U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Cover of Evidence Brief: Psychedelic Medications for Mental Health and Substance Use Disorders

Evidence Brief: Psychedelic Medications for Mental Health and Substance Use Disorders

, MD, MPP, Conceptualization, Methodology, Investigation, Formal analysis, Writing – original draft, Writing – review & editing, Supervision, , MPH, Methodology, Investigation, Writing – original draft, Writing – review & editing, Project administration, , NP, MPH, Investigation, Writing – original draft, Writing – review & editing, , BA, Project administration, Investigation, and , PhD, Conceptualization, Methodology, Formal analysis, Writing – original draft, Writing – review & editing, Project administration.

Author Information and Affiliations
Washington (DC): Department of Veterans Affairs (US); .

The ESP Coordinating Center (ESP CC) is responding to a request from the VA Office of Research and Development (ORD) and VHA Office of Mental Health and Suicide Prevention (OMHSP) for an Evidence Brief on the use of psychedelic substances for treating mental health and substance use disorders. Findings from this Evidence Brief will be used to inform VA research activities and clinical care in the areas of mental health and substance use disorder treatment.

PREFACE

The VA Evidence Synthesis Program (ESP) was established in 2007 to provide timely and accurate syntheses of targeted health care topics of importance to clinicians, managers, and policymakers as they work to improve the health and health care of Veterans. These reports help:

  • Develop clinical policies informed by evidence;
  • Implement effective services to improve patient outcomes and to support VA clinical practice guidelines and performance measures; and
  • Set the direction for future research to address gaps in clinical knowledge.

The program comprises four ESP Centers across the US and a Coordinating Center located in Portland, Oregon. Center Directors are VA clinicians and recognized leaders in the field of evidence synthesis with close ties to the AHRQ Evidence-based Practice Center Program. The Coordinating Center was created to manage program operations, ensure methodological consistency and quality of products, interface with stakeholders, and address urgent evidence needs. To ensure responsiveness to the needs of decision-makers, the program is governed by a Steering Committee composed of health system leadership and researchers. The program solicits nominations for review topics several times a year via the program website.

The present report was developed in response to a request from the VA Office of Research and Development (ORD) and VHA Office of Mental Health and Suicide Prevention (OMHSP). The scope was further developed with input from Operational Partners (below) and the ESP Coordinating Center review team.

ACKNOWLEDGMENTS

The authors are grateful to Kathryn Vela, MLIS for literature searching, Sarah Young, MPH for study selection assistance, and the following individuals for their contributions to this project:

Operational Partners

Operational partners are system-level stakeholders who help ensure relevance of the review topic to the VA, contribute to the development of and approve final project scope and timeframe for completion, provide feedback on the draft report, and provide consultation on strategies for dissemination of the report to the field and relevant groups.

  • Ilse Wiechers, MD
    Deputy Executive Director
    Office of Mental Health and Suicide Prevention

Peer Reviewers

The Coordinating Center sought input from external peer reviewers to review the draft report and provide feedback on the objectives, scope, methods used, perception of bias, and omitted evidence (see Appendix D in Supplemental Materials for disposition of comments). Peer reviewers must disclose any relevant financial or non-financial conflicts of interest. Because of their unique clinical or content expertise, individuals with potential conflicts may be retained.

The Coordinating Center works to balance, manage, or mitigate any potential nonfinancial conflicts of interest identified.

EXECUTIVE SUMMARY

Background

The Evidence Synthesis Program Coordinating Center is responding to a request from the VA Office of Research and Development (ORD) and VHA Office of Mental Health and Suicide Prevention (OMHSP) for an Evidence Brief on the use of psychedelics for treating mental health and substance use disorders. Findings from this Evidence Brief will be used to inform VA research and clinical care.

Methods

To identify studies, we searched MEDLINE, Cochrane Database of Systematic Reviews, Cochrane Central Register of Controlled Trials, and other sources up to April 2022. We used prespecified criteria for study selection, data abstraction, and rating internal validity and strength of the evidence. See the Methods section and our PROSPERO protocol for full details of our methodology.

Key Findings

  • In a clinical research setting, use of MDMA with intensive psychotherapy led by 2 mental health professionals may improve posttraumatic stress disorder (PTSD symptom severity and result in remission for some participants in the short term (1 month to 18 weeks) (low strength of evidence [SOE]). Findings are based on several small trials of adults with moderate to severe symptoms at baseline who did not improve with prior medication or therapy trials. Most studies did not include Veterans.
  • Psilocybin-assisted psychotherapy may reduce depression severity and lead to sustained remission for some participants at 12 months compared to wait list controls, but these benefits were not observed when psilocybin-assisted psychotherapy was compared to intensive psychotherapy and daily escitalopram (low SOE).
  • Psilocybin-assisted psychotherapy may reduce heavy drinking days and average daily alcohol consumption among adults with alcohol use disorder (low SOE).
  • Ayahuasca use may be associated with a short-term reduction in depression symptoms and suicidality (low SOE). Whether ayahuasca is effective for substance use disorder treatment is unclear.
  • Ibogaine may not reduce opioid withdrawal symptoms or short-term opioid use (low SOE) and has been associated with serious adverse events including prolonged QTc, cardiac arrhythmia, and death.
  • Studies conducted in the US have predominantly included non-Veteran, young, non-Hispanic white populations with access to a clinical research center or treatment program. Whether benefits can be replicated in more diverse populations and treatment settings including within VHA is unclear.
  • Critical gaps to address in future research include whether treatment effects vary according to patient characteristics and disease severity and whether benefits are durable over time. The feasibility of implementing intensive psychotherapy protocols in real-world settings is another critical gap, as is determining how psychedelic treatments could fit into current standard of care and usual care pathways.

CLINICAL BOTTOM LINE

MDMA-assisted psychotherapy for PTSD has been the most extensively studied therapeutic intervention involving psychedelics. Results from several small RCTs demonstrate that this treatment may improve PTSD symptom severity to a clinically meaningful degree and lead to disease remission for some individuals in the short term. Similarly, psilocybin-assisted psychotherapy for depression shows some promise. However, study interventions included intensive psychotherapy protocols that may be challenging to implement in everyday practice. Moreover, US studies primarily enrolled young non-Hispanic white individuals and few studies included Veterans. Results may not be generalizable to more diverse populations and settings including within the Veterans Health Administration (VHA). Evidence on psychedelics for treatment of mental health and substance use disorders is therefore very preliminary. Several critical gaps need to be addressed by future research.

OVERVIEW

The term psychedelics refers to a group of substances that temporarily alter perception and mood and may impact numerous cognitive processes. The term is used to describe naturally derived substances with these properties, such as psilocybin, as well as synthetic substances like lysergic acid diethylamide (LSD) and 3,4-methylenedioxymethamphetamine (MDMA). Naturally derived psychedelics have been used for millennia, and starting in the 1940s through the 1960s, natural and synthetic psychedelics were used in clinical research as potential treatments for a range of medical conditions. However, study of psychedelics largely paused in the US in 1970 when most were codified as Schedule I drugs with no accepted medical use. In recent years, a growing number of researchers have been revisiting the potential therapeutic benefits of psychedelics and have sought out legal and regulatory pathways to study them.

Psychedelics and psychedelic-assisted psychotherapy have been proposed as novel treatments for adults with mental health and substance use disorders based on the supposition that intentional use of psychedelics over a limited period can disrupt problematic thought patterns, facilitate psychotherapy, and potentially lead to behavior change. New treatment options are needed given the limitations of existing therapies. The aim of this review is to inform VHA program planning by synthesizing available evidence on the benefits and harms of psychedelics as primary or adjunct treatment for mental health and substance use disorders.

From 2,532 potentially relevant articles, 15 RCTs and 23 observational studies (in 44 publications) met eligibility criteria. Studies enrolled adults with PTSD, depression, obsessive compulsive disorder (OCD), social anxiety, opioid use disorder (OUD), alcohol use disorder, tobacco use, or mixed substance use disorders and mood symptoms. Interventions involved treatment with MDMA, psilocybin, ayahuasca, 5-methoxy-N,N-dimethyltryptamine, and ibogaine with or without psychotherapy or other co-interventions.

The most extensively studied intervention involving psychedelics is MDMA-assisted psychotherapy for PTSD. We identified 9 small RCTs (including 1 study that pooled results from 6 smaller trials) and 3 observational studies of MDMA-assisted psychotherapy among adults with moderate or severe PTSD who had not improved with at least 1 past trial of medication or psychotherapy. All trials were conducted by researchers affiliated with the Multidisciplinary Association for Psychedelic Studies (MAPS) or were sponsored by this organization and thus had several features in common. Intervention group participants typically received MDMA 80–125mg with or without a 40–62.5mg supplemental dose. Comparator group participants received an 0mg placebo or a low dose of MDMA (low-dose placebo) intended to maintain participant blinding by producing some sensations of having ingested a psychedelic. Intervention and control group participants underwent the same intensive psychotherapy protocol, typically consisting of several preparatory sessions and follow-up sessions in addition to day-long MDMA or placebo dosing sessions. Psychotherapy sessions were all co-led by 2 mental health professionals using a standardized therapy manual. If participants were receiving treatment for PTSD prior to the study start, they were required to stop it to avoid confounding and potential drug interactions with MDMA. Studies reported outcomes in terms of changes in mean total severity scores on the Clinician-Administered PTSD Scale (CAPS). Some studies reported whether participants experienced a clinically meaningful improvement in total CAPS score (defined as at least 30% improvement) and PTSD remission.

MDMA paired with intensive psychotherapy may reduce PTSD symptom severity and lead to clinically meaningful improvement and remission for some. In trials reporting these outcomes, 50% to 100% of participants experienced clinically meaningful improvements on CAPS total severity scores and remission rates ranged from 33% to 88% when assessed at 1 month to 18 weeks. Additionally, results of a meta-analysis of 7 trials indicate that participants receiving MDMA experienced significantly greater improvements in mean symptom severity compared with control group participants (pooled standardized mean difference in change scores = −0.91, 95% CI [−1.33, −0.50]). Symptoms among control group participants also improved in most studies regardless of the type of placebo used, although to a lesser degree than MDMA group participants. While the relative importance of different intervention components (MDMA, psychotherapy, and other factors such as participation effects) is unclear, a comparison of findings across trials using different forms of placebo suggests that benefits are at least partially attributable to the effects of MDMA. Although results are fairly consistent and direct, trial sample sizes are small and at risk of prognostic imbalance (a failure of randomization to balance groups on participants’ responsiveness to treatment). Our confidence in these findings is low.

Other interventions involving psychedelics have been less extensively studied. Psilocybin-assisted psychotherapy for depression may reduce symptom severity and lead to sustained remission at 12 months for a proportion of recipients compared to wait list controls, but these same benefits were not observed (in a separate trial) when psilocybin-assisted psychotherapy was compared with the same psychotherapy protocol and daily escitalopram. We have low confidence in these findings, which are based on 1 small trial for each comparator. Ayahuasca use for depression may be associated with a reduction in short-term symptoms and suicidality. However, our confidence in these findings is also low due to a smaller number of trials overall with small sample sizes and some inconsistency.

Among treatments evaluated for substance use disorders, we have low confidence that psilocybin-assisted psychotherapy for alcohol use disorder may reduce alcohol consumption (heavy drinking days and the number of drinks per day) based on results of 1 small trial. Psilocybin has also been used as an adjunct to psychotherapy to reduce tobacco use, but results are limited to 1 small observational study and are insufficient to draw conclusions. Ibogaine, used most often to treat opioid withdrawal, did not improve withdrawal symptoms in 1 small trial despite observational data showing benefits. Unlike other psychedelics studied, ibogaine has been associated with serious adverse events, including death and cardiac events.

Psychedelic treatments for mental health and substance use disorders are still in an early phase of development and many gaps in the evidence exist, including whether treatment effects vary by patient characteristics or disease severity and whether observed benefits are maintained over time. Other critical gaps in the evidence include whether psychedelic treatment protocols are reproducible and scalable in real-world settings and whether the benefits of discontinuing mental health medications to undergo psychedelic treatments outweigh the risks. In addition to addressing these gaps, areas for future research include study of Veterans with combat-related trauma and study of more diverse populations, particularly with respect to race/ethnicity, socioeconomic status, and rurality.

ES Table. Summary of Evidence.

ES Table

Summary of Evidence.

INTRODUCTION

PURPOSE

The ESP Coordinating Center (ESP CC) is responding to a request from the VA Office of Research and Development (ORD) and VHA Office of Mental Health and Suicide Prevention (OMHSP) for an Evidence Brief on the use of psychedelic substances for treating mental health and substance use disorders. Findings from this Evidence Brief will be used to inform VA research activities and clinical care in the areas of mental health and substance use disorder treatment.

BACKGROUND

The term psychedelics refers to a group of substances that temporarily alter perception and mood and may impact numerous cognitive processes.41,42 The term is used to describe naturally derived substances with these properties, such as psilocybin, as well as synthetic substances like lysergic acid diethylamide (LSD) and 3,4-methylenedioxymethamphetamine (MDMA).43 Over time, various other terms have been used to describe and distinguish psychedelic substances including hallucinogens, psychotomimetics, psycholytics, and in the case of MDMA, entactogens.44,45 For brevity, the term psychedelics is used in this report. Table 1 presents the derivation and proposed mechanism of selected psychedelic substances.

Naturally derived psychedelics have been used for millennia, often in the context of religious or spiritual practices. From the 1940s through the 1960s, psychedelics were studied in clinical research as potential treatments for a range of medical conditions including mood and substance use disorders. Despite some promising early findings,46 research on psychedelic substances largely ceased in the US with passage of the 1970 Controlled Substances Act,47 which codified psychedelics as drugs with no accepted medical use.48 In recent years, a growing number of researchers have been revisiting the potential therapeutic benefits of psychedelics and have sought out legal and regulatory pathways to study them. Alongside renewed research interest, increasing media attention has raised public awareness of potential therapeutic uses of psychedelics. Although psychedelics are still categorized as Schedule I controlled substances, MDMA and psilocybin have received “breakthrough therapy” designation from the Food and Drug Administration (FDA) to be studied in adults with post-traumatic stress disorder (PTSD) and treatment-resistant depression, respectively.49,50 In 2020, Oregon was the first state to create a regulatory framework for therapeutic psilocybin use,51 with plans to allow psilocybin service centers with licensed facilitators to begin operating in 2023.

Most psychedelics used in research act as agonists or partial agonists at serotonin 5-hydroxytryptamine 2A receptors in the brain. This group includes LSD, psilocybin, and ayahuasca and are often referred to as classical psychedelics. In clinical contexts, classical psychedelics have most often been used to treat depression and substance use disorders. For these indications, it is thought that psychedelic use can disrupt problematic thought patterns and potentially lead to behavior change.30 So-called non-classical psychedelics include MDMA and ibogaine. MDMA has most often been used to treat PSTD, as it is thought to reduce the anxiety and fear that can accompany memories of past trauma and thereby facilitate psychotherapy.52

MDMA works by complex mechanisms to promote release of serotonin, (nor)epinephrine, and dopamine and may stimulate signaling of oxytocin and other hormones. Ibogaine, used predominantly to treat substance use disorders, has a poorly understood mechanism of action but may work through a mix of neurotransmitters.23 In general, psychedelics are intended to be used for therapeutic purposes once or a limited number of times and are not meant to be used as long-term treatment of mental health or substance use disorders.

Table 1. Characteristics of Selected Psychedelic Substances.

Table 1

Characteristics of Selected Psychedelic Substances.

The effectiveness of psychedelics is thought to in part depend on the treatment intentions of the patient and the physical environment in which psychedelics are administered (“set” and “setting,” respectively).53 In a clinical research setting, psychedelics are typically administered over a day-long dosing session in a monitored and controlled environment designed to promote relaxation and minimize disruption (eg, in a comfortable setting with the participant wearing eyeshades and listening to pre-selected music through headphones). In many cases, a specific number of structured psychotherapy sessions are delivered before and after psychedelic dosing. For example, treatment may involve 1–2 psychotherapy sessions before an extended 8–10-hour session in which the therapist or other health care personnel administers the psychedelic substance and monitors the participant for safety. The psychedelic session may then by followed by additional psychotherapy sessions to debrief and analyze the participant’s psychedelic experience, often referred to as integration sessions. The cycle of psychedelic administration followed by post-psychedelic integration sessions may be repeated 2–3 times. Contemporary studies of psychedelic substances have used different treatment protocols for a range of disorders and have employed study designs with differing research aims (eg, safety, feasibility, efficacy, effectiveness, comparative effectiveness).

Given the prevalence of mental health and substance use disorders within the Veteran population, any novel or emerging treatments with the potential to improve clinical outcomes are of great interest to VA patients, researchers, and other stakeholders.54 Although VA services include comprehensive care for mental health and substance use disorders in a range of settings including residential rehabilitation,55 a proportion of Veterans may not experience improvement with existing treatment options.56 Therefore, psychedelics and psychedelic-assisted psychotherapy represent a potential new option for those with mental health or substance use disorders who have not responded to available treatments. The aim of this review is to inform VHA program planning by synthesizing available evidence on the benefits and harms of psychedelics as primary or adjunct treatment for mental health and substance use disorders.

METHODS

PROTOCOL

A preregistered protocol for this review can be found on the PROSPERO international prospective register of systematic reviews (http://www.crd.york.ac.uk/PROSPERO/; registration number CRD42022330065).

KEY QUESTIONS

The following key questions (KQs) were the focus of this review:

KQ1.

What are the benefits and harms of psychedelic substances as primary or adjunct treatment for mental health and substance use disorders?

KQ1a.

Do benefits or harms of psychedelic substances vary based on patient characteristics (eg, race/ethnicity, gender identity, age, comorbid mental health or substance use disorders, index trauma type), disorder type, or disorder severity?

ANALYTIC FRAMEWORK

The analytic framework shown in Figure 1 provides a conceptual overview of this review. The population of interest was adults with mental health and/or substance use disorders. Eligible outcomes included mental health and/or substance use disorder symptoms (Key Questions 1 and 1a) and treatment harms (Key Question 1). Whether benefits and/or risks of the intervention differ by patient characteristics (eg, patient demographics, comorbidities, disease severity) or treatment protocol (eg, number of sessions, dosage) was also of interest (Key Question 1a).

Figure 1. Analytic Framework.

Figure 1

Analytic Framework. Abbreviations. LSD=lysergic acid; MDMA=3,4-methylenedioxymethamphetamine; PSTD=posttraumatic stress disorder.

