
209. Grob CS, Danforth AL, Chopra GS, et al. PIlot study of psilocybin
treatment for anxiety in patients with advanced-stage cancer. Arch
Gen Psychiatry. 2011;68:71–78.
210. Johnson MW, Garcia-Romeu A, Cosimano MP, et al. Pilot Study of
the 5-HT2AR Agonist Psilocybin in the Treatment of Tobacco
Addiction. J Psychopharmacol Oxf Engl. 2014;28:983–992.
211. Carhart-Harris RL, Bolstridge M, Rucker J, et al. Psilocybin with
psychological support for treatment-resistant depression: an
open-label feasibility study. Lancet Psychiatry. 2016;3:619–627.
212. Griffiths RR, Johnson MW, Richards WA, et al. Psilocybin-
occasioned mystical-type experience in combination with medita-
tion and other spiritual practices produces enduring positive
changes in psychological functioning and in trait measures of
prosocial attitudes and behaviors. J Psychopharmacol (Oxf.).
2018;32:49–69.
213. Lyons T, Carhart-Harris RL. Increased nature relatedness and decreased
authoritarian political views after psilocybin for treatment-resistant
depression. J Psychopharmacol (Oxf.). 2018;32:811–819.
214. Erritzoe D, Roseman L, Nour MM, et al. Effects of psilocybin therapy
on personality structure. Acta Psychiatr Scand. 2018;138:368–378.
215. Studerus E, Gamma A, Kometer M, et al. Prediction of psilocybin
response in healthy volunteers. PLoS ONE. 2012;7:e30800.
216. Haijen ECHM, Kaelen M, Roseman L, et al. Predicting responses to
psychedelics: a prospective study. Front Pharmacol. 2018;9:897.
217. Hendricks PS, Clark CB, Johnson MW, et al. Hallucinogen use pre-
dicts reduced recidivism among substance-involved offenders
under community corrections supervision. J Psychopharmacol
(Oxf.). 2014;28:62–66.
218. Walsh Z, Hendricks PS, Smith S, et al. Hallucinogen use and inti-
mate partner violence: prospective evidence consistent with pro-
tective effects among men with histories of problematic substance
use. J Psychopharmacol (Oxf.). 2016;30:601–607.
219. . Thiessen MS, Walsh Z, Bird BM. et al. Psychedelic use and intimate
partner violence: the role of emotion regulation.
J Psychopharmacol (Oxf.). 2018; 0269881118771782.
220. Passie T, Halpern JH, Stichtenoth DO, et al. The pharmacology of
lysergic acid diethylamide: a review. CNS Neurosci Ther.
2008;14:295–314.
221. Lowe LM, Peterson BL, Couper FJ. A case review of the first analy-
tically confirmed 25i-nbome-related death in washington state.
J Anal Toxicol. 2015;39:668–671.
222. Nutt DJ, King LA, Phillips LD. Drug harms in the UK: a multicriteria
decision analysis. Lancet. 2010;376:1558–1565.
223. Krebs TS, Johansen PO. Lysergic acid diethylamide (LSD) for alco-
holism: meta-analysis of randomized controlled trials.
J Psychopharmacol (Oxf.). 2012;26:994–1002.
224. Mithoefer MC, Feduccia AA, Jerome L, et al. MDMA-assisted psy-
chotherapy for treatment of PTSD: study design and rationale for
phase 3 trials based on pooled analysis of six phase 2 randomized
controlled trials. Psychopharmacology (Berl). 2019;236:2735–2745.
225. Mithoefer MC, Wagner MT, Mithoefer AT, et al. The safety and
efficacy of ±3,4-methylenedioxymethamphetamine-assisted psy-
chotherapy in subjects with chronic, treatment-resistant posttrau-
matic stress disorder: the first randomized controlled pilot study.
J Psychopharmacol (Oxf.). 2011;25:439–452.
226. Mithoefer MC, Wagner MT, Mithoefer AT, et al. Durability of
improvement in post-traumatic stress disorder symptoms and
absence of harmful effects or drug dependency after
3,4-methylenedioxymethamphetamine-assisted psychotherapy:
a prospective long-term follow-up study. J Psychopharmacol Oxf
Engl. 2013;27:28–39.
227. Danforth AL, Grob CS, Struble C, et al. Reduction in social anxiety
after MDMA-assisted psychotherapy with autistic adults:
a randomized, double-blind, placebo-controlled pilot study.
Psychopharmacology (Berl). 2018;235:3137–3148.
228. Cooper MD, Rosenblat JD, Cha DS, et al. Strategies to mitigate
dissociative and psychotomimetic effects of ketamine in the treat-
ment of major depressive episodes: a narrative review. World J Biol
Psychiatry. 2017;18:410–423.
229. Ross C, Jain R, Bonnett CJ, et al. High-dose ketamine infusion for
the treatment of posttraumatic stress disorder in combat veterans.
Ann Clin Psychiatry. 2019;31:271.
230. Kolp E, Friedman HL, Young MS, et al. Ketamine enhanced psy-
chotherapy: preliminary clinical observations on its effectiveness in
treating alcoholism. Humanist Psychol. 2006;34:399–422.
231. Krupitsky E, Burakov A, Romanova T, et al. Ketamine psychotherapy
for heroin addiction: immediate effects and two-year follow-up.
J Subst Abuse Treat. 2002;23:273–283.
232. Dakwar E, Nunes EV, Hart CL, et al. A single ketamine infusion
combined with mindfulness-based behavioral modification to
treat cocaine dependence: a randomized clinical trial. Am
J Psychiatry. 2019;176:923.
233. Liu Y, Lin D, Wu B, et al. Ketamine abuse potential and use disorder.
Brain Res Bull. 2016;126:68–73.
234. McAndrew A, Lawn W, Stevens T, et al. A proof-of-concept inves-
tigation into ketamine as a pharmacological treatment for alcohol
dependence: study protocol for a randomised controlled trial.
Trials. 2017;18:159.
235. Dakwar E, Nunes EV, Hart CL, et al. A sub-set of psychoactive effects
may be critical to the behavioral impact of ketamine on cocaine
use disorder: results from a randomized, controlled laboratory
study. Neuropharmacology. 2018;142:270–276.
236. Dakwar E, Anerella C, Hart CL, et al. Therapeutic infusions of
ketamine: do the psychoactive effects matter? Drug Alcohol
Depend. 2014;136:153–157.
• A three-armed randomized trial investigating how ketamine’s
psychoactive effects potentially mediate anti-addiction
properties.
237. Sos P, Klirova M, Novak T, et al. Relationship of ketamine’s anti-
depressant and psychotomimetic effects in unipolar depression.
Neuro Endocrinol Lett. 2013;34:287–293.
238. Niciu MJ, Shovestul BJ, Jaso BA, et al. Features of dissociation
differentially predict antidepressant response to ketamine in
treatment-resistant depression. J Affect Disord. 2018;232:310–315.
239. Michaels TI, Purdon J, Collins A, et al. Inclusion of people of color in
psychedelic-assisted psychotherapy: a review of the literature. BMC
Psychiatry. 2018;18:245.
EXPERT REVIEW OF CLINICAL PHARMACOLOGY 15
... Emotional breakthrough is defined by engagement with negative emotional material and resolution-related cognitions. Consistent with the common mechanisms model (Greenway et al., 2020), we feel there is value in exploring to what degree the emotional breakthrough experience/construct converges with treatment mechanisms from other psychotherapeutic approaches. We consider three psychotherapeutic models here to connect the psychedelic literature to the broader clinical literature. ...
... Moderation analyses can be used to infer on a treatment's main causal actions, but these actions could also be shared across treatments, as is implied by the common factors model of psychotherapeutic action for example (Greenway et al., 2020). Moderation-based analyses are, however, vulnerable to validity threats related to correlational methods. ...