ELIGIBILITY CRITERIA

The ESP included studies that met the following criteria:

PopulationAdults with mental health and/or substance use disorders (eg, depression, PTSD, opioid use disorder). Studies among adults with serious medical illness (eg, terminal cancer, end-stage renal disease) with mental health symptoms or substance use were excluded.
InterventionPsychedelic substances (eg, ayahuasca/DMT, LSD, MDMA, psilocybin; excluding cannabinoids and ketamine) used as a primary treatment or as an adjunct to psychotherapy or another treatment (ie, psychedelic-assisted therapy)
ComparatorAny (eg, placebo, treatment as usual)
Outcomes
  • Disorder symptoms
  • Quality of life/functioning
  • Harms (eg, treatment-emergent adverse events)
Study DesignSystematic reviews, randomized controlled trials (RCTs), and concurrently controlled cohort studies. We considered longitudinal studies without comparison groups (ie, pre-post studies) and case series (ie, N > 1) only to address gaps in evidence from studies with control groups. We excluded case reports (ie, N = 1).

DATA SOURCES AND SEARCHES

To identify articles relevant to the key questions, a research librarian searched Ovid MEDLINE, Ovid PsycINFO, and ClinicalTrials.gov, as well as AHRQ, Cochrane Database of Systematic Reviews, and HSR&D through April 2022 using terms for hallucinogen, psychedelic, and mental disorders (see Appendix A in Supplemental Materials for complete search strategies). Additional citations were identified from hand-searching reference lists. We limited the search to published and indexed articles involving human subjects available in the English language. Study selection was based on the eligibility criteria described above. We excluded studies of cannabis and ketamine (a dissociative anesthetic) because these substances have distinct mechanisms of action compared to psychedelics and have been reviewed extensively elsewhere.5759 To ensure that study findings would be most applicable to contemporary diagnostic and management practices, we limited inclusion to studies published in 1994 or after to coincide with the publication of Diagnostic and Statistical Manual of Mental Disorders-4.60

Titles, abstracts, and full-text articles were independently reviewed by 2 investigators. All disagreements were resolved by consensus or discussion with a third investigator.

DATA ABSTRACTION AND ASSESSMENT

Effect information and population, intervention, and comparator characteristics were abstracted from all included studies. The internal validity (risk of bias) of each included study was rated using the Cochrane risk of bias tools.61,62 All data abstraction and internal validity ratings were first completed by 1 investigator and then checked by another; disagreements were resolved by consensus or discussion with a third investigator.

We graded the strength of the evidence for each outcome based on the AHRQ Methods Guide for Comparative Effectiveness Reviews.63 This approach provides a rating of confidence in reported findings based on study methodology (design, quality, and risk of bias), consistency (whether effects are in the same direction and have a consistent magnitude), directness (whether assessed outcomes are clinically important to patients and providers), and precision of findings (eg, confidence intervals). For this review, we applied the following general algorithm: high strength evidence consisted of multiple, large trials with low risk of bias, consistent and precise findings, and clinically relevant outcomes; moderate strength evidence consisted of multiple trials with low to unclear risk of bias, consistent and precise findings, and clinically relevant outcomes; low strength evidence consisted of a single trial or multiple small trials and/or multiple observational studies, with unclear to high risk of bias, consistent findings, and clinically relevant outcomes; and insufficient evidence consisted of a single or multiple small observational studies with unclear or high risk of bias, or no available studies.

SYNTHESIS

We grouped studies by disorder and psychedelic type and described mean changes in symptoms, when possible including whether the psychedelic intervention led to a clinically meaningful improvement in symptom severity (as defined by the study) or disease remission. We focused synthesis on disorder-specific outcomes (eg, PTSD symptom severity) and did not describe outcomes related to the psychedelic experience itself or measures of mood or perception not directly relevant to the disorder being treated.

Trials of MDMA-assisted psychotherapy for PTSD used the Clinician-Administered PTSD Scale (CAPS) total severity score (CAPS-4 or CAPS-5, rated 0-136 or 0-80, respectively) to report mean changes in PTSD symptom severity from baseline for intervention and comparator groups. For these trials, we synthesized bias-adjusted standardized mean differences (SMDs; Hedges’ g) in CAPS change scores between groups using random-effects meta-analysis. Precision of study-level and pooled mean differences is reported using 95% confidence intervals (CIs), which were also used to evaluate statistical significance at a significance level of .05. Heterogeneity in true study effects was estimated using restricted maximum-likelihood estimation and is presented as 95% prediction intervals (PIs). A PI encompassing values that substantively differ in direction and/or magnitude from the pooled MD indicates the presence of considerable heterogeneity.64,65 The Hartung-Knapp-Sidik-Jonkman method was used to better account for uncertainty in estimating the amount of heterogeneity.66 Meta-analyses were conducted using the metafor67 package for R (R Foundation for Statistical Computing, Vienna, Austria).

RESULTS

LITERATURE FLOW

The literature flow diagram (Figure 2) summarizes the results of the study selection process (a full list of excluded studies is available in Appendix B in Supplemental Materials).

Figure 2. Literature Flowchart.

Figure 2

Literature Flowchart. Notes. 38 studies in 44 publications. Abbreviations. CDSR=Cochrane Database of Systematic Reviews; SR=systematic review.

LITERATURE OVERVIEW

Our search identified 2,532 potentially relevant articles. We included 38 studies (in 44 publications),136,3840,6871 which are summarized in Table 2. Additional study details are provided in each section below and in Appendix C in the Supplemental Materials. Fifteen studies were RCTs, 21 were pre-post studies, and 2 were cohorts compared to healthy, non-treated controls. Participants included adults with PTSD, depression, obsessive compulsive disorder (OCD), social anxiety, opioid use disorder (OUD), alcohol use disorder, tobacco use, and mixed substance use disorders and mood symptoms. In most cases, studies described participants as having moderate to severe disease who had not improved with past treatment trials (more detail is provided in each section below). Adults with a personal or family history of a psychotic disorder or bipolar I disorder were generally excluded from study participation, as were those with major medical comorbidities and pregnant individuals. Studies inconsistently reported whether participants had previously used psychedelics; in some cases, recent or past use was a reason for exclusion. Sample sizes ranged from 3–191; the largest study37 (N = 191) was of adults seeking treatment for opioid or cocaine dependence with ibogaine.

Most studies (61%) were conducted exclusively outside the US, with Brazil and the United Kingdom being the most common locations. However, most studies did not report whether participants were local to the region or had travelled there for treatment purposes. Given the legal restrictions on psychedelic use in many locations, it is likely that some studies included participants from multiple countries. Participant mean age ranged from 25 to 51 in studies reporting this information. Among studies conducted completely or partially in the US, most participants were non-Hispanic white. Only 1 study exclusively enrolled US Veterans: a pre-post study of 51 Special Operations Forces Veterans who were treated for a range of mood symptoms with ibogaine and 5-MeO-DMT during a 3-day residential program in Mexico.40 Three studies of MDMA-assisted psychotherapy for PTSD included Veterans (range: 14–85% of total sample).1,2,11

MDMA was the only psychedelic used in studies of adults with PTSD. Psilocybin and ayahuasca were used most often in studies of adults with depression or substance use. Ibogaine was used in studies of adults with OUD and mixed substance use disorders. No studies of LSD published after 1994 met our inclusion criteria. Most studies of MDMA and psilocybin included intensive psychotherapy as a co-intervention. In contrast, most studies of ayahuasca and ibogaine did not include a therapy co-intervention. Comparators in RCTs included 0mg placebo, low-dose placebo (a lower dose of the same psychedelic used in the intervention group), or another non-psychedelic medication (eg, escitalopram, diphenhydramine). Most studies of PTSD required participants to discontinue usual mental health treatments except for sedative hypnotics or anxiolytics used between MDMA sessions (2 trials did not clearly report whether discontinuation was required). In most studies of depression, participants were not on other medical treatments for depression or were required to discontinue their usual treatments (usually antidepressant medications including selective serotonin reuptake inhibitors) for at least 2 weeks prior to the study period. Detail on whether studies did or did not require participants to discontinue their usual treatment are in Appendix C of the Supplemental Materials.

Most trials of MDMA for PTSD and psilocybin for depression were designed to evaluate treatment efficacy, while studies of ayahuasca and ibogaine were more often designed to evaluate safety and feasibility. Common methodological limitations of RCTs were small sample sizes, unclear intervention adherence, and unclear handling and/or extent of missing data. Observational studies, which mostly used a pre-post design, were limited by the potential for selection bias and lack of control groups.

We identified 70 in-progress studies (see Appendix E in Supplemental Materials) examining psychedelic use in populations with mood disorders, anxiety disorders, and substance use disorders.

Table 2. Characteristics of Included Studies by Treatment Indication.

Table 2

Characteristics of Included Studies by Treatment Indication.

POSTTRAUMATIC STRESS DISORDER

In a clinical research setting, MDMA-assisted psychotherapy may reduce PTSD symptoms, resulting in a clinically meaningful improvement (defined as a reduction in CAPS score by at least 30%) and/or disease remission in the short term (1 month to 18 weeks). We have low confidence in these findings based on several small trials with consistent and direct results with some imprecision (wide confidence intervals). Few serious adverse events were reported across studies, and only 1 event was ultimately attributed to MDMA.

We identified 7 unique RCTs17 of MDMA-assisted psychotherapy compared to psychotherapy with a 0mg placebo or low-dose placebo, and 1 RCT68 that pooled results from 6 of these trials. Studies used the CAPS total severity score (CAPS-4 or CAPS-5, rated 0–136 or 0–80, respectively) to report mean change from baseline in PTSD symptom severity for intervention and comparator groups. CAPS is conducted by a semi-structured interview that accesses an individual’s trauma history as defined by the Diagnostic and Statistical Manual of Mental Disorders. All studies included participants with moderate (total severity score 40–59 on CAPS-472 and 23–34 on CAPS-51) or severe (≥60 on CAPS-4 and ≥35 on CAPS-5) symptoms at baseline (Table 3). Most studies included participants who had been experiencing PTSD symptoms for years and had previously undergone at least 1 medication or psychotherapy trial.

Participants assigned to intervention groups received MDMA 80–125mg with or without a 40–62.5mg supplemental dose. Three studies used a 0mg placebo, and 4 studies used a low-dose placebo (MDMA 25–40mg with or without a 15mg supplemental dose). Low-dose placebos were intended to maintain participant blinding by producing some sensations of having ingested a psychedelic but without the same therapeutic psychedelic effect as the active treatment dose. Intervention and control group participants underwent the same intensive psychotherapy protocol, which typically consisted of 2–3 preparatory sessions, 2–3 MDMA or placebo dosing sessions, and 4–6 follow-up sessions (integration sessions) which were all co-led by 2 mental health professionals. More information regarding the MDMA dosing sessions is available in Appendix C of the Supplemental Materials. All trials were conducted by researchers affiliated with the Multidisciplinary Association for Psychedelic Studies (MAPS) or were sponsored by this organization and all reported using the same standardized manual to guide psychotherapy sessions (publicly available at http://maps.org/treatment-manual). No study compared MDMA-assisted psychotherapy to another active treatment (ie, psychotherapy paired with a different psychedelic at therapeutic dosages) or to no treatment. Follow-up duration in most trials was 1–2 months.

Table 3. Psychedelic-assisted Psychotherapy for PTSD: Study Characteristics.

Table 3

Psychedelic-assisted Psychotherapy for PTSD: Study Characteristics.

Although all available trials reported mean changes in CAPS total severity scores, only 5 also reported the proportion of participants who no longer met criteria for PTSD (ie, experienced disease remission) or the proportion who experienced a clinically meaningful improvement in symptoms (defined as ≥30% decrease in CAPS total severity score compared to baseline). Across these 5 trials,15 a higher proportion of participants who received MDMA-assisted psychotherapy experienced PTSD remission and/or at least a 30% decrease in CAPS scores compared to comparison group participants (Table 4).

Table 4. Effects of MDMA-assisted Psychotherapy on PTSD Diagnosis and Clinically Meaningful Improvement in CAPS Total Severity Score.

Table 4

Effects of MDMA-assisted Psychotherapy on PTSD Diagnosis and Clinically Meaningful Improvement in CAPS Total Severity Score.

Results of a meta-analysis of 7 trials (Figure 3) indicate that participants receiving MDMA experienced significantly greater improvements in mean CAPS total severity scores compared with participants receiving 0mg or low-dose placebo (pooled standardized mean difference in change scores = −0.91, 95% CI [−1.33, −0.50]). Heterogeneity in true effects appeared to be minimal (95% PI [−1.33, −0.50]). No studies stratified results by patient characteristics or PTSD severity at baseline. These findings are consistent with results of a small RCT69 conducted in Brazil that used a different assessment of PTSD symptomatology (Severity of Symptoms Scale for Post-traumatic Stress Disorder) and was terminated early due to reasons unrelated to trial conduct, as well as 3 observational studies.911

Across most studies, control group participants also experienced symptom improvements regardless of the type of placebo used, although to a lesser degree than treatment group participants (median standardized symptom change of −0.90 and −1.54, respectively). Because both treatment and control group participants received psychotherapy in all studies, this observation suggests that treatment benefits are at least partially attributable to MDMA. Nonetheless, the small number and size of available studies limits the extent to which firm conclusions can be drawn about the relative contributions of receiving MDMA, undergoing intensive psychotherapy, and other factors (such as participation effects) to observed symptom improvement.

Additionally, the precise magnitude of benefit that could be expected from MDMA is difficult to determine from the available evidence, in part because very small RCTs are at risk of prognostic imbalance (a failure of randomization to balance groups on participants’ responsiveness to treatment). The largest available RCT1 is less likely to be impacted by this bias, and provides an estimate of added treatment benefit due to MDMA that corresponds to an approximately 5 to 15-point greater reduction in PTSD symptom severity compared to psychotherapy only (unstandardized mean difference in CAPS-5 change scores = −10.5, 95% CI [−15.6, −5.4]). Like 2 other studies, however, this study used a 0mg placebo, which likely compromised participant blinding and may have led to biases in observed treatment effects.

Figure 3. Change in Total CAPS Score Following MDMA-assisted Psychotherapy Compared to 0mg Placebo or Low-dose Placebo.

Figure 3

Change in Total CAPS Score Following MDMA-assisted Psychotherapy Compared to 0mg Placebo or Low-dose Placebo. Abbreviations. Ctrl=control group; NR=not reported; Supp=supplemental dosage; RE=random-effects model; Tx=treatment group.

MAJOR DEPRESSIVE DISORDER

Psilocybin-assisted psychotherapy may reduce depression severity and lead to a clinically significant improvement or sustained remission for some recipients at 12 months. Ayahuasca use may be associated with a short-term reduction in mean depression symptom scores and suicidality. Our confidence in these findings is low given study methodological limitations, inconsistency, and small sample sizes. No serious adverse events were reported for either intervention.

We identified 2 small RCTs12,17 of psilocybin-assisted psychotherapy and 3 small observational studies1316 in participants with moderate to severe depression (Table 5). The intervention in both trials consisted of 2 doses of psilocybin 20–30mg paired with pre-and post-treatment psychotherapy sessions led by 2 mental health professionals. In 1 of the trials,12 conducted in the US among 24 adults with depression symptoms for a mean of 22 years, outcomes among the intervention group were compared to wait list controls who underwent the same treatment 8 weeks later. At baseline, participants had moderate depression according to the GRID-Hamilton Depression Rating Scale (GRID-HAMD; rated as no depression (0–7), mild depression (8–16), moderate depression (17–23), or severe depression (≥24)). If participants were taking antidepressants, they were required to refrain from using them prior to study screening and through completion of the primary outcome assessment but could start or restart antidepressant medication afterward. At 4-week and 3-, 6-, and 12-month follow-up, respectively, 0 (0%), 3 (12.5%), 5 (20.8%), and 8 (33.3%) of participants reported antidepressant use. Participants were allowed to continue psychotherapy throughout the trial if the type and frequency of psychotherapy had been stable and was expected to remain stable. At 4-week and 3-, 6-, and 12-month follow-up, respectively, 8 (33.3%), 9 (37.5%), 10 (41.7%), and 10 (41.7%) of participants reported receiving psychotherapy since receiving psilocybin.

Depression symptoms as measured by the GRID-HAMD significantly improved at 8 weeks compared to the delayed treatment group (standardized mean difference [SMD] = 2.6, 95% CI [1.5, 3.7]). In the immediate treatment group, the mean (SD) GRID-HAMD scores were 22.9 (3.6) at baseline, 8.0 (7.1) at week 5, and 8.5 (5.7) at week 8. Scores in the delayed treatment group remained stable: 22.5 (4.4) at baseline, 23.8 (5.4) at week 5, and 23.5 (6.0) at week 8. At 12 months, improvement on GRID-HAMD for all participants (immediate and delayed treatment groups) was also significant (SMD = 2.4, 95% CI [1.6, 3.2]).70 At 12 months, 18/24 (75%) of participants experienced a clinically significant response, which was defined by the study as a decrease of 50% or more from their pre-treatment GRID-HAMD total score, and 14/24 (58%) met criteria for disease remission, defined as a GRID-HAMD total score of 7 or lower. GRID-HAMD scores at 12 months were not significantly different among participants who reported receiving psychotherapy during the follow-up period (the same analysis was not reported for antidepressant use). Suicidal ideation as measured by the Columbia Suicide Severity Rating Scale (CSSRS) severity of ideation subscale (score range of 0–5,with higher scores indicating presence of ideation with at least some intent to die) was 1.2 (1.2), 0.3 (0.7), 0.2 (0.7), 0.5 (1.0), 0.6 (1.1), 0.6 (1.1), respectively, at baseline, 1-week, 4-week and 3-, 6-, and 12-month follow-up (N = 23 due to missing data). No serious adverse events were reported and there were no reported suicide attempts or instances of self-harm at 12 months. The trial did not stratify results by patient characteristics or disease severity.

In a second trial17 of psilocybin-assisted psychotherapy conducted in the UK that included 59 adults, the comparison group was treated with a low-dose placebo (psilocybin 1mg, presumed to have negligible activity) and escitalopram 10-20mg daily. The primary outcome measure was change from baseline scores on the Quick Inventory of Depressive Symptomatology-Self-Report (QIDS-SR-16; rated as no depression (0–5), mild depression (6–10), moderate depression (11–15), severe depression (16–20), or very severe depression (21–27)). At baseline, participants in the psilocybin group (N = 30) and escitalopram groups (N = 29), respectively, had mean QIDS-SR-16 scores (SD, range) of 14.5 (3.9; 7–23) and 16.4 (4.1; 6–22). Psilocybin group participants had depression for a mean of 22 years, had previously tried an average of 2 psychiatric medications, and 93% had previously undergone psychotherapy. Escitalopram group participants had depression for a mean of 15 years, had tried an average of 2 psychiatric medications, and 90% had previously undergone psychotherapy. Alcohol use was more common in the escitalopram group (average weekly use of 68 grams compared to 37 grams). Participants were required to stop any use of psychiatric medication at least 2 weeks before starting the trial and stop psychotherapy at least 3 weeks beforehand.