Article
Full-text available
The mechanisms by which Psilocybin Therapy (PT) improves depression remain an important object of study, with scientists actively exploring acute psychological experiences and neurobiological processes as candidates. In a phase 2, double-blind, randomized, active comparator controlled trial involving patients with moderate-to-severe major depressive disorder, we investigated whether acute psychological experiences could meaningfully account for the unique efficacy of PT versus Escitalopram Treatment over a core 6-week trial period. An exploratory-factor-analysis-derived single-factor of depression was used as the outcome. Among a comprehensive set of acute experiences related to psilocybin, so-called “mystical experience” and “ego dissolution” were unique in mediating the effect of treatment condition on depressive response with high specificity. Higher reported levels of mystical experience, emotional breakthrough, and intense responses to music-listening were furthermore associated with greater antidepressant response. These results provide qualified support for the causal mechanistic role of acute psychological experiences in the treatment of depression via PT.
... Additionally, both ketamine and meditation practice can give rise to mystical experiences (Millière et al., 2018), which can lead to significant perspective shifts (Krupitsky et al., 2002(Krupitsky et al., , 2007Morgan et al., 2017) that can help contribute to its therapeutic effects. Additionally, the psychedelic model (Greenway et al., 2020) proposes that ketamine experiences produce 'breakthrough realisations', generate enhanced awareness of the unconscious processes that underlie addiction, and improve feelings of internal and external connectedness, which can also be beneficial in addiction recovery (Garel et al., 2022: 11). Considering that the esketamine condition experienced significantly more mystical experiences and dissociative states, this could be an additional explanation for the observed transient reductions in alcohol cravings. ...
Article
Full-text available
Background Alcohol use disorder (AUD) is a major public health issue, posing harmful consequences for individuals and society. Recent advances in addiction research have highlighted the therapeutic potential of ketamine-assisted therapy for AUD. However, the exact mechanisms underlying its effectiveness remain unknown. Aims This double-blind, pilot study aimed to investigate esketamine combined with mindfulness-based intervention (MBI) to examine whether esketamine enhances engagement in MBI for individuals with alcohol misuse problems and whether enhanced engagement has any impact on alcohol-related outcomes. Methods In all, 28 individuals with alcohol problems were randomly assigned to receive sublingual esketamine hydrochloride (AWKN002: 115.1 mg) or vitamin C (placebo) in an oral thin film and took part in 2 weeks of daily MBI. Participants were assessed on various self-report measures, including mindfulness, engagement in MBI (physical and psychological), alcohol cravings and consumption. Results Esketamine enhanced psychological engagement with a daily MBI, compared to placebo, and led to transient decreases in alcohol cravings. Esketamine also resulted in significantly greater mystical experiences and dissociative states compared to placebo. Conclusions The findings suggest that esketamine may improve treatment outcomes when combined with mindfulness-based therapies through its ability to increase engagement with meditative practice.
... Agreeing with others who have pointed out that psychedelic therapy shares substantial "common factors" with other psychotherapies (Aday et al., 2023;Greenway et al., 2020;Nayak and Johnson, 2020), we assume that therapeutically effective psychedelic experiences are similar to those experiences which are purposefully induced in effective psychotherapy (Gründer et al., 2023;Jungaberle et al., 2008;Wolff et al., 2020Wolff et al., , 2022. Our contextual-experiential model ( Figure 1) suggests that such psychotherapy-like psychedelic experiences are most likely to occur in the context of formal psychotherapy and other social contexts that engender interpersonal trust, emotional openness, introspective attention, and self-inquiry. ...
Article
Full-text available
Abstract Background: Therapeutic and salutogenic effects of psychedelic drugs have been attributed to psychotherapeutic or psychotherapy-like processes that can unfold during the acute psychedelic experience and beyond. Currently, there are no psychometric instruments available to comprehensively assess psychotherapeutic processes (as conceptualized by empirical psychotherapy research) in the context of psychedelic experiences. Aims: We report the initial validation of the General Change Mechanisms Questionnaire (GCMQ), a self-report instrument designed to measure five empirically established general change mechanisms (GCMs) of psychotherapy—(1) resource activation, (2) therapeutic relationship, (3) problem actuation, (4) clarification, and (5) mastery—in the context of psychedelic experiences. Methods: An online survey in a sample of 1153 English-speaking and 714 German-speaking psychedelic users was conducted to evaluate simultaneously developed English- and German-language versions of the GCMQ. Results: The theory-based factor structure was confirmed. The five GCMQ scales showed good internal consistency. Evidence for convergent validity with external measures was obtained. Significant associations with different settings and with therapeutic, hedonic, and escapist use motives confirmed the hypothesized context dependence of GCM-related psychedelic experiences. Indicating potential therapeutic effects, the association between cumulative stressful life events and well-being was significantly moderated by resource activation, clarification, and mastery. Factor mixture modeling revealed five distinct profiles of GCM-related psychedelic experiences. Conclusion: Initial testing indicates that the GCMQ is a valid and reliable instrument that can be used in future clinical and nonclinical psychedelic research. The five identified profiles of GCM-related experiences may be relevant to clinical uses of psychedelics and psychedelic harm reduction.
... PAT is a therapeutic intervention that consists of administering a psychedelic substance in a carefully controlled environment with preparatory and follow-up psychological support centered around the acute experience (Schenberg, 2018). In this way, PAT represents a combined pharmaco-and psychotherapy approach that leverages both the acute psycho-emotional experience elicited by the drug, as well as its potentially experience-agnostic neurobiological effects, toward lasting positive change in behavior and well-being (Greenway et al., 2020). Evidence suggests that PAT can generate profound psychosocial changes, leading to marked symptom reductions across several mental disorders (Andersen et al., 2021) (Table 1). ...
Article
Full-text available
Functional seizures (FS), the most common subtype of functional neurological disorder (FND), cause serious neurological disability and significantly impact quality of life. Characterized by episodic disturbances of functioning that resemble epileptic seizures, FS coincide with multiple comorbidities and are treated poorly by existing approaches. Novel treatment approaches are sorely needed. Notably, mounting evidence supports the safety and efficacy of psychedelic-assisted therapy (PAT) for several psychiatric conditions, motivating investigations into whether this efficacy also extends to neurological disorders. Here, we synthesize past empirical findings and frameworks to construct a biopsychosocial mechanistic argument for the potential of PAT as a treatment for FS. In doing so, we highlight FS as a well-defined cohort to further understand the large-scale neural mechanisms underpinning PAT. Our synthesis is guided by a complexity science perspective which we contend can afford unique mechanistic insight into both FS and PAT, as well as help bridge these two domains. We also leverage this perspective to propose a novel analytic roadmap to identify markers of FS diagnostic specificity and treatment success. This endeavor continues the effort to bridge clinical neurology with psychedelic medicine and helps pave the way for a new field of psychedelic neurology.
... These experiences can have lasting impacts on life perspectives, beliefs, and feelings of well-being, and have been shown to treat depression, anxiety (including end-of-life anxiety), posttraumatic stress disorder, and alcohol use disorder, among others (Barrett & Griffiths, 2015;Bogenschutz et al., 2022;Goodwin et al., 2023;Griffiths et al., 2006Griffiths et al., , 2016Grob et al., 2011;Nayak et al., 2023). In treating these conditions with psychedelics, it is clear that set and setting have a profound impact on the experience (Feduccia et al., 2023;Greenway, Garel, Jerome, & Feduccia, 2020). Research into another psychedelic, ketamine, posits that resetting of the homeostatic or baseline neuronal signaling may be the biochemical explanation for its healing attributes Kim, Suzuki, Kavalali, & Monteggia, 2023). ...
Article
Full-text available
Psilocybe zapotecorum is a strongly blue-bruising psilocybin mushroom used by indigenous groups in southeastern Mexico and beyond. While this species has a rich history of ceremonial use, research into its chemistry and genetics has been limited. Herein, we report on mushroom morphology, cultivation parameters, chemical profile, and the full genome sequence of P. zapotecorum. First, we detail growth and cloning methods that are simple, and reproducible. In combination with high resolution microscopic analysis, the strain was identified by DNA barcoding, confirming the field identification. Full genome sequencing reveals the architecture of the psilocybin gene cluster in P. zapotecorum, and can serve as a reference genome for Psilocybe clade I. Characterization of the tryptamine profile revealed a psilocybin concentration of 17.9 ± 1.7 mg/g, with a range of 10.6–25.7 mg/g (n = 7), and similar tryptamines (psilocin, baeocystin, norbaeocystin, norpsilocin, aeruginascin, and 4-HO-tryptamine) in lesser concentrations for a combined tryptamine concentration of 22.5 ± 3.2 mg/g. These results show P. zapotecorum to be a potent and chemically variable Psilocybe mushroom. Chemical profiling, genetic analysis, and cultivation assist in demystifying these mushrooms. As clinical studies with psilocybin gain traction, understanding the diversity of Psilocybe expands the conversation beyond the molecule.