Both the psilocybin and escitalopram groups had reductions in mean QIDS-SR-16 scores. However, the change in mean scores from baseline to week 6 was not significantly different between the 2 groups (mean difference [MD] = −2.0, 95% CI [−5.0, 0.9]). A clinically meaningful response (defined as a decrease in QIDS-SR-16 by at least 50%) occurred in 21/30 (70%) of the psilocybin group and 14/29 (48%) in the escitalopram group. Remission (defined as QIDS-SR-16 score <5) occurred in 17/30 (57%) in the psilocybin group and 8/29 (28%) in the escitalopram group. Neither response nor remission rates were significantly different between groups (Table 5). Changes in suicidal ideation as measured by the Suicidal Ideation Attributes Scale (SIDAS) were also not significantly different. No serious adverse events occurred. The trial did not stratify results by patient characteristics or disease severity.

The same group of researchers who conducted the UK trial also published 4 small observational studies on an intervention consisting of 2 doses of psilocybin (10mg followed by 25mg) combined with psychological support before, during, and after psilocybin treatments.1316 The number of participants who were exposed to this intervention ranged from 12–20 in each study, but importantly, some participants overlapped between studies. Results consistently showed a reduction in depression severity (as measured by QIDS-16 and the Beck Depression Inventory) at 1–8 weeks following the intervention. In the study with the longest duration, the change in QIDS-SR-16 from baseline to 6 months among 20 participants was significant (within-group SMD = 1.4, p < 0.001).14 These observational studies aimed to test the feasibility of studying psilocybin-assisted psychotherapy and explore associations of psilocybin use with factors related to depression such as future life event forecasting and emotional face recognition (both outcomes are outside the scope of this review). Although necessary to establish a basis for further study, results of these observational studies did not weigh heavily in our assessment of the evidence compared with findings from RCTs.

Table 5. Study Characteristics and Outcomes of Psilocybin-assisted Psychotherapy for Depression.

Table 5

Study Characteristics and Outcomes of Psilocybin-assisted Psychotherapy for Depression.

We identified 1 small RCT18 (N = 29) on ayahuasca for treatment of depression and 2 small pre-post studies (Table 6). In the trial conducted in Brazil, participants had moderate to severe depression as defined by mean Hamilton Depression Rating (HAM-D; rated as mild (10–13), mild to moderate (14–17), or moderate to severe (>17)) with a mean (SD) baseline score of 21.83 (5.35), had tried at least 2 antidepressant medications, and most (80%) had previously tried psychotherapy. Participants in the intervention group received 1 dose of ayahuasca without additional co-interventions aside from supervision during the dosing session and follow-up study assessments. The control group received an 0mg placebo designed to mimic the taste and color of ayahuasca. Mean HAM-D scores 1 week after treatment were significantly different between groups and favored ayahuasca (SMD = 0.98, 95% CI [0.21, 1.75]). A higher proportion of participants in the ayahuasca group met criteria for a clinically meaningful response (reduction in HAM-D score by at least 50%) and remission (HAM-D ≤7) (Table 6). This trial did not follow participants longer than 1 week, so the durability of these responses is unknown. Suicidal ideation decreased in both groups, but between-group differences were not significant. It is notable that 76% of participants had a comorbid personality disorder, 31% had comorbid anxiety disorder, and all were under regular use of benzodiazepines during the trial. Whether these factors influenced results is unclear, and the trial did not stratify results by patient characteristics.

The same researchers who conducted this RCT also conducted 2 small observational studies19,20,22 that identified similar reductions in depression symptom severity in the 3 weeks following use of ayahuasca by 23 adults with recurrent depression. Among 15 patients with suicidality at baseline, ayahuasca use was associated with a reduction in suicidality as measured by Montgomery-Åsberg Depression Rating Scale-Suicidality Item (MADRS-SI) in the acute period (up to 180 minutes after ayahuasca administration) and for the next 21 days. These observational studies contributed little to our assessment of the evidence given that RCT data are available. No serious adverse events were reported for any studies of ayahuasca. 30

Table 6. Study Characteristics and Outcomes of Ayshuasca for Depression.

Table 6

Study Characteristics and Outcomes of Ayshuasca for Depression.

OPIOID USE

Ibogaine may not reduce short-term opioid withdrawal symptoms compared to placebo. Our confidence in this finding is low based on the small number of studies available, study methodologic limitations, and inconsistent findings. Although several pre-post studies2428 found that ibogaine use was associated with a reduction in opioid withdrawal symptoms, this finding was not replicated in an RCT (Table 7).23 Whether ibogaine use is associated with long-term opioid use reduction is unclear. Only 3 small uncontrolled observational studies with high attrition have evaluated long-term outcomes. Available evidence is therefore insufficient to draw conclusions about the long-term efficacy of ibogaine. Importantly, ibogaine has been associated with serious adverse events, namely prolonged QTc interval on an electrocardiogram (ECG) and cardiac arrhythmia. Across studies, 1 death occurred during treatment.

Only 1 small RCT has been conducted on ibogaine use to treat opioid withdrawal symptoms. In this trial23 (which was conducted in New Zealand and designed to evaluate ibogaine safety), 27 participants on methadone treatment for OUD were randomized to receive 1 of 3 doses of noribogaine (an ibogaine active metabolite at 60mg, 120mg, or 240mg) or placebo in the immediate time period following their last use of an opioid. In the week before receiving noribogaine, participants stopped methadone and received long-acting morphine for 6 days followed by short-acting morphine for 1 day. Approximately 2 hours after their last dose of short-acting morphine, participants received noribogaine or placebo and were monitored for withdrawal symptoms. The study’s primary efficacy outcome was the time in hours before participants requested a dose of morphine or methadone. The trial found that noribogaine at any dose did not result in a longer time to resuming opioid use (and did not significantly reduce withdrawal symptoms) compared to placebo, noting that participants in both groups were treated in the same physical space and could have influenced each other. None of the participants experienced serious adverse effects. Norigobaine was associated with dose-dependent effect on QTc interval prolongation on electrocardiogram, which is a risk factor for life-threatening cardiac arrhythmias. The trial did not follow participants long term.

In a small uncontrolled observational study27 in the Netherlands with similar research aims as the New Zealand trial, 14 adults on methadone treatment for OUD who were transitioned to morphine received ibogaine 10mg/kg and were monitored for withdrawal. Most participants (11/14, 79%) did not restart morphine within 24 hours, and among these participants the median Clinical Opiate Withdrawal Scale (COWS) score was 0, indicating no or mild withdrawal symptoms. Half of participants had a prolonged QTc >500ms during the treatment period (a change from baseline) and received magnesium infusions as a result, without any adverse consequences. All participants experienced ataxia with an inability to walk without support, which reversed within 48 hours. Like the New Zealand trial, this study did not follow participants long term.

Four additional uncontrolled observational studies2426,28 of adults receiving ibogaine with the intent of opioid detoxification (N = 14–88) found that participants had lower measures of opioid withdrawal and/or opioid craving post-ibogaine compared to pre-ibogaine use. In a small observational study26 conducted in New Zealand, 14 adults completed treatment for opioid withdrawal with ibogaine 25–55mg/kg, diazepam 5–30mg, and zopiclone 7.5–15mg. On average, participants reported having fewer withdrawal symptoms 12–24 hours after receiving ibogaine compared to baseline. One participant (not included in the analysis) died during treatment under circumstances that are not well described. In a report by New Zealand officials, the death was deemed very likely “related to ibogaine ingestion and most probably related to a cardiac arrhythmia.” A subset of participants (8/14, 57%) were followed for 12 months. Among these participants, the Addiction Severity Index Lite (ASI-Lite) for drug use was significantly lower at 12 months compared with baseline, and most participants (6/8, 75%) had negative drug screens at 12 months. Only 1 other study reported on long-term opioid use. In a retrospective study of 88 adults who received ibogaine at a treatment center in Mexico with a history of heroin (51%) or prescription opioid use (49%), 30% of participants reported never using opioids again, while 48% said that their opioid use was reduced compared to the period before ibogaine treatment, 17% reported no change, and 6% increased their use.25

Table 7. Study Characteristics and Outcomes of Psychedelic Treatment for OUD.

Table 7

Study Characteristics and Outcomes of Psychedelic Treatment for OUD.

ALCOHOL USE

Psilocybin-assisted psychotherapy may reduce heavy drinking days and average alcohol consumption per day among adults with alcohol use disorder. Although results are consistent and direct, our confidence in these findings is low due to study methodologic limitations and small sample sizes. MDMA-assisted psychotherapy for alcohol use has been less studied and evidence is currently insufficient to draw conclusions.

One RCT29 has been conducted on psilocybin-assisted psychotherapy for alcohol disorder (Table 8). In this US trial, 95 adults with an average of 14 years of alcohol dependence were randomized to receive 12 psychotherapy sessions with 2 psilocybin dosing sessions or diphenhydramine 50–100mg. At baseline, participants in the psilocybin group consumed a mean (SD) of 7.52 (4.58) standard drinks per day (defined as 14 grams of ethanol) compared to 7.10 (4.05) in the diphenhydramine group. The study did not report whether participants had previous experience with treatment for alcohol dependence. Participants receiving psilocybin experienced a significantly greater reduction in the percent of heavy drinking days and number of drinks per day compared to the diphenhydramine group at 32-week follow-up. Reduction in the percent of drinking days also favored psilocybin, but this finding was nonsignificant, and improvements on all outcomes were observed in both groups. No serious adverse events occurred among participants who received psilocybin.

In a small uncontrolled observational study conducted by the same group as the trial,30 10 adults were administered a 14-week psilocybin-assisted psychotherapy program that included 1–2 psilocybin dosing sessions starting in week 4. In the period after dosing (weeks 5–12), the percent of heavy drinking days and any drinking days were decreased compared with baseline and with the period prior to the initial dosing of psilocybin. At the study endpoint of 36 weeks, percent heavy drinking days and any drinking days were trending upward but still below baseline.

MDMA-assisted psychotherapy was evaluated as a treatment for alcohol use disorder in a UK study31 of 14 adults with mostly moderate to severe alcohol use disorder. Participants were enrolled in an 8-week treatment program consisting of 10 psychotherapy sessions, in 2 of which they received MDMA. At 9 months post-treatment, 9 participants were abstinent from alcohol and 2 had reduced their use to fewer than 14 units of alcohol per week.

Table 8. Study Characteristics and Outcomes of Psychedelic-assisted Psychotherapy for AUD.

Table 8

Study Characteristics and Outcomes of Psychedelic-assisted Psychotherapy for AUD.

TOBACCO USE

Evidence regarding psychedelic-assisted psychotherapy for tobacco use is limited to 1 small uncontrolled observational study of psilocybin use, which is insufficient to make conclusions about efficacy. The average participant in this US study32,33 (N = 15) was aged 51, smoked 19 cigarettes per day for a 31 years, and had 6 previous lifetime quit attempts (see Appendix C in the Supplemental Materials for additional study characteristics). The intervention consisted of intensive cognitive behavioral therapy delivered in 19 in-person sessions, with 3 sessions of psilocybin administration as well as brief daily phone calls for 2 weeks following participants’ target quit dates. At 12 months, 10/15 (67%) adults were abstinent from smoking as measured by smoking timeline follow-back assessments and verified by exhaled carbon monoxide and urinary cotinine levels. Among those who participated in longer-term follow-up (range: 16–57 months), 9/12 (60%) were abstinent from smoking by the same measures. No serious adverse events were reported.

In addition to its small sample size, a limitation of this study is the potential for selection bias. Although participants were typical of those seeking medical care for tobacco cessation in terms of tobacco use history and the number of quit attempts, most (10/15, 67%) reported past use of hallucinogens. It is possible that the participants with past hallucinogen use had different expectations regarding the potential of this treatment compared to those without past hallucinogen use.

COCAINE AND CANNABIS USE

Psychedelics, specifically ayahuasca and ibogaine, have also been evaluated in uncontrolled observational studies as treatment for other substance use including cocaine and cannabis. Due to study methodologic limitations, small sample sizes, indirectness, and inconsistency, this evidence is insufficient to draw conclusions about the effects of ayahuasca and ibogaine on use of cocaine and cannabis. See Appendix C in the Supplemental Materials for detailed study characteristics.

One observational study34 evaluated the use of ayahuasca among participants with mixed forms of substance use. In this Canadian pre-post study34 of 12 adults participating in a 4-day “Working with Addiction and Stress” retreat that incorporated ayahuasca ceremonies, the proportion of participants who reported using alcohol, tobacco, and cocaine declined. No change was observed for opiate or cannabis use, and no serious adverse events were reported.

Three observational studies36,37,71 of ibogaine have also been conducted among individuals with cocaine or opioid dependence who self-referred to a treatment program on St. Kitts in the West Indies. The largest and most recent study37 of this treatment program included 102 adults with opioid dependence and 89 with cocaine dependence who underwent 1 ibogaine dosing session during a 12-day inpatient detoxification program. Self-reported cravings as measured by the Heroin (HCQ-29) and Cocaine (CCQ-45) Craving Questionnaires were lower at the time of discharge compared to before treatment, and remained lower among 37 participants with opioid dependence and 32 with cocaine dependence who completed follow-up at 1 month. No serious adverse events were reported, though orthostatic hypotension occurred in 5% of participants and transient nausea, vomiting, and ataxia were reported as common side effects shortly after ibogaine administration.

In a retrospective study35 of 75 adults in Brazil who underwent 1–9 ibogaine dosing sessions in conjunction with outpatient psychotherapy, 61% of participants reported abstinence. The duration of reported abstinence was longer for participants who underwent multiple ibogaine dosing sessions compared to 1 dosing session (median 8.4 months compared to median 5.5 months). No serious adverse events were reported.

OTHER CONDITIONS

Psychedelic substances have been investigated as treatment for social anxiety, OCD, and for non-specific mood and anxiety symptoms in a single RCT and 2 uncontrolled observational studies. This evidence is insufficient to draw any conclusions about the effectiveness of psychedelics for these disorders. See Appendix C in the Supplemental Materials for detailed study characteristics.

An RCT38 of 17 adults with clinical symptoms of social anxiety disorder (SAD) found that Beck Anxiety Inventory scores were not significantly different after a single ayahuasca dosing session compared to 0mg placebo.38 However, ayahuasca was associated with a significant improvement in self-perception of speech performance as measured by the Self-Statements During Public Speaking Scale. In a pre-post study39 of 9 adults with OCD in the US who received variable doses of psilocybin for up to 4 total treatments, all participants experienced a decrease in OCD symptoms as measured by the Yale-Brown Obsessive Compulsive Scale (23–100% reduction in YBOCS scores).

Lastly, ibogaine plus 5-MeO-DMT has been used as treatment for US Special Forces Veterans with a range of mood and anxiety symptoms. A retrospective study40 of 51 Veterans who participated in a psychedelic clinical program in Mexico during which they received 1 dose of ibogaine and 3 doses of 5-MeO-DMT found significant reductions in PTSD symptoms (SMD PTSD Checklist-5 = −3.6, p < 0.001), depression (SMD Patient Health Questionnaire-2 = −3.7, p < 0.001), anxiety (SMD Generalized Anxiety Disorder 2-Item = −3.1, p < 0.001), and suicidal ideation (SMD Depressive Symptom Index Suicidality Subscale = −1.9, p < 0.001) 30-days post-treatment.

HARMS

Transient side effects were commonly reported and may occur before, during, or after a psychedelic session (for full list of observed harms see Supplemental Materials Appendix C).

Ibogaine has been associated with serious adverse events, including death in 1 study.26 Several studies found a dose-dependent effect of ibogaine on QTc interval prolongation on ECG, which is a risk factor for cardiac arrhythmia.23,27 Other common side effects of ibogaine include headache, nausea, imbalance, and visual impairment.

MDMA may also be associated with cardiac risks. Only 1 study reported a serious adverse event attributed to MDMA, a participant with premature ventricular contractions (PVCs) at baseline experienced an increase in PVCs requiring an overnight hospital stay for observation.2 Other transient side effects of MDMA include symptoms related to mood (eg, anxiety, irritability, restlessness), sleep (eg, fatigue, insomnia), sensation (eg, dizziness, impaired balance), pain (eg headache, muscle tension), and gastrointestinal systems (eg, nausea, vomiting, low appetite).

Psilocybin was associated with transient effects on heart rate (eg, tachycardia, palpitations) and elevated blood pressure, but these events were not described as serious. Common side effects of psilocybin include anxiety, fear, headache, and nausea. Ayahuasca was not associated with serious adverse events in included studies. Common side effects of ayahuasca include headache, nausea, and vomiting.

DISCUSSION

The aim of this review was to synthesize evidence on the benefits and harms of psychedelics as primary or adjunct treatment for mental health and substance use disorders. Given the prevalence of mental health and substance use disorders with the Veteran population, any novel or emerging treatments with the potential to improve clinical outcomes are of great interest.

MDMA-assisted psychotherapy for PTSD is the most extensively studied intervention involving psychedelics. In a clinical research setting, MDMA paired with intensive psychotherapy co-led by 2 mental health professionals may reduce PTSD symptom severity and lead to clinically meaningful improvement and/or disease remission for some participants in the short term. Although results are fairly consistent and direct, trial sample sizes are small and at risk of prognostic imbalance (a failure of randomization to balance groups on participants’ responsiveness to treatment). Common side effects of MDMA include anxiety, restlessness, headache, and nausea but serious adverse events appear to be rare.

Although promising, our confidence in these findings is low. The evidence on MDMA-assisted psychotherapy remains preliminary and several important research questions have yet to be addressed. Among these is whether outcomes vary by patient characteristics, index trauma type, disorder severity, or the number or types of past treatments tried. Available evidence also has unclear applicability to Veteran populations, as most studies did not include Veterans and none have been conducted in a VHA setting. Moreover, the psychotherapy component of studied interventions—typically requiring 30 hours or more of psychotherapy co-led by 2 mental health professionals—is more intensive than typically occurs in clinical practice. Researchers are beginning to study whether treatment protocols can be reproduced at scale,11 but this evidence is preliminary. Whether this level of treatment is feasible in everyday practice and whether treatment benefits may still be observed with lower intensity psychotherapy are questions for future research.

Other interventions involving psychedelics have been less extensively studied. Psilocybin-assisted psychotherapy for depression may reduce symptom severity and lead to sustained remission at 12 months for a proportion of recipients compared to wait list controls, but these same benefits were not observed (in a separate trial) when psilocybin-assisted psychotherapy was compared with psychotherapy and daily escitalopram. We have low confidence in these findings, which are based on 1 small trial for each comparator. Ayahuasca use for depression may be associated with a reduction in short-term symptoms and suicidality. However, our confidence in these findings is also low due to a smaller number of trials with small sample sizes and some inconsistency in results.

Among treatments evaluated for substance use disorders, psilocybin-assisted psychotherapy for alcohol use disorder may reduce alcohol consumption (heavy drinking days and the number of drinks per day), but we have low confidence is this finding (which is based on results of 1 small trial). MDMA-assisted psychotherapy has also been studied for treatment of alcohol use disorder, but results are limited to 1 small observational study and are insufficient to make conclusions. Similarly, evidence on psilocybin as an adjunct to psychotherapy to reduce tobacco use and ayahuasca as treatment for cocaine, cannabis, and other forms of substance use is limited to 1 or a small number of observational studies and is insufficient to make conclusions.

Ibogaine may not reduce opioid-related withdrawal symptoms or reduce long-term opioid use. We have low confidence in these findings, which are drawn from a small number of studies with small sample sizes, study methodologic limitations, and inconsistent findings. Ibogaine had also been studied as a treatment to reduce cocaine use and to reduce problematic general substance use, but results are limited to a small number of observational studies with mixed populations and evidence is insufficient to draw conclusions. Ibogaine plus 5-MeO-DMT was found to improve a range of mood and anxiety symptoms in a retrospective study of US Special Forces Veterans participating in a treatment program in Mexico. However, this study had a high risk of bias and results have not been replicated in other studies. Given safety concerns associated with ibogaine use, particularly the documented risk of QTc prolongation, cardiac arrhythmias, and death, treatment should only be undertaken in the context of a clinical trial with appropriate safety monitoring.

Finally, most available studies on psychedelics required participants to discontinue their usual mental health medications, most often for a least 2 weeks before the study start. Risk of harms arising from medication discontinuation may have been minimal in the research context (due to presumably close patient contact and oversight), but conceivably could be greater in real-world settings. If the recommendation to discontinue medication becomes accepted practice as psychedelics move through the FDA drug development pathway, the risks of doing so must be taken into consideration when assessing the overall balance of risks and benefits of psychedelic treatments.

LIMITATIONS

Limitations of our review methods include our use of sequential review (rather than dual independent review) for data abstraction and risk of bias assessment. Additionally, both statistical precision and heterogeneity can be poorly estimated in small meta-analyses. We took steps to ameliorate these concerns, including use of corrections to better account for uncertainty in the estimation of heterogeneity, but some caution should be used in interpreting reported meta-analytic confidence intervals and prediction intervals.

The available evidence on psychedelics as primary or adjunct treatment for mental health disorders and substance use also has several limitations. First, psychedelics have been studied in a limited number of populations and settings by a relatively small number of researchers and institutions, which has partly been in response to legal and regulatory restrictions. Despite broad recruitment efforts, it is possible that participants of studies to date are not representative of general adult populations with mental health or substance use disorders. For example, US studies have been predominantly conducted among young, mostly non-Hispanic white populations with moderate to severe disease and access to a clinical research center or treatment program. Whether treatment effects can be replicated in a broader array of populations and treatment settings is unclear. Similarly, little is known about the durability of psychedelic treatment effects, as most studies followed participants for no longer than a year. Whether exposure to psychedelics changes the long-term symptomatology of adults diagnosed with PTSD or depression, for example, is largely unknown.

Second, despite the use of low-dose placebos in some trials (eg, use of low-dose placebo MDMA in studies of MDMA-assisted psychotherapy for PTSD), many participants were able to correctly guess whether they were in the intervention or comparison groups (among trials that reported this information). Participant blinding was likely largely ineffective in studies that used a 0mg placebo, which may have biased observed treatment effects.

Lastly, because research on psychedelics is still in an early phase, whether and how psychedelic treatments could fit into usual care pathways is unclear. Even for areas with some promise, such as MDMA-assisted psychotherapy for PTSD, several clinical questions remain including whether benefits are limited to those with moderate to severe disease, whether adults should try more established treatments before considering psychedelic treatment, and if not, which criteria should be met to recommend psychedelics as primary treatment. Available evidence has also not addressed whether those who improve with psychedelic treatments should start or restart other therapies to maintain benefits long term. Additionally, the extent to which psychotherapy protocols employed in interventions involving psychedelics are consistent with current VA/Department of Defense (DoD) guidelines is also unclear. For example, studies of MDMA-assisted psychotherapy for PTSD used the MAPS standard treatment manual to guide psychotherapy sessions, but it is unclear to what degree the MAPS approach overlaps with currently recommended trauma-focused psychotherapies such as Prolonged Exposure as outlined in the applicable VA/DoD guideline (https://www.healthquality.va.gov/guidelines/mh/ptsd/).

FUTURE RESEARCH

Recommendations for future research on psychedelics as primary or adjunct treatment for mental health and substance use disorders include:

  • Inclusion of Veteran populations, particularly those with combat-related trauma, with detailed reporting of treatment histories and disease severity at baseline.
  • Inclusion of more diverse populations, particularly with respect to race/ethnicity, socioeconomic status, and rurality, to better understand variability in treatment effectiveness, safety, and acceptability.
  • Evaluation of the clinical implications of discontinuing mental health medications prior to undergoing psychedelic treatments and whether the benefits of doing so outweigh potential harms.
  • Use of low-dose placebo rather than 0mg placebo to improve the adequacy of blinding, and improved reporting of blinding effectiveness.

Questions to be addressed at the health system level include:

  • How could and should interventions involving psychedelics be incorporated into current standard of care and usual care pathways?
  • Are psychedelic treatment protocols scalable and reproducible across a broad range of regions and settings?
  • What are best practices for implementation of psychedelic treatment programs?
  • If implemented, how can access to these resource-intensive treatments be distributed equitably?

A large number of underway studies were identified (see Appendix C in the Supplemental Materials for full details) in a wide range of populations (eg, adults with PTSD, mood disorders, and substance use). However, most are feasibility or pilot trials that may not fully address these research gaps.

CONCLUSIONS

Novel approaches to treatment of adults with mental health and substance use disorders are needed given the limitations of existing therapies. MDMA-assisted psychotherapy for PTSD, the most extensively studied intervention to date, may reduce PTSD symptom severity and lead to disease remission in a proportion of cases. Psilocybin-assisted psychotherapy and ayahuasca use for depression and psilocybin for alcohol use disorder may also be effective. However, our confidence in these findings is low, mainly due to the small number of trials available and the small sample sizes of these studies. Treatments that utilize psychedelics are still in an early phase of development and many gaps in the evidence need to be addressed. The most critical gaps include whether treatment effects are durable and consistent across patient characteristics and disease severity, whether the benefits of discontinuing mental health medications to undergo psychedelic treatments outweigh risks, and whether psychedelic treatment protocols are reproducible and scalable in everyday practice, including in VHA settings.

REFERENCES

1.
Mitchell JM, Bogenschutz M, Lilienstein A, et al. MDMA-assisted therapy for severe PTSD: a randomized, double-blind, placebo-controlled phase 3 study. Nature Medicine. 2021;27(6):1025–1033. [PMC free article: PMC8205851] [PubMed: 33972795]
2.
Mithoefer MC, Mithoefer AT, Feduccia AA, et al. 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy for post-traumatic stress disorder in military veterans, firefighters, and police officers: a randomised, double-blind, dose-response, phase 2 clinical trial. The Lancet Psychiatry. 2018;5(6):486–497. [PubMed: 29728331]
3.
Mithoefer MC, Wagner MT, Mithoefer AT, Jerome L, Doblin R. The safety and efficacy of {+/-}3,4-methylenedioxymethamphetamine-assisted psychotherapy in subjects with chronic, treatment-resistant posttraumatic stress disorder: the first randomized controlled pilot study. Journal of Psychopharmacology. 2011;25(4):439–452. [PMC free article: PMC3122379] [PubMed: 20643699]
4.
Ot’alora GM, Grigsby J, Poulter B, et al. 3,4-Methylenedioxymethamphetamine-assisted psychotherapy for treatment of chronic posttraumatic stress disorder: A randomized phase 2 controlled trial. Journal of Psychopharmacology. 2018;32(12):1295–1307. [PMC free article: PMC6247454] [PubMed: 30371148]
5.
Oehen P, Traber R, Widmer V, Schnyder U. A randomized, controlled pilot study of MDMA (+/− 3,4-Methylenedioxymethamphetamine)-assisted psychotherapy for treatment of resistant, chronic Post-Traumatic Stress Disorder (PTSD). Journal of Psychopharmacology. 2013;27(1):40–52. [PubMed: 23118021]
6.
Kotler M. A Randomized, Double-Blind, Active Placebo-Controlled Phase 2 Pilot Study of MDMA-assisted Psychotherapy in People With Chronic, Treatment-Resistant Posttraumatic Stress Disorder (PTSD). NCT01689740. Multidisplinary Association for Psychedelic Studies. https:​//clinicaltrials​.gov/ct2/show/record​/NCT01689740?term=NCT01689740&draw​=2&rank=1. Published 2017. Accessed August 22, 2022.
7.
Pacey I. A Randomized, Double-Blind, Controlled Phase 2 Pilot Study of Manualized 3,4-methylenedioxymethamphetamine (MDMA)-Assisted Psychotherapy in 12 Subjects With Treatment-Resistant Posttraumatic Stress Disorder (PTSD) - Canada. NCT01958593. Multidisciplinary Association for Psychedelic Studies. https:​//clinicaltrials​.gov/ct2/show/record​/NCT01958593?term=NCT01958593&draw​=2&rank=1. Published 2017. Accessed August 22, 2022.
8.
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. Journal of Psychopharmacology. 2013;27(1):28–39. [PMC free article: PMC3573678] [PubMed: 23172889]
9.
Jardim AV, Jardim DV, Chaves BR, et al. 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy for victims of sexual abuse with severe post-traumatic stress disorder: an open label pilot study in Brazil. Revista Brasileira de Psiquiatria. 2021;43(2):181–185. [PMC free article: PMC8023155] [PubMed: 32638920]
10.
Monson CM, Wagner AC, Mithoefer AT, et al. MDMA-facilitated cognitive-behavioural conjoint therapy for posttraumatic stress disorder: An uncontrolled trial. European Journal of Psychotraumatology. 2020;11(1):1840123. [PMC free article: PMC7748057] [PubMed: 33408811]
11.
Wang JB, Lin J, Bedrosian L, et al. Scaling Up: Multisite Open-Label Clinical Trials of MDMA-Assisted Therapy for Severe Posttraumatic Stress Disorder. Journal of Humanistic Psychology. 2021;0(0):00221678211023663.
12.
Davis AK, Barrett FS, May DG, et al. Effects of Psilocybin-Assisted Therapy on Major Depressive Disorder: A Randomized Clinical Trial. JAMA Psychiatry. 2021;78(5):481–489. [PMC free article: PMC7643046] [PubMed: 33146667]
13.
Carhart-Harris RL, Bolstridge M, Rucker J, et al. Psilocybin with psychological support for treatment-resistant depression: an open-label feasibility study. The Lancet Psychiatry. 2016;3(7):619–627. [PubMed: 27210031]
14.
Carhart-Harris RL, Bolstridge M, Day CMJ, et al. Psilocybin with psychological support for treatment-resistant depression: six-month follow-up. Psychopharmacology. 2018;235(2):399–408. [PMC free article: PMC5813086] [PubMed: 29119217]
15.
Lyons T, Carhart-Harris RL. More realistic forecasting of future life events after psilocybin for treatment-resistant depression. Frontiers in Psychology. 2018;9. [PMC free article: PMC6194345] [PubMed: 30369890]
16.
Stroud JB, Freeman TP, Leech R, et al. Psilocybin with psychological support improves emotional face recognition in treatment-resistant depression. Psychopharmacology. 2018;235(2):459–466. [PMC free article: PMC5813058] [PubMed: 29085980]
17.
Carhart-Harris R, Giribaldi B, Watts R, et al. Trial of Psilocybin versus Escitalopram for Depression. New England Journal of Medicine. 2021;384(15):1402–1411. [PubMed: 33852780]
18.
Palhano-Fontes F, Barreto D, Onias H, et al. Rapid antidepressant effects of the psychedelic ayahuasca in treatment-resistant depression: a randomized placebo-controlled trial. Psychological Medicine. 2019;49(4):655–663. [PMC free article: PMC6378413] [PubMed: 29903051]
19.
Sanches RF, de Lima Osorio F, Dos Santos RG, et al. Antidepressant Effects of a Single Dose of Ayahuasca in Patients With Recurrent Depression: A SPECT Study. Journal of Clinical Psychopharmacology. 2016;36(1):77–81. [PubMed: 26650973]
20.
Osorio L, Sanches RF, Macedo LR, et al. Antidepressant effects of a single dose of ayahuasca in patients with recurrent depression: a preliminary report. Revista Brasileira de Psiquiatria. 2015;37(1):13–20. [PubMed: 25806551]
21.
Zeifman RJ, Palhano-Fontes F, Hallak J, Arcoverde E, Maia-Oliveira JP, Araujo DB. The Impact of Ayahuasca on Suicidality: Results From a Randomized Controlled Trial. Frontiers in Pharmacology. 2019;10. [PMC free article: PMC6878725] [PubMed: 31798447]
22.
Zeifman RJ, Singhal N, Dos Santos RG, et al. Rapid and sustained decreases in suicidality following a single dose of ayahuasca among individuals with recurrent major depressive disorder: results from an open-label trial. Psychopharmacology. 2021;238(2):453–459. [PubMed: 33118052]
23.
Glue P, Cape G, Tunnicliff D, et al. Ascending Single-Dose, Double-Blind, Placebo-Controlled Safety Study of Noribogaine in Opioid-Dependent Patients. Clinical Pharmacology in Drug Development. 2016;5(6):460–468. [PubMed: 27870477]
24.
Brown TK, Alper K. Treatment of opioid use disorder with ibogaine: detoxification and drug use outcomes. American Journal of Drug & Alcohol Abuse. 2018;44(1):24–36. [PubMed: 28541119]
25.
Davis AK, Barsuglia JP, Windham-Herman A-M, Lynch M, Polanco M. Subjective effectiveness of ibogaine treatment for problematic opioid consumption: Short- and long-term outcomes and current psychological functioning. Journal of Psychedelic Studies. 2017;1(2):65–73. [PMC free article: PMC6157925] [PubMed: 30272050]
26.
Noller GE, Frampton CM, Yazar-Klosinski B. Ibogaine treatment outcomes for opioid dependence from a twelve-month follow-up observational study. American Journal of Drug & Alcohol Abuse. 2018;44(1):37–46. [PubMed: 28402682]
27.
Knuijver T, Schellekens A, Belgers M, et al. Safety of ibogaine administration in detoxification of opioid-dependent individuals: a descriptive open-label observational study. Addiction. 2022;117(1):118–128. [PMC free article: PMC9292417] [PubMed: 33620733]
28.
Malcolm BJ, Polanco M, Barsuglia JP. Changes in Withdrawal and Craving Scores in Participants Undergoing Opioid Detoxification Utilizing Ibogaine. Journal of Psychoactive Drugs. 2018;50(3):256–265. [PubMed: 29608409]
29.
Bogenschutz MP, Ross S, Bhatt S, et al. Percentage of Heavy Drinking Days Following Psilocybin-Assisted Psychotherapy vs Placebo in the Treatment of Adult Patients With Alcohol Use Disorder: A Randomized Clinical Trial. JAMA Psychiatry. 2022. [PMC free article: PMC9403854] [PubMed: 36001306]
30.
Bogenschutz MP, Forcehimes AA, Pommy JA, Wilcox CE, Barbosa PC, Strassman RJ. Psilocybin-assisted treatment for alcohol dependence: a proof-of-concept study. Journal of Psychopharmacology. 2015;29(3):289–299. [PubMed: 25586396]
31.
Sessa B, Higbed L, O’Brien S, et al. First study of safety and tolerability of 3,4-methylenedioxymethamphetamine-assisted psychotherapy in patients with alcohol use disorder. Journal of Psychopharmacology. 2021;35(4):375–383. [PubMed: 33601929]
32.
Johnson MW, Garcia-Romeu A, Cosimano MP, Griffiths RR. Pilot study of the 5-HT2AR agonist psilocybin in the treatment of tobacco addiction. Journal of Psychopharmacology. 2014;28(11):983–992. [PMC free article: PMC4286320] [PubMed: 25213996]
33.
Johnson MW, Garcia-Romeu A, Griffiths RR. Long-term follow-up of psilocybin-facilitated smoking cessation. The American Journal of Drug and Alcohol Abuse. 2017;43(1):55–60. [PMC free article: PMC5641975] [PubMed: 27441452]
34.
Thomas G, Lucas P, Capler NR, Tupper KW, Martin G. Ayahuasca-assisted therapy for addiction: results from a preliminary observational study in Canada. Current Drug Abuse Reviews. 2013;6(1):30–42. [PubMed: 23627784]
35.
Schenberg EE, de Castro Comis MA, Chaves BR, da Silveira DX. Treating drug dependence with the aid of ibogaine: a retrospective study. Journal of Psychopharmacology. 2014;28(11):993–1000. [PubMed: 25271214]
36.
Mash DC, Kovera CA, Pablo J, et al. Ibogaine: complex pharmacokinetics, concerns for safety, and preliminary efficacy measures. Annals of the New York Academy of Sciences. 2000;914:394–401. [PubMed: 11085338]
37.
Mash DC, Duque L, Page B, Allen-Ferdinand K. Ibogaine Detoxification Transitions Opioid and Cocaine Abusers Between Dependence and Abstinence: Clinical Observations and Treatment Outcomes. Front Pharmacol. 2018;9:529. [PMC free article: PMC5996271] [PubMed: 29922156]
38.
Dos Santos RG, Osorio FL, Rocha JM, et al. Ayahuasca Improves Self-perception of Speech Performance in Subjects With Social Anxiety Disorder: A Pilot, Proof-of-Concept, Randomized, Placebo-Controlled Trial. Journal of Clinical Psychopharmacology. 2021;41(5):540–550. [PubMed: 34166299]
39.
Moreno FA, Wiegand CB, Taitano EK, Delgado PL. Safety, tolerability, and efficacy of psilocybin in 9 patients with obsessive-compulsive disorder. Journal of Clinical Psychiatry. 2006;67(11):1735–1740. [PubMed: 17196053]
40.
Davis AK, Averill LA, Sepeda ND, Barsuglia JP, Amoroso T. Psychedelic treatment for trauma-related psychological and cognitive impairment among US Special Operations Forces Veterans. Chronic Stress. 2020;4:2470547020939564. [PMC free article: PMC7359647] [PubMed: 32704581]
41.
Nichols DE. Psychedelics. Pharmacol Rev. 2016;68(2):264–355. [PMC free article: PMC4813425] [PubMed: 26841800]
42.
Garcia-Romeu A, Kersgaard B, Addy PH. Clinical applications of hallucinogens: A review. Exp Clin Psychopharmacol. 2016;24(4):229–268. [PMC free article: PMC5001686] [PubMed: 27454674]
43.
Tupper KW, Wood E, Yensen R, Johnson MW. Psychedelic medicine: a re-emerging therapeutic paradigm. CMAJ Canadian Medical Association Journal. 2015;187(14):1054–1059. [PMC free article: PMC4592297] [PubMed: 26350908]
44.
Nichols DE. Entactogens: How the Name for a Novel Class of Psychoactive Agents Originated. Front Psychiatry. 2022;13:863088. [PMC free article: PMC8990025] [PubMed: 35401275]
45.
Sessa B. The History of Psychedelics in Medicine. Handbuch Psychoaktive Substanzen 2016.
46.
Nichols DE, Walter H. The History of Psychedelics in Psychiatry. Pharmacopsychiatry. 2021;54(4):151–166. [PubMed: 33285579]
47.
Psychotropic Substances Act of 1978. In. Public Law 95-633. Vol Statute 92: Senate and House of Representatives of the United States of America; 1978.
48.
Marks M. Psychedelic medicine for mental illness and substance use disorders: overcoming social and legal obstacles. NYUJ Legis & Pub Pol’y. 2018;21:69.
49.
Compass Pathways. COMPASS Pathways Receives FDA Breakthrough Therapy Designation for Psilocybin Therapy for Treatment-Resistant Depression. Cision PR Newswire. https://www​.prnewswire​.com/news-releases​/compass-pathways-receives-fda-breakthrough-therapy-designation-for-psilocybin-therapy-for-treatment-resistant-depression-834088100.html. Published 2018. Accessed August 2018.
50.
Multidisciplinary Association for Psychedelic Studies. FDA Grants MAPS Breakthrough Therapy Designation for Phase 3 Trials of MDMA-Assisted Therapy for PTSD. Multidisciplinary Association for Psychedelic Studies. Newsletter: August 2017 Web site. https://maps​.org/2017​/08/16/fda-grants-maps-breakthrough-therapy-designation-for-phase-3-trials-of-mdma-assisted-psychotherapy-for-ptsd/. Published 2017. Accessed August 2022.
51.
An Act: Ballot Measure 109. In: Elections Division Secretary of State, ed: Oregon Secretary of State; 2020.
52.
Feduccia AA, Mithoefer MC. MDMA-assisted psychotherapy for PTSD: Are memory reconsolidation and fear extinction underlying mechanisms? Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2018;84(Pt A):221–228. [PubMed: 29524515]
53.
Carhart-Harris RL, Roseman L, Haijen E, et al. Psychedelics and the essential importance of context. J Psychopharmacol. 2018;32(7):725–731. [PubMed: 29446697]
54.
Averill LA, Abdallah CG. Investigational drugs for assisting psychotherapy for posttraumatic stress disorder (PTSD): emerging approaches and shifting paradigms in the era of psychedelic medicine. Expert Opinion on Investigational Drugs. 2022;31(2):133–137. [PubMed: 35188023]
55.
National Academies of Sciences Engineering and Medicine. In: Evaluation of the Department of Veterans Affairs Mental Health Services. Washington (DC): National Academies Press (US); 2018. [PubMed: 29738208]
56.
Gaynes BN. Identifying difficult-to-treat depression: differential diagnosis, subtypes, and comorbidities. J Clin Psychiatry. 2009;70 Suppl 6:10–15. [PubMed: 19922739]
57.
Goldberg SB, Shechet B, Nicholas CR, et al. Post-acute psychological effects of classical serotonergic psychedelics: a systematic review and meta-analysis. Psychological Medicine. 2020;50(16):2655–2666. [PMC free article: PMC7855004] [PubMed: 33143790]
58.
Wheeler SW, Dyer NL. A systematic review of psychedelic-assisted psychotherapy for mental health: An evaluation of the current wave of research and suggestions for the future. Psychology of Consciousness: Theory, Research, and Practice. 2020;7(3):279–315.
59.
Romeo B, Karila L, Martelli C, Benyamina A. Efficacy of psychedelic treatments on depressive symptoms: A meta-analysis. Journal of Psychopharmacology. 2020;34(10):1079–1085. [PubMed: 32448048]
60.
Diagnostic and Statistical Manual of Mental Disorders. 4th ed. Washington D.C.: American Psychiatric Association; 1994.
61.
Sterne JA, Hernán MA, Reeves BC, et al. ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ. 2016;355:i4919. [PMC free article: PMC5062054] [PubMed: 27733354]
62.
Sterne JAC, Savović J, Page MJ, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ. 2019;366:l4898. [PubMed: 31462531]
63.
Berkman ND, Lohr KN, Ansari M, et al. AHRQ Methods for Effective Health Care Grading the Strength of a Body of Evidence When Assessing Health Care Interventions for the Effective Health Care Program of the Agency for Healthcare Research and Quality: An Update. In: Methods Guide for Effectiveness and Comparative Effectiveness Reviews. Rockville (MD): Agency for Healthcare Research and Quality (US); 2008. [PubMed: 24404627]
64.
Borenstein M, Higgins JP, Hedges LV, Rothstein HR. Basics of meta-analysis: I(2) is not an absolute measure of heterogeneity. Res Synth Methods. 2017;8(1):5–18. [PubMed: 28058794]
65.
Parr NJ, Schweer-Collins ML, Darlington TM, Tanner-Smith EE. Meta-analytic approaches for examining complexity and heterogeneity in studies of adolescent development. J Adolesc. 2019;77:168–178. [PMC free article: PMC6934259] [PubMed: 31739275]
66.
Knapp G, Hartung J. Improved tests for a random effects meta-regression with a single covariate. Stat Med. 2003;22(17):2693–2710. [PubMed: 12939780]
67.
Viechtbauer W. metafor: Meta-analysis package for R. The Comprehensive R Archive Network. https://cran​.r-project​.org/web/packages/metafor/index.html. Published 2020. Accessed August 2022.
68.
Mithoefer MC, Feduccia AA, Jerome L, et al. MDMA-assisted psychotherapy for treatment of PTSD: study design and rationale for phase 3 trials based on pooled analysis of six phase 2 randomized controlled trials. Psychopharmacology. 2019;236(9):2735–2745. [PMC free article: PMC6695343] [PubMed: 31065731]
69.
Bouso JC, Doblin R, Farre M, Alcazar MA, Gomez-Jarabo G. MDMA-assisted psychotherapy using low doses in a small sample of women with chronic posttraumatic stress disorder. Journal of Psychoactive Drugs. 2008;40(3):225–236. [PubMed: 19004414]
70.
Gukasyan N, Davis AK, Barrett FS, et al. Efficacy and safety of psilocybin-assisted treatment for major depressive disorder: Prospective 12-month follow-up. Journal of Psychopharmacology. 2022;36(2):151–158. [PMC free article: PMC8864328] [PubMed: 35166158]
71.
Mash DC, Kovera CA, Pablo J, et al. Ibogaine in the treatment of heroin withdrawal. The Alkaloids Chemistry & Biology. 2001;56:155–171. [PubMed: 11705106]
72.
Weathers F, Keane T, Davidson J. Clinician-Administered PTSD Scale: A Review of the First Ten Years of Research. Depression & Anxiety. 2001;13:132–156. [PubMed: 11387733]