... Without psychotherapy there is no need to think about how it complicates the medical ethics of psychedelic therapy. Yet others believe that psychedelics differ from established psychiatric medicines in that they have no inherent therapeutic effect but serve as powerful catalysts of the therapeutic effects of psychotherapy [35,73,82]. If market approval is granted, psychiatrists might be free to use the drugs to assist whatever kind of psychotherapy they see fit. ...
Article
Full-text available
In the historical context of a crisis in biological psychiatry, psychedelic drugs paired with psychotherapy are globally re-emerging in research clinics as a potential transdiagnostic therapy for treating mood disorders, addictions, and other forms of psychological distress. The treatments are poised to soon shift from clinical trials to widespread service delivery in places like Australia, North America, and Europe, which has prompted ethical questions by social scientists and bioethicists. Taking a broader view, we argue that the ethics of psychedelic therapy concerns not simply how psychotherapies are different when paired with psychedelic drugs, but how psychedelic therapies shape and are shaped by different values, norms, and metaphysical commitments. Drawing from the published literature and interviews with seven psychedelic therapists working in clinical trials in the United States, Germany, Switzerland, and Australia, this article opens the black box of the treatments to consider the values and informal debates currently animating the therapies. Considering questions of patient autonomy, mechanisms of therapeutic action, and which therapies are best suited to pair with psychedelic substances, we examine the ethics of psychedelic therapy as an emergent form of life. To bring this form of life out in fuller relief, we conclude by comparing and contrasting it with ayahuasca use in Amazonian shamanism.
... Ketamine was administered with nonpharmacological treatment adjuncts inspired from the paradigm of psychedelic-assisted psychotherapy and included music, the use of blindfolds, and accompaniment by a trained physician who invited the patient to explore and make meaning of her ketamine experience. 14 Both treatments were very well tolerated with no significant increase in blood pressure, nausea, or headache. Immediately after the first treatment, the patient experienced significant improvements in mood, anhedonia, energy, concentration, and feelings of guilt: the improvement was not reflected by the modest improvement in the BDI-II where the symptoms of her post-COVID contributed heavily to the total score. ...
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
Full-text available
Background: Due to unmet clinical needs for efficient drugs with a rapid onset of antidepressant effects, we aimed to evaluate the efficacy of single-dose ketamine in different subgroups of patients with major depression and establish whether repeated ketamine administration could be a viable strategy to maintain treatment gains. Methods: Electronic databases (Medline via PubMed, Embase, Cochrane Library, Trip Database) were systematically searched until February 22, 2019, for published peer-reviewed randomized controlled trials (RCTs) concerning a single and repeated administration of ketamine in patients with major depression. All relevant RCTs were selected and critically appraised, and a meta-analysis of eligible studies was performed. Results: A total of 20 studies were included in the meta-analysis. The largest effect of ketamine vs. controls in reducing depressive symptoms was observed at 24 h (SMD = - 0.89; 95% CI - 1.24; - 0.53; p < 0.00001); however, a significant difference was shown for up to 7 days after a single dose. Significant differences compared with controls were observed for up to 7 days in treatment-resistant patients and when ketamine was added to ongoing antidepressant treatment, while there were no significant differences at 7 days when ketamine was used as monotherapy. In patients with major depression, initial antidepressant effects of ketamine were maintained during repeated dosing. At 2-3 weeks of repeated ketamine treatment, significant reduction of depression severity scores was observed: SMD = - 0.70; 95% CI - 1.15; - 0.25 or SMD = - 0.81; 95% CI - 1.41; - 0.20 (depending on the dosing regimen used); p ≤ 0.009 vs placebo. Conclusions: Our meta-analysis revealed rapid and robust antidepressant effects of single-dose ketamine in patients with treatment-resistant depression (TRD). By pooling data from RCTs, we showed for the first time that repeated ketamine administration is effective in sustaining initial antidepressant effects observed after single dosing.