Supplemental Materials

Download Supplemental Materials PDF

APPENDIX A. SEARCH STRATEGY

SYSTEMATIC REVIEWS

Search for current systematic reviews (limited to last 7 years)

Date Searched: 04-26-22

A. Bibliographic Databases:#Search StatementResults

MEDLINE: Systematic Reviews

Ovid MEDLINE(R) ALL 1946 to April 25, 2022

1 exp Hallucinogens/ad, ae, pd, tu, th, to [Administration & Dosage, Adverse Effects, Pharmacology, Therapeutic Use, Therapy, Toxicity]17439
2 (MDMA or LSD or DMT or ayahuasca or psilocybin or psychedelic*).ti,ab.14393
3 exp Mental Disorders/dt, pc, rh, tu, th [Drug Therapy, Prevention & Control, Rehabilitation, Therapeutic Use, Therapy]450492
4 exp Cannabinoids/ or exp Cannabis/24987
5 1 or 226925
6 3 and 51813
7 6 not 41468
8 limit 7 to (english language and humans)1078
9 (systematic review.ti. or meta-analysis.pt. or meta-analysis.ti. or systematic literature review.ti. or this systematic review.tw. or pooling project.tw. or (systematic review.ti,ab. and review.pt.) or meta synthesis.ti. or meta-analy*.ti. or integrative review.tw. or integrative research review.tw. or rapid review.tw. or umbrella review.tw. or consensus development conference.pt. or practice guideline.pt. or drug class reviews.ti. or cochrane database syst rev.jn. or acp journal club.jn. or health technol assess.jn. or evid rep technol assess summ.jn. or jbi database system rev implement rep.jn. or (clinical guideline and management).tw. or ((evidence based.ti. or evidence-based medicine/ or best practice*.ti. or evidence synthesis.ti,ab.) and (((review.pt. or diseases category/ or behavior.mp.) and behavior mechanisms/) or therapeutics/ or evaluation studies.pt. or validation studies.pt. or guideline.pt. or pmcbook.mp.)) or (((systematic or systematically).tw. or critical.ti,ab. or study selection.tw. or ((predetermined or inclusion) and criteri*).tw. or exclusion criteri*.tw. or main outcome measures.tw. or standard of care.tw. or standards of care.tw.) and ((survey or surveys).ti,ab. or overview*.tw. or review.ti,ab. or reviews.ti,ab. or search*.tw. or handsearch.tw. or analysis.ti. or critique.ti,ab. or appraisal.tw. or (reduction.tw. and (risk/ or risk.tw.) and (death or recurrence).mp.)) and ((literature or articles or publications or publication or bibliography or bibliographies or published).ti,ab. or pooled data.tw. or unpublished.tw. or citation.tw. or citations.tw. or database.ti,ab. or internet.ti,ab. or textbooks.ti,ab. or references.tw. or scales.tw. or papers.tw. or datasets.tw. or trials.ti,ab. or meta-analy*.tw. or (clinical and studies).ti,ab. or treatment outcome/ or treatment outcome.tw. or pmcbook.mp.))) not (letter or newspaper article).pt. 514028
10 8 AND 9 47

CDSR: Protocols and Reviews

EBM Reviews - Cochrane Database of Systematic Reviews 2005 to April 20, 2022

1hallucinogen*.ti,ab.1
2(MDMA or LSD or DMT or ayahuasca or psilocybin or psychedelic*).ti,ab.7
3mental disorder.ti,ab.16
4Cannabinoids or Cannabis.ti,ab.57
51 or 28
63 and 50
76 not 40
8limit 7 to (english language and humans)0

Search for current systematic reviews (limited to last 7 years)

Date Searched: 04-26-22

B. Non-bibliographic databases Evidence Results
AHRQ: evidence reports, technology assessments, U.S Preventative Services Task Force Evidence Synthesis

http://www​.ahrq.gov/research​/findings/evidence-based-reports/search.html

Search: (mental disorders OR substance use) AND (MDMA or LSD or DMT or ayahuasca or psilocybin or psychedelic)

AHRQ. 2020. Pharmacologic and Nonpharmacologic Treatments for Posttraumatic Stress Disorder: An Update of the PTSD Repository Evidence Base. https:​//effectivehealthcare​.ahrq.gov/products​/ptsd-repository-update/protocol

AHRQ. 2020. Pharmacologic and Nonpharmacologic Treatments for Posttraumatic Stress Disorder. https:​//effectivehealthcare​.ahrq.gov/products​/ptsd-repository-expanded/research

2
CADTH

https://www​.cadth.ca

Search: (mental disorders OR substance use) AND (MDMA or LSD or DMT or ayahuasca or psilocybin or psychedelic)

CADTH. 2021. Psychedelic-Assisted Psychotherapy for Post-Traumatic Stress Disorder, Anxiety Disorders, Mood Disorders, or Substance Use Disorders. https://www​.cadth.ca​/psychedelic-assisted-psychotherapy-post-traumatic-stress-disorder-anxiety-disorders-mood-disorders

1
ECRI Institute

https://guidelines​.ecri.org/

Search: (mental disorders OR substance use) AND (MDMA or LSD or DMT or ayahuasca or psilocybin or psychedelic)

0
HTA: Health Technology Assessments (UP TO 2016)

http://www​.ohsu.edu/xd​/education/library/

See CDSR search above

0
EPPI-Centre

http://eppi​.ioe.ac.uk/cms/Default​.aspx?tabid=62

Use browser search function [CNTL + F] for keyword search

Search: MDMA or LSD or DMT or ayahuasca or psilocybin or psychedelic

0
NLM

http://www​.ncbi.nlm.nih.gov/books

Search: (mental disorders OR substance use) AND (MDMA or LSD or DMT or ayahuasca or psilocybin or psychedelic)

0
VA Products - VATAP, PBM and HSR&D publications

A. http://www​.hsrd.research​.va.gov/research/default.cfm

Search: (mental disorders OR substance use) AND (MDMA or LSD or DMT or ayahuasca or psilocybin or psychedelic)

0

PRIMARY STUDIES

Search for primary literature

Date searched: 04-26-22

MEDLINE [Ovid MEDLINE(R) ALL 1946 to April 25, 2022]
#Search StatementResults
1exp Hallucinogens/ad, ae, pd, tu, th, to [Administration & Dosage, Adverse Effects, Pharmacology, Therapeutic Use, Therapy, Toxicity]17439
2(MDMA or LSD or DMT or ayahuasca or psilocybin or psychedelic*).ti,ab.14393
3exp Mental Disorders/dt, pc, rh, tu, th [Drug Therapy, Prevention & Control, Rehabilitation, Therapeutic Use, Therapy]450492
4exp Cannabinoids/ or exp Cannabis/24987
51 or 226925
63 and 51813
76 not 41468
8limit 7 to (english language and humans)1078
PsycINFO [APA PsycInfo 1806 to April Week 3 2022]
#Search StatementResults
1exp Hallucinogenic Drugs/4238
2(MDMA or LSD or DMT or ayahuasca or psilocybin or psychedelic*).ti,ab.6788
3exp Mental Disorders/929175
4exp Cannabinoids/ or exp Cannabis/15155
51 or 28737
63 and 52513
76 not 42333
8limit 7 to (english language and humans)1728

APPENDIX B. EXCLUDED STUDIES

Exclude reasons: 1=Ineligible population, 2=Ineligible intervention, 3=Ineligible comparator, 4=Ineligible outcome, 5=Ineligible timing, 6=Ineligible study design, 7=Ineligible publication type, 8=Outdated or ineligible systematic review, 9=non-English language, 10=unable to retrieve full-text, 11=study published before 1994.