Article
Full-text available
Lysergic acid diethylamide (LSD) is a classic psychedelic, 3,4-methylenedioxymethamphetamine (MDMA) is an empathogen, and d-amphetamine is a classic stimulant. All three substances are used recreationally. LSD and MDMA are being investigated as medications to assist psychotherapy, and d-amphetamine is used for the treatment of attention-deficit/hyperactivity disorder. All three substances induce distinct acute subjective effects. However, differences in acute responses to these prototypical psychoactive substances have not been characterized in a controlled study. We investigated the acute autonomic, subjective, and endocrine effects of single doses of LSD (0.1 mg), MDMA (125 mg), d-amphetamine (40 mg), and placebo in a randomized, double-blind, cross-over study in 28 healthy subjects. All of the substances produced comparable increases in hemodynamic effects, body temperature, and pupil size, indicating equivalent autonomic responses at the doses used. LSD and MDMA increased heart rate more than d-amphetamine, and d-amphetamine increased blood pressure more than LSD and MDMA. LSD induced significantly higher ratings on the 5 Dimensions of Altered States of Consciousness scale and Mystical Experience Questionnaire than MDMA and d-amphetamine. LSD also produced greater subjective drug effects, ego dissolution, introversion, emotional excitation, anxiety, and inactivity than MDMA and d-amphetamine. LSD also induced greater impairments in subjective ratings of concentration, sense of time, and speed of thinking compared with MDMA and d-amphetamine. MDMA produced greater ratings of good drug effects, liking, high, and ego dissolution compared with d-amphetamine. d-Amphetamine increased ratings of activity and concentration compared with LSD. MDMA but not LSD or d-amphetamine increased plasma concentrations of oxytocin. None of the substances altered plasma concentrations of brain-derived neurotrophic factor. These results indicate clearly distinct acute effects of LSD, MDMA, and d-amphetamine and may assist the dose-finding in substance-assisted psychotherapy research.
Article
Full-text available
Background: Depression is the single largest contributor to non-fatal health loss worldwide. Second-generation antidepressants are the first-line option for pharmacological management of depression. Optimising their use is crucial in reducing the burden of depression; however, debate about their dose dependency and their optimal target dose is ongoing. We have aimed to summarise the currently available best evidence to inform this clinical question. Methods: We did a systematic review and dose-response meta-analysis of double-blind, randomised controlled trials that examined fixed doses of five selective serotonin reuptake inhibitors (SSRIs; citalopram, escitalopram, fluoxetine, paroxetine, and sertraline), venlafaxine, or mirtazapine in the acute treatment of adults (aged 18 years or older) with major depression, identified from the Cochrane Central Register of Controlled Trials, CINAHL, Embase, LILACS, MEDLINE, PsycINFO, AMED, PSYNDEX, websites of drug licensing agencies and pharmaceutical companies, and trial registries. We imposed no language restrictions, and the search was updated until Jan 8, 2016. Doses of SSRIs were converted to fluoxetine equivalents. Trials of antidepressants for patients with depression and a serious concomitant physical illness were excluded. The main outcomes were efficacy (treatment response defined as 50% or greater reduction in depression severity), tolerability (dropouts due to adverse effects), and acceptability (dropouts for any reasons), all after a median of 8 weeks of treatment (range 4-12 weeks). We used a random-effects, dose-response meta-analysis model with flexible splines for SSRIs, venlafaxine, and mirtazapine. Findings: 28 554 records were identified through our search (24 524 published and 4030 unpublished records). 561 published and 121 unpublished full-text records were assessed for eligibility, and 77 studies were included (19 364 participants; mean age 42·5 years, SD 11·0; 7156 [60·9%] of 11 749 reported were women). For SSRIs (99 treatment groups), the dose-efficacy curve showed a gradual increase up to doses between 20 mg and 40 mg fluoxetine equivalents, and a flat to decreasing trend through the higher licensed doses up to 80 mg fluoxetine equivalents. Dropouts due to adverse effects increased steeply through the examined range. The relationship between the dose and dropouts for any reason indicated optimal acceptability for the SSRIs in the lower licensed range between 20 mg and 40 mg fluoxetine equivalents. Venlafaxine (16 treatment groups) had an initially increasing dose-efficacy relationship up to around 75-150 mg, followed by a more modest increase, whereas for mirtazapine (11 treatment groups) efficacy increased up to a dose of about 30 mg and then decreased. Both venlafaxine and mirtazapine showed optimal acceptability in the lower range of their licensed dose. These results were robust to several sensitivity analyses. Interpretation: For the most commonly used second-generation antidepressants, the lower range of the licensed dose achieves the optimal balance between efficacy, tolerability, and acceptability in the acute treatment of major depression. Funding: Japan Society for the Promotion of Science, Swiss National Science Foundation, and National Institute for Health Research.