CitationExclude Reason
Abramson HA. LSD in psychotherapy and alcoholism. American Journal of Psychotherapy. 1966;20(3):415–438. [PubMed: 5337647] E7
Abramson HA. Lysergic acid diethylamide (LSD 25). XXXXI. The use of LSD as an adjunct to psychotherapy: fact and fiction. Journal of Asthma Research. 1973;10(4):227–235. [PubMed: 4744918] E7
Abuzzahab FS, Sr., Anderson BJ. A review of LSD treatment in alcoholism. International Pharmacopsychiatry. 1971;6(4):223–235. [PubMed: 4950565] E7
Aday JS, Mitzkovitz CM, Bloesch EK, Davoli CC, Davis AK. Long-term effects of psychedelic drugs: A systematic review. Neuroscience and Biobehavioral Reviews. 2020;113:179–189. [PubMed: 32194129] E8
Agin-Liebes G. The role of self-compassion in psilocybin-assisted motivational enhancement therapy to treat alcohol dependence: A randomized controlled trial. Dissertation Abstracts International: Section B: The Sciences and Engineering. 2021;82(9-B). E10
Albaugh BJ, Anderson PO. Peyote in the treatment of alcoholism among American Indians. American Journal of Psychiatry. 1974;131(11):1247–1250. [PubMed: 4424469] E7
Alexander L. Mind and body in biological psychiatry. Biological Psychiatry. 1973;5(3):225–238. [PubMed: 4708678] E10
Almond K, Allan R. Incorporating MDMA as an adjunct in emotionally focused couples therapy with clients impacted by trauma or PTSD. The Family Journal. 2019;27(3):293–299. E7
Alper KR. Ibogaine: a review. The Alkaloids Chemistry & Biology. 2001;56:1–38. [PubMed: 11705103] E7
Alper KR, Glick SD. Psychotropic complementary medicines. British Journal of Psychiatry. 2006;188:587; author reply 587-588. [PubMed: 16738355] E7
Alper KR, Lotsof HS, Frenken GM, Luciano DJ, Bastiaans J. Treatment of acute opioid withdrawal with ibogaine. American Journal on Addictions. 1999;8(3):234–242. [PubMed: 10506904] E11
Amoroso T. The Psychopharmacology of +/-3,4 Methylenedioxymethamphetamine and its Role in the Treatment of Posttraumatic Stress Disorder. Journal of Psychoactive Drugs. 2015;47(5):337–344. [PubMed: 26579955] E7
Amoroso T, Workman M. Treating posttraumatic stress disorder with MDMA-assisted psychotherapy: A preliminary meta-analysis and comparison to prolonged exposure therapy. Journal of Psychopharmacology. 2016;30(7):595–600. [PubMed: 27118529] E8
Andersen KAA, Carhart-Harris R, Nutt DJ, Erritzoe D. Therapeutic effects of classic serotonergic psychedelics: A systematic review of modern-era clinical studies. Acta Psychiatrica Scandinavica. 2021;143(2):101–118. [PubMed: 33125716] E8
Anonymous. LSD: the search for definite conclusions. JAMA. 1966;196(4):Suppl:32–33. [PubMed: 5952211] E7
Anonymous. Dependence on LSD and other hallucinogenic drugs. JAMA. 1967;202(1):141–144. [PubMed: 6072003] E7
Anonymous. Preventing misuse of L.S.D. Lancet. 1967;1(7483):202. [PubMed: 4163134] E7
Anonymous. Control of amphetamines and L.S.D. Lancet. 1970;1(7649):708. [PubMed: 4191006] E7
Anonymous. L.S.D. in psychiatry. Lancet. 1970;2(7678):877–878. [PubMed: 4097784] E7
Anonymous. Treatment of alcoholism. II. Maryland State Medical Journal. 1973;22(4):17–19. [PubMed: 4785592] E7
Anonymous. Think harder about ecstasy. Nature. 2004;429(6988):113. [PubMed: 15141170] E7
Anonymous. Magic mushroom compound is a potential treatment for patients with major depression. Nursing Standard. 2016;30(41):15. [PubMed: 27286599] E7
Apud Pelaez IE. Personality Traits in Former Spanish Substance Users Recovered with Ayahuasca. Journal of Psychoactive Drugs. 2020;52(3):264–272. [PubMed: 32362241] E4
Argento E, Capler R, Thomas G, Lucas P, Tupper KW. Exploring ayahuasca-assisted therapy for addiction: A qualitative analysis of preliminary findings among an Indigenous community in Canada. Drug & Alcohol Review. 2019;38(7):781–789. [PubMed: 31489731] E6
Argento E, Tupper KW, Socias ME. The tripping point: The potential role of psychedelic-assisted therapy in the response to the opioid crisis. International Journal of Drug Policy. 2019;66:80–81. [PubMed: 30743091] E7
Avancena ALV, Kahn JG, Marseille E. The Costs and Health Benefits of Expanded Access to MDMA-assisted Therapy for Chronic and Severe PTSD in the USA: A Modeling Study. Clinical Drug Investigation. 2022;42(3):243–252. [PMC free article: PMC8930905] [PubMed: 35230652] E6
Averill LA, Abdallah CG. Investigational drugs for assisting psychotherapy for posttraumatic stress disorder (PTSD): emerging approaches and shifting paradigms in the era of psychedelic medicine. Expert Opinion on Investigational Drugs. 2022;31(2):133–137. [PubMed: 35188023] E7
Bahji A, Forsyth A, Groll D, Hawken ER. Efficacy of 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy for posttraumatic stress disorder: A systematic review and meta-analysis. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2020;96:109735. [PubMed: 31437480] E8
Barabasz-Gembczyk A, Kucia K. Ayahuasca - potential therapeutic properties in psychiatry. Research review. Psychiatria Polska. 2020;54(2):381–389. [PubMed: 32772067] E7
Barker ET, Buck MF. LSD in a coercive milieu therapy program. Canadian Psychiatric Association Journal. 1977;22(6):311–314. [PubMed: 922641] E4
Barnett BS, Greer GR. Psychedelic psychiatry and the consult-liaison psychiatrist: A primer. Journal of the Academy of Consultation Liaison Psychiatry. 2021;62(4):460–471. [PubMed: 34210406] E7
Barrios AA. An explanation of the behavioral and therapeutic effects of the hallucinogens. International Journal of Neuropsychiatry. 1965;1(6):574–592. [PubMed: 5328850] E7
Basky G. Policy in focus: Is psilocybin the next cannabis? CMAJ Canadian Medical Association Journal. 2021;193(45):E1741–E1742. [PMC free article: PMC8594542] [PubMed: 34782381] E7
Bateman C. Ibogaine may help drug addicts live the ‘never again’. South African Medical Journal Suid-Afrikaanse Tydskrif Vir Geneeskunde. 2006;96(7):580, 582. [PubMed: 16909175] E7
Bedi G. 3,4-Methylenedioxymethamphetamine as a Psychiatric Treatment. JAMA Psychiatry. 2018;75(5):419–420. [PubMed: 29562086] E7
Bergman RL. Navajo peyote use: its apparent safety. American Journal of Psychiatry. 1971;128(6):695–699. [PubMed: 5147724] E1
Bienemann B, Ruschel NS, Campos ML, Negreiros MA, Mograbi DC. Self-reported negative outcomes of psilocybin users: A quantitative textual analysis. PLoS ONE. 2020;15(2). [PMC free article: PMC7034876] [PubMed: 32084160] E1
Bird CIV, Modlin NL, Rucker JJH. Psilocybin and MDMA for the treatment of trauma-related psychopathology. International Review of Psychiatry. 2021;33(3):229–249. [PubMed: 34121583] E7
Boer AP, Sipprelle CN. Induced anxiety in the treatment for LSD effects. Psychotherapy & Psychosomatics. 1969;17(2):108–113. [PubMed: 5404497] E6
Bogenschutz MP, Johnson MW. Classic hallucinogens in the treatment of addictions. Progress in Neuro-Psychopharmacology & Biological Psychiatry. 2016;64:250–258. [PubMed: 25784600] E7
Bonny HL, Pahnke WN. The use of music in psychedelic (LSD) psychotherapy. Journal of Music Therapy. 1972;9(2):64–87. E7
Bouso JC, Ryan C. Using MDMA in the treatment of post-traumatic stress disorder. Holland, Julie [Ed] (2001). 2001;Ecstasy:The complete guide: A comprehensive look at the risks and benefits of MDMA. (pp. 248–260). x. E7
Bowen WT, Soskin RA, Chotlos JW. Lysergic acid diethylamide as a variable in the hospital treatment of alcoholism. Journal of Nervous and Mental Disease. 1970;150(2):111–118. [PubMed: 4906920] E11
Bramble FB. Dependency needs in chronic hallucinogenic drug abusers. Dissertation Abstracts International. 1974;34(7-B). E4
Breeksema JJ, Niemeijer AR, Krediet E, Vermetten E, Schoevers RA. Psychedelic Treatments for Psychiatric Disorders: A Systematic Review and Thematic Synthesis of Patient Experiences in Qualitative Studies. CNS Drugs. 2020;34(9):925–946. [PMC free article: PMC7447679] [PubMed: 32803732] E6
Brewerton TD, Lafrance A, Mithoefer MC. The potential use of N-methyl-3,4-methylenedioxyamphetamine (MDMA) assisted psychotherapy in the treatment of eating disorders comorbid with PTSD. Medical Hypotheses. 2021;146:110367. [PubMed: 33203569] E6
Brewerton TD, Wang JB, Lafrance A, et al. MDMA-assisted therapy significantly reduces eating disorder symptoms in a randomized placebo-controlled trial of adults with severe PTSD. Journal of Psychiatric Research. 2022;149:128–135. [PubMed: 35272210] E4
Brown TK, Noller GE, Denenberg JO. Ibogaine and Subjective Experience: Transformative States and Psychopharmacotherapy in the Treatment of Opioid Use Disorder. Journal of Psychoactive Drugs. 2019;51(2):155–165. [PubMed: 30967101] E4
Bryce JC. An evaluation of LSD in the treatment of chronic alcoholism. Canadian Psychiatric Association Journal. 1970;15(1):77–78. [PubMed: 5415468] E7
Buckman J. Theoretical aspects of L.S.D. therapy. International Journal of Social Psychiatry. 1967;13(2):126–138. [PubMed: 4167447] E7
Carbonaro TM, Gatch MB. Neuropharmacology of N,N-dimethyltryptamine. Brain Research Bulletin. 2016;126(Pt 1):74–88. [PMC free article: PMC5048497] [PubMed: 27126737] E7
Chabrol H. MDMA assisted psychotherapy found to have a large effect for chronic post-traumatic stress disorder. Journal of Psychopharmacology. 2013;27(9):865–866. [PubMed: 23981728] E7
Chang AF, Caldwell AB, Moss T. The stability of personality traits in alcoholics during and after treatment as measured by the MMPI: A one year follow-up study. Proceedings of the Annual Convention of the American Psychological Association. 1973:389–390. E11
Check E. Psychedelic drugs: the ups and downs of ecstasy. Nature. 2004;429(6988):126–128. [PubMed: 15141183] E7
Cheek FE, Osmond H, Sarett M. Observations regarding the use of LSD-25 in the treatment of alcoholism. Journal of Psychopharmacology. 1966;1(2):56–74. E10
Chessick RD, Haertzen CA, Wikler A. Tolerance to LSD-25 in schizophrenic subjects. Archives of General Psychiatry. 1964;10(6):653–658. [PubMed: 14159265] E4
Chi T, Gold JA. A review of emerging therapeutic potential of psychedelic drugs in the treatment of psychiatric illnesses. Journal of the Neurological Sciences. 2020;411:116715. [PubMed: 32044687] E7
Cholden LS, Kurland A, Savage C. Clinical reactions and tolerance to LSD in chronic schizophrenia. Journal of Nervous & Mental Disease. 1955;122(3):211–221. [PubMed: 13295823] E1
Cipriani A, Cowen PJ. 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy for post-traumatic stress disorder in service personnel. The Lancet Psychiatry. 2018;5(6):453–455. [PubMed: 29728332] E7
Cline HS, Freeman H. Resistance to lysergic acid in schizophrenic patients. Psychiatric Quarterly. 1956;30(4):676–683. [PubMed: 13400977] E11
Clyman RC. LSD psychotherapy: a review of the literature and some proposals for future research. Rhode Island Medical Journal. 1972;55(9):282–286. [PubMed: 4561308] E7
Cohen RS. Adverse symptomatology and suicide associated with the use of methylenedioxymethamphetamine (MDMA; “Ecstasy”). Biological Psychiatry. 1996;39(9):819–820. [PubMed: 8731525] E1
Corey VR, Pisano VD, Halpern JH. Effects of 3,4-Methylenedioxymethamphetamine on Patient Utterances in a Psychotherapeutic Setting. Journal of Nervous & Mental Disease. 2016;204(7):519–523. [PubMed: 26998697] E4
Corkery JM. Ibogaine as a treatment for substance misuse: Potential benefits and practical dangers. Progress in Brain Research. 2018;242:217–257. [PubMed: 30471681] E7
Cox DJ, Garcia-Romeu A, Johnson MW. Predicting changes in substance use following psychedelic experiences: natural language processing of psychedelic session narratives. American Journal of Drug & Alcohol Abuse. 2021;47(4):444–454. [PubMed: 34096403] E10
Crowley N. A role for psychedelics in psychiatry? British Journal of Psychiatry. 2005;187:483; author reply 484-485. [PubMed: 16260829] E7
Curran HV, Nutt D, de Wit H. Psychedelics and related drugs: therapeutic possibilities, mechanisms and regulation. Psychopharmacology. 2018;235(2):373–375. [PubMed: 29445838] E7
Curry AE. Elixir of anguish: The phenomenology of the alcoholic’s perennial quest. Psychiatric Quarterly Supplement. 1964;38(1):13–20. [PubMed: 14297898] E4
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. 2018;235(11):3137–3148. [PMC free article: PMC6208958] [PubMed: 30196397] E1
Danforth AL, Struble CM, Yazar-Klosinski B, Grob CS. MDMA-assisted therapy: A new treatment model for social anxiety in autistic adults. Progress in NeuroPsychopharmacology & Biological Psychiatry. 2016;64:237–249. [PubMed: 25818246] E7
Davey CG. Out of the night-time and into the day: Ketamine and MDMA as therapies for mental disorders. Australian & New Zealand Journal of Psychiatry. 2021;55(8):741–743. [PubMed: 34311555] E7
Davis AK, Barrett FS, Griffiths RR. Psychological flexibility mediates the relations between acute psychedelic effects and subjective decreases in depression and anxiety. Journal of Contextual Behavioral Science. 2020;15:39–45. [PMC free article: PMC7451132] [PubMed: 32864325] E1
Davis AK, Griffiths RR. Errors in a response rate and in effect sizes in study of psilocybin-assisted therapy for major depressive disorder. JAMA Psychiatry. 2021;78(5):569. [PubMed: 33566073] E7
Davis AK, Renn E, Windham-Herman AM, Polanco M, Barsuglia JP. A Mixed-Method Analysis of Persisting Effects Associated with Positive Outcomes Following Ibogaine Detoxification. Journal of Psychoactive Drugs. 2018;50(4):287–297. [PMC free article: PMC6247813] [PubMed: 30020025] E4
Davis AK, So S, Lancelotta R, Barsuglia JP, Griffiths RR. 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) used in a naturalistic group setting is associated with unintended improvements in depression and anxiety. American Journal of Drug & Alcohol Abuse. 2019;45(2):161–169. [PMC free article: PMC6430661] [PubMed: 30822141] E1
Daws RE, Timmermann C, Giribaldi B, et al. Increased global integration in the brain after psilocybin therapy for depression. Nature Medicine. 2022;28(4):844–851. [PubMed: 35411074] E4
Day J. The role and reaction of the psychiatrist in LSD therapy. Journal of Nervous & Mental Disease. 1957;125(3):437–438. [PubMed: 13481751] E7
de Almeida RN, de Menezes Galvao AC, da Silva FS, et al. Modulation of serum brain-derived neurotrophic factor by a single dose of ayahuasca: Observation from a randomized controlled trial. Frontiers in Psychology. 2019;10. [PMC free article: PMC6558429] [PubMed: 31231276] E4
De Gregorio D, Aguilar-Valles A, Preller KH, et al. Hallucinogens in Mental Health: Preclinical and Clinical Studies on LSD, Psilocybin, MDMA, and Ketamine. Journal of Neuroscience. 2021;41(5):891–900. [PMC free article: PMC7880300] [PubMed: 33257322] E7
de Kleine RA, Rothbaum BO, van Minnen A. Pharmacological enhancement of exposure-based treatment in PTSD: A qualitative review. European Journal of Psychotraumatology. 2013;4:21626. [PMC free article: PMC3800126] [PubMed: 24147208] E8
de Veen BT, Schellekens AF, Verheij MM, Homberg JR. Psilocybin for treating substance use disorders? Expert Review of Neurotherapeutics. 2017;17(2):203–212. [PubMed: 27684102] E7
Denber HC. Studies with mescaline. Rivista di Neurobiologia. 1964;10(4):Suppl:1157–1168. [PubMed: 5827093] E7
Denber HC. Mescaline and lysergic acid diethylamide: therapeutic implications of the drug-induced state. Diseases of the Nervous System. 1969;30(2):Suppl:23–27. [PubMed: 5775311] E10
Denson R. Dissociative delirium after treatment with lysergide. Canadian Medical Association Journal. 1967;97(20):1222–1224. [PMC free article: PMC1923613] [PubMed: 6054295] E6
Denson R. Lysergide therapy and the mauve factor. Acta Psychiatrica Scandinavica. 1968;44(3):280–288. [PubMed: 5722604] E11
Denson R. Complications of therapy with lysergide. Canadian Medical Association Journal. 1969;101(11):53–57. [PMC free article: PMC1946366] [PubMed: 5353149] E11
Denson R, Sydiaha D. A controlled study of LSD treatment in alcoholism and neurosis. British Journal of Psychiatry. 1970;116(533):443–445. [PubMed: 4392561] E1
Ditman KS, et al. Psychological and drug variables in the LSD experiences of alcoholics. Psychotherapy and Psychosomatics. 1967;15(1):15. E10
DiVito AJ, Leger RF. Psychedelics as an emerging novel intervention in the treatment of substance use disorder: a review. Molecular Biology Reports. 2020;47(12):9791–9799. [PubMed: 33231817] E7
Dominguez-Clave E, Soler J, Pascual JC, et al. Ayahuasca improves emotion dysregulation in a community sample and in individuals with borderline-like traits. Psychopharmacology. 2019;236(2):573–580. [PubMed: 30406413] E1
Donnelly JR. The need for ibogaine in drug and alcohol addiction treatment. Journal of Legal Medicine. 2011;32(1):93–114. [PubMed: 21391058] E7
Dos Santos RG, Balthazar FM, Bouso JC, Hallak JE. The current state of research on ayahuasca: A systematic review of human studies assessing psychiatric symptoms, neuropsychological functioning, and neuroimaging. Journal of Psychopharmacology. 2016;30(12):1230–1247. [PubMed: 27287824] E1
Dos Santos RG, Bouso JC, Alcazar-Corcoles MA, Hallak JEC. Efficacy, tolerability, and safety of serotonergic psychedelics for the management of mood, anxiety, and substance-use disorders: a systematic review of systematic reviews. Expert Review of Clinical Pharmacology. 2018;11(9):889–902. [PubMed: 30102078] E8
Dos Santos RG, Hallak JE, Baker G, Dursun S. Hallucinogenic/psychedelic 5HT2A receptor agonists as rapid antidepressant therapeutics: Evidence and mechanisms of action. Journal of Psychopharmacology. 2021;35(4):453–458. [PubMed: 33740877] E7
Dos Santos RG, Osorio FL, Crippa JA, Hallak JE. Antidepressive and anxiolytic effects of ayahuasca: a systematic literature review of animal and human studies. Revista Brasileira de Psiquiatria. 2016;38(1):65–72. [PMC free article: PMC7115465] [PubMed: 27111702] E8
dos Santos RG, Osorio FL, Crippa JAS, Bouso JC, Hallak JEC. The therapeutic potential of ayahuasca and other serotonergic hallucinogens in the treatment of social anxiety. Osorio, Flavia de Lima [Ed]; Donadon, Mariana Fortunata [Ed] (2018). 2018;Social anxiety disorder:Recognition. E7
Doss MK, Povazan M, Rosenberg MD, et al. Psilocybin therapy increases cognitive and neural flexibility in patients with major depressive disorder. Transl Psychiatry Psychiatry. 2021;11(1):574. [PMC free article: PMC8575795] [PubMed: 34750350] E4
Eagle CT. Music and LSD: An empirical study. Journal of Music Therapy. 1972;9(1):23–36. E6
Erritzoe D, Roseman L, Nour MM, et al. Effects of psilocybin therapy on personality structure. Acta Psychiatrica Scandinavica. 2018;138(5):368–378. [PMC free article: PMC6220878] [PubMed: 29923178] E4
Fadiman J, Korb S. Might microdosing psychedelics be safe and beneficial? An initial exploration. Journal of Psychoactive Drugs. 2019;51(2):118–122. [PubMed: 30925850] E6
Faillace LA. Clinical use of psychotomimetic drugs. Comprehensive Psychiatry. 1966;7(1):13–20. [PubMed: 5322648] E7
Faillace LA, Vourlekis A, Szara S. Clinical evaluation of some hallucinogenic tryptamine derivatives. Journal of Nervous & Mental Disease. 1967;145(4):306–313. [PubMed: 6076017] E4
Faillace LA, Vourlekis A, Szara S. Hallucinogenic drugs in the treatment of alcoholism: a two-year follow-up. Comprehensive Psychiatry. 1970;11(1):51–56. [PubMed: 5411214] E11
Fauvel B, Strika-Bruneau L, Piolino P. Changes in self-rumination and self-compassion mediate the effect of psychedelic experiences on decreases in depression, anxiety, and stress. Psychology of Consciousness: Theory, Research, and Practice. 2021(Pagination). E1
Feduccia AA, Jerome L, Mithoefer MC, Holland J. Discontinuation of medications classified as reuptake inhibitors affects treatment response of MDMA-assisted psychotherapy. Psychopharmacology. 2021;238(2):581–588. [PMC free article: PMC7826309] [PubMed: 33221932] E4
Feduccia AA, Jerome L, Yazar-Klosinski B, Emerson A, Mithoefer MC, Doblin R. Breakthrough for trauma treatment: Safety and efficacy of mdma-assisted psychotherapy compared to paroxetine and sertraline. Frontiers in Psychiatry. 2019;10:650. [PMC free article: PMC6751381] [PubMed: 31572236] E8
Fink M, Simeon J, Haque W, Itil T. Prolonged adverse reactions to LSD in psychotic subjects. Archives of General Psychiatry. 1966;15(5):450–454. [PubMed: 5926592] E6
Frenken G. From the roots up: ibogaine and addict self-help. The Alkaloids Chemistry & Biology. 2001;56:283–292. [PubMed: 11705113] E7
Fuentes JJ, Fonseca F, Elices M, Farre M, Torrens M. Therapeutic use of LSD in psychiatry: A systematic review of randomized-controlled clinical trials. Frontiers in Psychiatry. 2020;10:943. [PMC free article: PMC6985449] [PubMed: 32038315] E8
Galvao-Coelho NL, Marx W, Gonzalez M, et al. Classic serotonergic psychedelics for mood and depressive symptoms: a meta-analysis of mood disorder patients and healthy participants. Psychopharmacology. 2021;238(2):341–354. [PMC free article: PMC7826317] [PubMed: 33427944] E8
Garcia-Romeu A, Barrett FS, Carbonaro TM, Johnson MW, Griffiths RR. Optimal dosing for psilocybin pharmacotherapy: Considering weight-adjusted and fixed dosing approaches. Journal of Psychopharmacology. 2021;35(4):353–361. [PMC free article: PMC8056712] [PubMed: 33611977] E4
Garcia-Romeu A, Davis AK, Erowid F, Erowid E, Griffiths RR, Johnson MW. Cessation and reduction in alcohol consumption and misuse after psychedelic use. Journal of Psychopharmacology. 2019;33(9):1088–1101. [PubMed: 31084460] E2
Garcia-Romeu A, Griffiths RR, Johnson MW. Psilocybin-occasioned mystical experiences in the treatment of tobacco addiction. Current Drug Abuse Reviews. 2014;7(3):157–164. [PMC free article: PMC4342293] [PubMed: 25563443] E4
Gilbert CS, Earleywine M, Mian MN, Altman BR. Symptom specificity of ayahuasca’s effect on depressive symptoms. Journal of Psychedelic Studies. 2021;5(1):37–43. E1
Gill H, Gill B, Chen-Li D, et al. The emerging role of psilocybin and MDMA in the treatment of mental illness. Expert Review of Neurotherapeutics. 2020;20(12):1263–1273. [PubMed: 32954860] E7
Giovannetti C, Garcia Arce S, Rush B, Mendive F. Pilot Evaluation of a Residential Drug Addiction Treatment Combining Traditional Amazonian Medicine, Ayahuasca and Psychotherapy on Depression and Anxiety. Journal of Psychoactive Drugs. 2020;52(5):472–481. [PubMed: 32748709] E4
Goldberg SB, Pace BT, Nicholas CR, Raison CL, Hutson PR. The experimental effects of psilocybin on symptoms of anxiety and depression: A meta-analysis. Psychiatry Research. 2020;284:112749. [PubMed: 31931272] E8
Goldberg SB, Shechet B, Nicholas CR, et al. Post-acute psychological effects of classical serotonergic psychedelics: a systematic review and meta-analysis. Psychological Medicine. 2020;50(16):2655–2666. [PMC free article: PMC7855004] [PubMed: 33143790] E8
Gomez-Busto FJ, Ortiz MI. Virtual reality and psychedelics for the treatment of psychiatric disease: A systematic literature review. Clinical Neuropsychiatry: Journal of Treatment Evaluation. 2020;17(6):365–380. [PMC free article: PMC8629068] [PubMed: 34909015] E8
Gorman I, Belser AB, Jerome L, et al. Posttraumatic Growth After MDMA-Assisted Psychotherapy for Posttraumatic Stress Disorder. Journal of Traumatic Stress. 2020;33(2):161–170. [PMC free article: PMC7216948] [PubMed: 32073177] E8
Greenway KT, Garel N, Jerome L, Feduccia AA. Integrating psychotherapy and psychopharmacology: psychedelic-assisted psychotherapy and other combined treatments. Expert Review of Clinical Pharmacology. 2020;13(6):655–670. [PubMed: 32478631] E7
Greer GR, Tolbert R. A method of conducting therapeutic sessions with MDMA. Journal of Psychoactive Drugs. 1998;30(4):371–379. [PubMed: 9924843] E6
Grinspoon L, Bakalar JB. The psychedelic drug therapies. Current Psychiatric Therapies. 1981;20:275–283. [PubMed: 7326971] E10
Grof S, Soskin RA, Richards WA, Kurland AA. DPT as an adjunct in psychotherapy of alcoholics. International Pharmacopsychiatry. 1973;8(1):104–115. [PubMed: 4150711] E11
Halpern JH, Sherwood AR, Hudson JI, Yurgelun-Todd D, Pope HG, Jr. Psychological and Cognitive Effects of Long-Term Peyote Use Among Native Americans. Biological Psychiatry. 2005;58(8):624–631. [PubMed: 16271313] E1
Hamill J, Hallak J, Dursun SM, Baker G. Ayahuasca: Psychological and Physiologic Effects, Pharmacology and Potential Uses in Addiction and Mental Illness. Current Neuropharmacology. 2019;17(2):108–128. [PMC free article: PMC6343205] [PubMed: 29366418] E7
Harris R, Gurel L. A study of ayahuasca use in North America. Journal of Psychoactive Drugs. 2012;44(3):209–215. [PubMed: 23061320] E1
Hatrick JA, Dewhurst K. Delayed psychosis due to L.S.D. Lancet. 1970;2(7676):742–744. [PubMed: 4195978] E1
Hausner M, Dolezal V. Follow-up studies in group and individual LSD psychotherapy. Activitas Nervosa Superior. 1966;8(1):87–95. [PubMed: 5913553] E1
Hausner M, Dolezal V. Follow-up evaluation of LSD psychotherapy of inpatients. Activitas Nervosa Superior. 1968;10(3):282–283. [PubMed: 5702527] E1
Heal DJ, Gosden J, Smith SL. Evaluating the abuse potential of psychedelic drugs as part of the safety pharmacology assessment for medical use in humans. Neuropharmacology. 2018;142:89–115. [PubMed: 29427652] E7
Heink A, Katsikas S, Lange-Altman T. Examination of the Phenomenology of the Ibogaine Treatment Experience: Role of Altered States of Consciousness and Psychedelic Experiences. Journal of Psychoactive Drugs. 2017;49(3):201–208. [PubMed: 28266890] E1
Heink AL. Examination of the phenomeonology of the ibogaine treatment experience on change: Role of altered states of consciousness and psychedelic experiences. Dissertation Abstracts International: Section B: The Sciences and Engineering. 2016;76(11-B(E). E10
Hendricks PS. Back to the future: a return to psychedelic treatment models for addiction. Journal of Psychopharmacology. 2014;28(11):981–982. [PubMed: 25253275] E7
Hendricks PS, Clark CB, Johnson MW, Fontaine KR, Cropsey KL. Hallucinogen use predicts reduced recidivism among substance-involved offenders under community corrections supervision. Journal of Psychopharmacology. 2014;28(1):62–66. [PubMed: 24399338] E4
Hendricks PS, Thorne CB, Clark CB, Coombs DW, Johnson MW. Classic psychedelic use is associated with reduced psychological distress and suicidality in the United States adult population. Journal of Psychopharmacology. 2015;29(3):280–288. [PubMed: 25586402] E1
Heuschkel K, Kuypers KPC. Depression, mindfulness, and psilocybin: Possible complementary effects of mindfulness meditation and psilocybin in the treatment of depression. A review. Frontiers in Psychiatry. 2020;11:224. [PMC free article: PMC7136554] [PubMed: 32296353] E8
Hittner JB, Quello SB. Combating substance abuse with ibogaine: pre- and posttreatment recommendations and an example of successive model fitting analyses. Journal of Psychoactive Drugs. 2004;36(2):191–199. [PubMed: 15369200] E6
Hoch PH. Remarks on LSD and mescaline. Journal of Nervous & Mental Disease. 1957;125(3):442–444. [PubMed: 13481753] E7
Hoelen DW, Spiering W, Valk GD. Long-QT syndrome induced by the antiaddiction drug ibogaine. New England Journal of Medicine. 2009;360(3):308–309. [PubMed: 19144953] E6
Hofling CK, Winslow WW, Stone WN. Drug therapy. Progress in Neurology & Psychiatry. 1969;24:478–497. [PubMed: 4905451] E7
Hollister LE, Shelton J, Krieger G. A controlled comparison of lysergic acid diethylamide (LSD) and dextroamphetmine in alcoholics. American Journal of Psychiatry. 1969;125(10):1352–1357. [PubMed: 4886242] E11
Holoyda B. Psychedelic Psychiatry: Preparing for Novel Treatments Involving Altered States of Consciousness. Psychiatric Services. 2020;71(12):1297–1299. [PubMed: 33050796] E7
Hosanagar A, Cusimano J, Radhakrishnan R. Therapeutic Potential of Psychedelics in Treatment of Psychiatric Disorders, Part 2: Review of the Evidence. Journal of Clinical Psychiatry. 2021;82(3):23. [PubMed: 33988930] E7
Hosanagar A, Cusimano J, Radhakrishnan R. Therapeutic Potential of Psychedelics in the Treatment of Psychiatric Disorders, Part 1: Psychopharmacology and Neurobiological Effects. Journal of Clinical Psychiatry. 2021;82(2):23. [PubMed: 33988926] E7
Hoskins MD, Sinnerton R, Nakamura A, et al. Pharmacological-assisted psychotherapy for post-traumatic stress disorder: A systematic review and meta-analysis. European Journal of Psychotraumatology. 2021;12(1):1853379. [PMC free article: PMC7874936] [PubMed: 33680344] E8
Hostiuc S, Buda O, Ion DA. Harmine for catatonic schizophrenia. A forgotten experiment. Schizophrenia Research. 2014;159(1):249–250. [PubMed: 25195064] E7
Hrdina PD, Bakish D, Ravindran A, Chudzik J, Cavazzoni P, Lapierre YD. Platelet serotonergic indices in major depression: up-regulation of 5-HT2A receptors unchanged by antidepressant treatment. Psychiatry Research. 1997;66(2–3):73–85. [PubMed: 9075272] E4
Huang DS. The efficacy of psychedelic-assisted psychotherapy. Dissertation Abstracts International: Section B: The Sciences and Engineering. 2020;81(12-B). E10
Hungerford DA, Taylor KM, Shagass C, LaBadie GU, Balaban GB, Paton GR. Cytogenetic effects of LSD 25 therapy in man. JAMA. 1968;206(10):2287–2291. [PubMed: 5696083] E4
Hutchison CA, Bressi SK. MDMA-assisted psychotherapy for posttraumatic stress disorder: Implications for social work practice and research. Clinical Social Work Journal. 2020;48(4):421–430. E7
Hutten NRPW, Mason NL, Dolder PC, Kuypers KPC. Self-rated effectiveness of microdosing with psychedelics for mental and physical health problems among microdosers. Frontiers in Psychiatry. 2019;10:672. [PMC free article: PMC6753862] [PubMed: 31572246] E2
Illingworth BJ, Lewis DJ, Lambarth AT, et al. A comparison of MDMA-assisted psychotherapy to non-assisted psychotherapy in treatment-resistant PTSD: A systematic review and meta-analysis. Journal of Psychopharmacology. 2021;35(5):501–511. [PubMed: 33345689] E8
Itil TM, Keskiner S, Holden JM. The use of LSD and ditran in the treatment of therapy resistant schizophrenics (symptom provocation approach). Diseases of the Nervous System. 1969;30(2):Suppl:93–103. [PubMed: 4388281] E4
Jerome L, Feduccia AA, Wang JB, et al. Long-term follow-up outcomes of MDMA-assisted psychotherapy for treatment of PTSD: a longitudinal pooled analysis of six phase 2 trials. Psychopharmacology. 2020;237(8):2485–2497. [PMC free article: PMC7351848] [PubMed: 32500209] E8
Jerome L, Schuster S, Yazar-Klosinski BB. Can MDMA play a role in the treatment of substance abuse? Current Drug Abuse Reviews. 2013;6(1):54–62. [PubMed: 23627786] E7
Johnson FG. LSD in the treatment of alcoholism. American Journal of Psychiatry. 1969;126(4):481–487. [PubMed: 5806791] E11
Johnson FG. A comparison of short-term treatment effects of intravenous sodium amytal-methedrine and LSD in the alcoholic. Canadian Psychiatric Association Journal. 1970;15(5):493–497. [PubMed: 5492189] E4
Johnson MW, Garcia-Romeu A, Johnson PS, Griffiths RR. An online survey of tobacco smoking cessation associated with naturalistic psychedelic use. Journal of Psychopharmacology. 2017;31(7):841–850. [PMC free article: PMC6753943] [PubMed: 28095732] E2
Johnson MW, Griffiths RR. Potential Therapeutic Effects of Psilocybin. Neurotherapeutics. 2017;14(3):734–740. [PMC free article: PMC5509636] [PubMed: 28585222] E7
Johnson MW, Griffiths RR, Hendricks PS, Henningfield JE. The abuse potential of medical psilocybin according to the 8 factors of the Controlled Substances Act. Neuropharmacology. 2018;142:143–166. [PMC free article: PMC6791528] [PubMed: 29753748] E7
Johnstad PG. A dangerous method? Psychedelic therapy at Modum Bad, Norway, 1961-76. History of Psychiatry. 2020;31(2):217–226. [PubMed: 31928087] E7
Jones G, Ricard JA, Lipson J, Nock MK. Associations between classic psychedelics and opioid use disorder in a nationally-representative U.S. adult sample. Scientific Reports. 2022;12(1):4099. [PMC free article: PMC8990065] [PubMed: 35393455] E2
Jones GM, Nock MK. Lifetime use of MDMA/ecstasy and psilocybin is associated with reduced odds of major depressive episodes. Journal of Psychopharmacology. 2022;36(1):57–65. [PubMed: 34983261] E2
Kadriu B, Greenwald M, Henter ID, et al. Ketamine and Serotonergic Psychedelics: Common Mechanisms Underlying the Effects of Rapid-Acting Antidepressants. International Journal of Neuropsychopharmacology. 2021;24(1):8–21. [PMC free article: PMC7816692] [PubMed: 33252694] E7
Kalechstein AD, De La Garza R, II, Mahoney JJ, III, Fantegrossi WE, Newton TF. MDMA use and neurocognition: A meta-analytic review. Psychopharmacology. 2007;189(4):531–537. [PubMed: 17082969] E1
Knuijver T, Belgers M, Markus W, Verkes RJ, van Oosteren T, Schellekens A. Hallucinogen Persisting Perception Disorder After Ibogaine Treatment for Opioid Dependence. Journal of Clinical Psychopharmacology. 2018;38(6):646–648. [PubMed: 30300292] E6
Krebs TS, Johansen P-O. Psychedelics and mental health: A population study. PLoS ONE. 2013;8(8). [PMC free article: PMC3747247] [PubMed: 23976938] E2
Krebs TS, Johansen PO. Lysergic acid diethylamide (LSD) for alcoholism: meta-analysis of randomized controlled trials. Journal of Psychopharmacology. 2012;26(7):994–1002. [PubMed: 22406913] E8
Krediet E, Bostoen T, Breeksema J, van Schagen A, Passie T, Vermetten E. Reviewing the Potential of Psychedelics for the Treatment of PTSD. International Journal of Neuropsychopharmacology. 2020;23(6):385–400. [PMC free article: PMC7311646] [PubMed: 32170326] E7
Krystal JH, Kelmendi B, Petrakis IL. Psychotherapy-supported MDMA treatment for PTSD. Cell Reports Medicine. 2021;2(8):100378. [PMC free article: PMC8385322] [PubMed: 34467253] E7
Kupferschmidt K. Can ecstasy treat the agony of PTSD? Science. 2014;345(6192):22–23. [PubMed: 24994631] E7
Kurland AA, Unger S, Shaffer JW, Savage C. Psychedelic therapy utilizing LSD in the treatment of the alcoholic patient: a preliminary report. American Journal of Psychiatry. 1967;123(10):1202–1209. [PubMed: 4381477] E4
Kverno KS, Mangano E. Treatment-Resistant Depression: Approaches to Treatment. Journal of Psychosocial Nursing & Mental Health Services. 2021;59(9):7–11. [PubMed: 34459676] E7
Lajoie M. Can ayahuasca enhance the treatment of addiction? A comprehensive literature review. Dissertation Abstracts International: Section B: The Sciences and Engineering. 2018;79(1-B(E). E10
Lappin JM, Sara GE. Psychostimulant use and the brain. Addiction. 2019;114(11):2065–2077. [PubMed: 31321819] E7
Larova V. The evolution of posttraumatic stress disorder treatment: Lessons learned from MDMA-assisted psychotherapy. Dissertation Abstracts International: Section B: The Sciences and Engineering. 2021;82(2-B). E8
Larsen JK. LSD treatment in Scandinavia: emphasizing indications and short-term treatment outcomes of 151 patients in Denmark. Nordic Journal of Psychiatry. 2017;71(7):489–495. [PubMed: 28678583] E1
Larsen JK. Early LSD treatment in Denmark from 1960 to 1974: An analysis of possible and long-lasting changes in the adult personality following psychedelic treatment. A historical retrospective cohort study. Medicine. 2021;100(23):e26300. [PMC free article: PMC8202585] [PubMed: 34115036] E1