Article
Full-text available
Introduction: Combat veterans are at high risk for the development of posttraumatic stress disorder (PTSD) and substance use disorders. Ketamine has been shown to be an effective treatment for numerous mental health disorders, although research on its efficacy in combat-related PTSD in veterans is very limited. Methods: The study population consisted of 30 US military veterans with combat-related PTSD. Participants underwent a standard induction series of six 1-hour ketamine infusions with the goal of obtaining a transpersonal dissociative experience. Participants were given a series of self-report questionnaires to assess for changes in symptoms of depression, PTSD, and substance use prior to the first and sixth infusions. Results: Symptoms of depression as measured by change in score on the Patient Health Questionnaire decreased significantly from an average of 18.9 to 9.5 (P < .001). Similarly, symptoms of PTSD as measured by change in score on the PSTD Checklist for DSM-5 dropped significantly from an average of 56.2 to 31.3 (P < .001). Self-reported levels of substance use did not significantly decrease during the study period, although the level of use trended down. Conclusions: This observational study suggests that high-dose ketamine infusion therapy, which induced a transpersonal dissociative experience, could be a valuable tool in the treatment of combat-related PTSD. Further study is needed to better elucidate ketamine's mechanism of action with regards to the treatment of PTSD.
Book
Sacred Knowledge is the first well-documented, sophisticated account of the effect of psychedelics on biological processes, human consciousness, and revelatory religious experiences. Based on nearly three decades of legal research with volunteers, William A. Richards argues that, if used responsibly and legally, psychedelics have the potential to assuage suffering and constructively affect the quality of human life. Richards’s analysis contributes to social and political debates over the responsible integration of psychedelic substances into modern society. His book serves as an invaluable resource for readers who, whether spontaneously or with the facilitation of psychedelics, have encountered meaningful, inspiring, or even disturbing states of consciousness and seek clarity about their experiences. Testing the limits of language and conceptual frameworks, Richards makes the most of experiential phenomena that stretch our conception of reality, advancing new frontiers in the study of belief, spiritual awakening, psychiatric treatment, and social well-being. His findings enrich humanities and scientific scholarship, expanding work in philosophy, anthropology, theology, and religious studies and bringing depth to research in mental health, psychotherapy, and psychopharmacology.
Article
Objectives: To assess the evidence and discuss the risks and clinical relevance of ketamine for the treatment of various disease states impacting the adult critically ill population. Data sources: A literature review was performed using PubMed evaluating primary literature published until August 2018. Study selection: Case reports, observational studies (cohort, case-control), and randomized controlled trials involving patients 18 years and older in a nonperioperative setting using either IV or intramuscular ketamine were included for analysis. Uses of ketamine discussed focused on critically ill patients in the ICU and emergency department settings. Data extraction: Included studies were evaluated for dosing, outcomes, and adverse effects of ketamine. For each study, the design, population, intervention, investigated outcomes, and results were assessed. Data synthesis: The evidence was organized according to use of ketamine, which included pain, sedation, status asthmaticus, alcohol withdrawal syndrome, status epilepticus, and acute behavioral psychologic disturbances. Evaluation of the evidence was based on the included primary literature along with any related guideline recommendations. Conclusions: Ketamine has suggested potential benefit in several disease states impacting critically ill patients including pain, alcohol withdrawal syndrome, status epilepticus, and acute agitation. Further supporting evidence is needed to validate its use in the setting of critical illness.
Article
Objective: To evaluate long-term safety and efficacy of esketamine nasal spray plus a new oral antidepressant (OAD) in patients with treatment-resistant depression (TRD). Methods: This phase 3, open-label, multicenter, long-term (up to 1 year) study was conducted between October 2015 and October 2017. Patients (≥ 18 years) with TRD (DSM-5 diagnosis of major depressive disorder and nonresponse to ≥ 2 OAD treatments) were enrolled directly or transferred from a short-term study (patients aged ≥ 65 years). Esketamine nasal spray (28-mg, 56-mg, or 84-mg) plus new OAD was administered twice a week in a 4-week induction (IND) phase and weekly or every-other-week for patients who were responders and entered a 48-week optimization/maintenance (OP/MAINT) phase. Results: Of 802 enrolled patients, 86.2% were direct-entry and 13.8% were transferred-entry; 580 (74.5%) of 779 patients who entered the IND phase completed the phase, and 150 (24.9%) of 603 who entered the OP/MAINT phase completed the phase. Common treatment-emergent adverse events (TEAEs) were dizziness (32.9%), dissociation (27.6%), nausea (25.1%), and headache (24.9%). Seventy-six patients (9.5%) discontinued esketamine due to TEAEs. Fifty-five patients (6.9%) experienced serious TEAEs. Most TEAEs occurred on dosing days, were mild or moderate in severity, and resolved on the same day. Two deaths were reported; neither was considered related to esketamine. Cognitive performance generally either improved or remained stable postbaseline. There was no case of interstitial cystitis or respiratory depression. Treatment-emergent dissociative symptoms were transient and generally resolved within 1.5 hours postdose. Montgomery-Åsberg Depression Rating Scale total score decreased during the IND phase, and this reduction persisted during the OP/MAINT phase (mean [SD] change from baseline of respective phase to endpoint: IND, -16.4 [8.76]; OP/MAINT, 0.3 [8.12]). Conclusions: Long-term esketamine nasal spray plus new OAD therapy had a manageable safety profile, and improvements in depression appeared to be sustained in patients with TRD. Trial registration: ClinicalTrials.gov identifier: NCT02497287.
Article
Background: Most guidelines for major depressive disorder recommend initial treatment with either a second-generation antidepressant (SGA) or cognitive behavioral therapy (CBT). Although most trials suggest that these treatments have similar efficacy, their health economic implications are uncertain. Objective: To quantify the cost-effectiveness of CBT versus SGA for initial treatment of depression. Design: Decision analytic model. Data sources: Relative effectiveness data from a meta-analysis of randomized controlled trials; additional clinical and economic data from other publications. Target population: Adults with newly diagnosed major depressive disorder in the United States. Time horizon: 1 to 5 years. Perspectives: Health care sector and societal. Intervention: Initial treatment with either an SGA or group and individual CBT. Outcome measures: Costs in 2014 U.S. dollars, quality-adjusted life-years (QALYs), and incremental cost-effectiveness ratios. Results of base-case analysis: In model projections, CBT produced higher QALYs (3 days more at 1 year and 20 days more at 5 years) with higher costs at 1 year (health care sector, $900; societal, $1500) but lower costs at 5 years (health care sector, -$1800; societal, -$2500). Results of sensitivity analysis: In probabilistic sensitivity analyses, SGA had a 64% to 77% likelihood of having an incremental cost-effectiveness ratio of $100 000 or less per QALY at 1 year; CBT had a 73% to 77% likelihood at 5 years. Uncertainty in the relative risk for relapse of depression contributed the most to overall uncertainty in the optimal treatment. Limitation: Long-term trials comparing CBT and SGA are lacking. Conclusion: Neither SGAs nor CBT provides consistently superior cost-effectiveness relative to the other. Given many patients' preference for psychotherapy over pharmacotherapy, increasing patient access to CBT may be warranted. Primary funding source: Department of Veterans Affairs, National Institute of Mental Health.