Latimer D, Stocker MD, Sayers K, et al. MDMA to Treat PTSD in Adults. Psychopharmacology Bulletin. 2021;51(3):125–149. [PMC free article: PMC8374929] [PubMed: 34421149] E7
Lea T, Amada N, Jungaberle H, Schecke H, Scherbaum N, Klein M. Perceived outcomes of psychedelic microdosing as self-managed therapies for mental and substance use disorders. Psychopharmacology. 2020;237(5):1521–1532. [PubMed: 32043165] E2
Ledermann EK. Existential psychotherapy. Israel Annals of Psychiatry & Related Disciplines. 1970;8(3):255–262. [PubMed: 4929280] E7
Leger RF, Unterwald EM. Assessing the effects of methodological differences on outcomes in the use of psychedelics in the treatment of anxiety and depressive disorders: A systematic review and meta-analysis. Journal of Psychopharmacology. 2022;36(1):20–30. [PubMed: 34519567] E8
Li N-X, Hu Y-R, Chen W-N, Zhang B. Dose effect of psilocybin on primary and secondary depression: A preliminary systematic review and meta-analysis. Journal of Affective Disorders. 2022;296:26–34. [PubMed: 34587546] E8
Liechti ME. Modern Clinical Research on LSD. Neuropsychopharmacology. 2017;42(11):2114–2127. [PMC free article: PMC5603820] [PubMed: 28447622] E7
Loizaga-Velder A, Verres R. Therapeutic effects of ritual ayahuasca use in the treatment of substance dependence--qualitative results. Journal of Psychoactive Drugs. 2014;46(1):63–72. [PubMed: 24830187] E6
Lotsof HS, Alexander NE. Case studies of ibogaine treatment: implications for patient management strategies. The Alkaloids Chemistry & Biology. 2001;56:293–313. [PubMed: 11705114] E7
Luciano D. Observations on treatment with ibogaine. American Journal on Addictions. 1998;7(1):89–90. [PubMed: 9522011] E7
Ludwig A, Levine J, Stark L, Lazar R. A clinical study of LSD treatment in alcoholism. American Journal of Psychiatry. 1969;126(1):59–69. [PubMed: 5798383] E11
Ludwig AM. Studies in alcoholism and LSD. I. Influence of therapist attitudes on treatment outcome. American Journal of Orthopsychiatry. 1968;38(4):733–737. [PubMed: 5661557] E4
Luz M, Mash DC. Evaluating the toxicity and therapeutic potential of ibogaine in the treatment of chronic opioid abuse. Expert Opinion On Drug Metabolism & Toxicology. 2021;17(9):1019–1022. [PubMed: 34139922] E7
Lyons T, Carhart-Harris RL. Increased nature relatedness and decreased authoritarian political views after psilocybin for treatment-resistant depression. Journal of Psychopharmacology. 2018;32(7):811–819. [PMC free article: PMC6047302] [PubMed: 29338538] E4
MacCallum WA. The use of L.S.D. 25 in psychiatry. Ulster Medical Journal. 1968;37(2):151–154. [PMC free article: PMC2384848] [PubMed: 5700239] E1
Maciulaitis R, Kontrimaviciute V, Bressolle FM, Briedis V. Ibogaine, an anti-addictive drug: pharmacology and time to go further in development. A narrative review. Human & Experimental Toxicology. 2008;27(3):181–194. [PubMed: 18650249] E7
Madsen JD, Hoffart A. Psychotherapy with the aid of LSD. Nordic Journal of Psychiatry. 1996;50(6):477–486. E4
Madsen JD, Oyslebo T, Hoffart A. A follow-up study of psycholytic therapy with the aid of LSD. Nordic Journal of Psychiatry. 1996;50(6):487–494. E4
Malitz S. The role of mescaline and D-lysergic acid in psychiatric treatment. Diseases of the Nervous System. 1966;7 Suppl(7):39–42. [PubMed: 5965283] E7
Malleson N. Acute adverse reactions to LSD in clinical and experimental use in the United Kingdom. British Journal of Psychiatry. 1971;118(543):229–230. [PubMed: 4995932] E4
Marks M, Cohen IG. Psychedelic therapy: a roadmap for wider acceptance and utilization. Nature Medicine. 2021;27(10):1669–1671. [PubMed: 34608331] E7
Marschall J, Fejer G, Lempe P, et al. Psilocybin microdosing does not affect emotion-related symptoms and processing: A preregistered field and lab-based study. Journal of Psychopharmacology. 2022;36(1):97–113. [PMC free article: PMC8801668] [PubMed: 34915762] E1
Marseille E, Kahn JG, Yazar-Klosinski B, Doblin R. The cost-effectiveness of MDMA-assisted psychotherapy for the treatment of chronic, treatment-resistant PTSD. PLoS ONE [Electronic Resource]. 2020;15(10):e0239997. [PMC free article: PMC7556534] [PubMed: 33052965] E6
Marseille E, Mitchell JM, Kahn JG. Updated cost-effectiveness of MDMA-assisted therapy for the treatment of posttraumatic stress disorder in the United States: Findings from a phase 3 trial. PLoS ONE [Electronic Resource]. 2022;17(2):e0263252. [PMC free article: PMC8880875] [PubMed: 35213554] E6
Martin AJ. A case of early paranoiac psychosis treated by Lysergic Acid Diethylamide (LSD). Acta Psychotherapeutica. 1964;12(2):119–130. [PubMed: 14155989] E6
Mash DC, Kovera CA, Buck BE, et al. Medication development of ibogaine as a pharmacotherapy for drug dependence. Annals of the New York Academy of Sciences. 1998;844:274–292. [PubMed: 9668685] E7
McCabe OL, Savage C, Kurland A, Unger S. Psychedelic (LSD) therapy of neurotic disorders: Short term effects. Journal of Psychedelic Drugs. 1972;5(1):18–28. E4
Mechaneck R, Feldstein S, Dahlberg CC, Jaffe J. Experimental investigation of LSD as a psychotherapeutic adjunct. Comprehensive Psychiatry. 1968;9(5):490–498. [PubMed: 4879643] E7
Merlis S. The effects of mescaline sulfate in chronic schizophrenia. Journal of Nervous & Mental Disease. 1957;125(3):432–434. [PubMed: 13481749] E4
Mertens LJ, Preller KH. Classical Psychedelics as Therapeutics in Psychiatry - Current Clinical Evidence and Potential Therapeutic Mechanisms in Substance Use and Mood Disorders. Pharmacopsychiatry. 2021;54(4):176–190. [PubMed: 33472250] E7
Mertens LJ, Wall MB, Roseman L, Demetriou L, Nutt DJ, Carhart-Harris RL. Therapeutic mechanisms of psilocybin: Changes in amygdala and prefrontal functional connectivity during emotional processing after psilocybin for treatment-resistant depression. Journal of Psychopharmacology. 2020;34(2):167–180. [PubMed: 31941394] E4
Mian MN, Altman BR, Earleywine M. Ayahuasca’s antidepressant effects covary with behavioral activation as well as mindfulness. Journal of Psychoactive Drugs. 2020;52(2):130–137. [PubMed: 31570056] E2
Mithoefer M. MDMA-assisted psychotherapy for the treatment of post-traumatic stress disorder. Winkelman, Michael J [Ed]; Roberts, Thomas B [Ed] (2007). 2007;Psychedelic medicine:New evidence for hallucinogenic substances as treatments. E10
Mithoefer MC, Feduccia AA, Jerome L, et al. MDMA-assisted psychotherapy for treatment of PTSD: study design and rationale for phase 3 trials based on pooled analysis of six phase 2 randomized controlled trials. Psychopharmacology. 2019;236(9):2735–2745. [PMC free article: PMC6695343] [PubMed: 31065731] E8
Molla S. The personal experiences of ayahuasca brew users as a therapeutic catalyst for substance dependence: A qualitative exploratory approach. Dissertation Abstracts International: Section B: The Sciences and Engineering. 2021;82(9-B). E2
Moreno FA, Delgado PL. Hallucinogen-induced relief of obsessions and compulsions. American Journal of Psychiatry. 1997;154(7):1037–1038. [PubMed: 9210762] E6
Motta RW. MDMA (“Ecstasy”) for PTSD. Motta, Robert W (2020). 2020;Alternative therapies for PTSD:The science of mind-body treatments. (pp. 163–178). 229 pp. Washington. E7
Mottin JL. Drug-induced attenuation of alcohol consumption. A review and evaluation of claimed, potential or current therapies. Quarterly Journal of Studies on Alcohol. 1973;34(2):444–472. [PubMed: 4270100] E7
Mullard A. Will psychedelics be ‘a revolution in psychiatry’? Nature Reviews Drug Discovery. 2021;20(6):418–419. [PubMed: 34011999] E7
Muthukumaraswamy SD, Forsyth A, Lumley T. Blinding and expectancy confounds in psychedelic randomized controlled trials. Expert Review of Clinical Pharmacology. 2021;14(9):1133–1152. [PubMed: 34038314] E7
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. [PubMed: 31382100] E8
Nafees T, Wase HA, Amir Malik MF. Is the 3,4-Methylenedioxymethamphetamine assisted psychotherapy a novel approach to managing post-traumatic stress disorder? JPMA - Journal of the Pakistan Medical Association. 2019;69(8):1229. [PubMed: 31431790] E7
Nayak SM, Gukasyan N, Barrett FS, Erowid E, Erowid F, Griffiths RR. Classic psychedelic coadministration with lithium, but not lamotrigine, is associated with seizures: An analysis of online psychedelic experience reports. Pharmacopsychiatry. 2021;54(5):240–245. [PubMed: 34348413] E1
Nicholas CR, Wang JB, Coker A, et al. The effects of MDMA-assisted therapy on alcohol and substance use in a phase 3 trial for treatment of severe PTSD. Drug & Alcohol Dependence. 2022;233:109356. [PMC free article: PMC9750500] [PubMed: 35286849] E4
Noorani T, Garcia-Romeu A, Swift TC, Griffiths RR, Johnson MW. Psychedelic therapy for smoking cessation: Qualitative analysis of participant accounts. Journal of Psychopharmacology. 2018;32(7):756–769. [PubMed: 29938565] E4
Nour MM, Krzanowski J. Therapeutic potential of psychedelic agents. British Journal of Psychiatry. 2015;206(5):433–434. [PubMed: 25934306] E7
Nunes AA, dos Santos RG, Osorio FL, Sanches RF, Crippa JAS, Hallak JEC. Effects of ayahuasca and its alkaloids on drug dependence: A systematic literature review of quantitative studies in animals and humans. Journal of Psychoactive Drugs. 2016;48(3):195–205. [PubMed: 27230395] E8
O’Shea B, Fagan J. Lysergic acid diethylamide. Irish Medical Journal. 2001;94(7):217. [PubMed: 11693215] E7
Pahnke WN, Kurland AA, Unger S, Savage C, Grof S. The experimental use of psychedelic (LSD) psychotherapy. Internationale Zeitschrift fur Klinische Pharmakologie, Therapie und Toxikologie. 1971;4(4):446–454. [PubMed: 5566727] E1
Palladino L. Vine of soul: A phenomenological study of ayahuasca and its effect on depression. Dissertation Abstracts International: Section B: The Sciences and Engineering. 2010;71(1-B):668. E2
Parashos AJ. The psilocybin-induced “state of drunkenness” in normal volunteers and schizophrenics. Behavioral Neuropsychiatry. 1976;8(1–12):1976–1977. [PubMed: 1052267] E4
Parker LA, Siegel S. Modulation of the effects of rewarding drugs by ibogaine. The Alkaloids Chemistry & Biology. 2001;56:211–225. [PubMed: 11705109] E7
Parrott AC. The psychotherapeutic potential of MDMA (3,4-methylenedioxymethamphetamine): an evidence-based review. Psychopharmacology. 2007;191(2):181–193. [PubMed: 17297639] E7
Pearson C, Siegel J, Gold JA. Psilocybin-assisted psychotherapy for depression: Emerging research on a psychedelic compound with a rich history. Journal of the Neurological Sciences. 2022;434:120096. [PubMed: 34942586] E7
Perrine DM. Hallucinogens and obsessive-compulsive disorder. American Journal of Psychiatry. 1999;156(7):1123. [PubMed: 10401480] E7
Ponte L, Jerome L, Hamilton S, et al. Sleep Quality Improvements After MDMA-Assisted Psychotherapy for the Treatment of Posttraumatic Stress Disorder. Journal of Traumatic Stress. 2021;34(4):851–863. [PMC free article: PMC8453707] [PubMed: 34114250] E4
Popik P, Layer RT, Skolnick P. 100 years of ibogaine: neurochemical and pharmacological actions of a putative anti-addictive drug. Pharmacological Reviews. 1995;47(2):235–253. [PubMed: 7568327] E7
Pos R. LSD-25 as an adjunct to long-term psychotherapy. Canadian Psychiatric Association Journal. 1966;11(4):330–342. [PubMed: 5967401] E4
Prue RE. King alcohol to chief peyote: A grounded theory investigation of the supportive factors of the native American church for drug and alcohol abuse recovery. Dissertation Abstracts International Section A: Humanities and Social Sciences. 2009;69(11-A). E2
Reiff CM, McDonald WM. MDMA-assisted psychotherapy for the treatment of PTSD. Revista Brasileira de Psiquiatria. 2021;43(2):123–124. [PMC free article: PMC8023168] [PubMed: 33053043] E7
Reiff CM, Richman EE, Nemeroff CB, et al. Psychedelics and Psychedelic-Assisted Psychotherapy. American Journal of Psychiatry. 2020;177(5):391–410. [PubMed: 32098487] E8
Renelli M, Fletcher J, Tupper KW, Files N, Loizaga-Velder A, Lafrance A. An exploratory study of experiences with conventional eating disorder treatment and ceremonial ayahuasca for the healing of eating disorders. Eating & Weight Disorders: EWD. 2020;25(2):437–444. [PubMed: 30474794] E2
Rhead JC, Soskin RA, Turek I, et al. Psychedelic drug (DPT)-assisted psychotherapy with alcoholics: A controlled study. Journal of Psychedelic Drugs. 1977;9(4):287–300. E10
Rodrigues AV, Almeida FJ, Vieira-Coelho MA. Dimethyltryptamine: Endogenous Role and Therapeutic Potential. Journal of Psychoactive Drugs. 2019;51(4):299–310. [PubMed: 31018803] E7
Romeo B, Hermand M, Petillion A, Karila L, Benyamina A. Clinical and biological predictors of psychedelic response in the treatment of psychiatric and addictive disorders: A systematic review. Journal of Psychiatric Research. 2021;137:273–282. [PubMed: 33730602] E8
Romeo B, Karila L, Martelli C, Benyamina A. Efficacy of psychedelic treatments on depressive symptoms: A meta-analysis. Journal of Psychopharmacology. 2020;34(10):1079–1085. [PubMed: 32448048] E8
Roseman L, Demetriou L, Wall MB, Nutt DJ, Carhart-Harris RL. Increased amygdala responses to emotional faces after psilocybin for treatment-resistant depression. Neuropharmacology. 2018;142:263–269. [PubMed: 29288686] E4
Rucker J, Jafari H, Mantingh T, et al. Psilocybin-assisted therapy for the treatment of resistant major depressive disorder (PsiDeR): protocol for a randomised, placebo-controlled feasibility trial. BMJ Open. 2021;11(12):e056091. [PMC free article: PMC8638462] [PubMed: 34853114] E7
Rucker JJ, Jelen LA, Flynn S, Frowde KD, Young AH. Psychedelics in the treatment of unipolar mood disorders: a systematic review. Journal of Psychopharmacology. 2016;30(12):1220–1229. [PubMed: 27856684] E8
Rydzynski Z, Gruszczynski W. Treatment of alcoholism with psychotomimetic drugs. A follow-up study. Activitas Nervosa Superior. 1978;20(1):81–82. [PubMed: 636792] E4
Sandison RA. Psychological aspects of the LSD treatment of the neuroses. Journal of Mental Science. 1954;100(419):508–515. [PubMed: 13175012] E4
Sandison RA, Whitelaw JD. Further studies in the therapeutic value of lysergic acid diethylamide in mental illness. Journal of Mental Science. 1957;103(431):332–343. [PubMed: 13429304] E1
Sarett M, Cheek F, Osmond H. Reports of wives of alcoholics of effects of LSD-25 treatment of their husbands. Archives of General Psychiatry. 1966;14(2):171–178. [PubMed: 5948087] E1
Sarris J, Pinzon Rubiano D, Day K, Galvao-Coelho NL, Perkins D. Psychedelic medicines for mood disorders: current evidence and clinical considerations. Current Opinion in Psychiatry. 2022;35(1):22–29. [PubMed: 34855694] E7
Sartori SB, Singewald N. Novel pharmacological targets in drug development for the treatment of anxiety and anxiety-related disorders. Pharmacology & Therapeutics. 2019;204:107402. [PubMed: 31470029] E7
Savage C, Fadiman J, Mogar R, Allen MH. The effects of psychedelic (LSD) therapy on values, personality, and behavior. International Journal of Neuropsychiatry. 1966;2(3):241–254. [PubMed: 5961962] E4
Savage C, McCabe OL. Residential psychedelic (LSD) therapy for the narcotic addict. A controlled study. Archives of General Psychiatry. 1973;28(6):808–814. [PubMed: 4575166] E1
Savage C, McCabe OL, Kurland AA, Hanlon T. LSD-assisted psychotherapy in the treatment of severe chronic neurosis. Journal of Altered States of Consciousness. 1973;1(1):31–47. E4
Schenberg EE. Psychedelic drugs as new tools in psychiatric therapeutics. Revista Brasileira de Psiquiatria. 2021;43(2):121–122. [PMC free article: PMC8023166] [PubMed: 32578688] E7
Schenk S, Newcombe D. Methylenedioxymethamphetamine (MDMA) in Psychiatry: Pros, Cons, and Suggestions. Journal of Clinical Psychopharmacology. 2018;38(6):632–638. [PubMed: 30303861] E7
Schep LJ, Slaughter RJ, Galea S, Newcombe D. Ibogaine for treating drug dependence. What is a safe dose? Drug & Alcohol Dependence. 2016;166:1–5. [PubMed: 27426011] E7
Schmid Y, Gasser P, Oehen P, Liechti ME. Acute subjective effects in LSD- and MDMA-assisted psychotherapy. Journal of Psychopharmacology. 2021;35(4):362–374. [PubMed: 33853422] E4
Sessa B. Self-medication of LSD and MDMA to treat mental disorders: A case series. Merrick, Joav [Ed]. 2012. E2
Sessa B, Aday JS, O’Brien S, et al. Debunking the myth of ‘Blue Mondays’: No evidence of affect drop after taking clinical MDMA. Journal of Psychopharmacology. 2022;36(3):360–367. [PubMed: 34894842] E4
Sessa B, Johnson MW. Can psychedelic compounds play a part in drug dependence therapy? British Journal of Psychiatry. 2015;206(1):1–3. [PubMed: 25561484] E7
Sessa B, Sakal C, O’Brien S, Nutt D. First study of safety and tolerability of 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy in patients with alcohol use disorder: preliminary data on the first four participants. BMJ Case Reports. 2019;12(7):15. [PMC free article: PMC6663239] [PubMed: 31308191] E4
Shagass C, Bittle RM. Therapeutic effects of LSD: a follow-up study. Journal of Nervous & Mental Disease. 1967;144(6):471–478. [PubMed: 4860930] E1
Sheppard SG. A preliminary investigation of ibogaine: case reports and recommendations for further study. Journal of Substance Abuse Treatment. 1994;11(4):379–385. [PubMed: 7966509] E6
Sherwood AM, Prisinzano TE. Novel psychotherapeutics - a cautiously optimistic focus on Hallucinogens. Expert Review of Clinical Pharmacology. 2018;11(1):1–3. [PMC free article: PMC6121772] [PubMed: 29224406] E7
Shirvaikar RV, Kelkar YW. Therapeutic trial of lysergic Acid diethylamide (LSD) and thioridazine in chronic schizophrenia. Neurology India. 1966;14(2):97–101. [PubMed: 5330728] E4
Siegel AN, Meshkat S, Benitah K, et al. Registered clinical studies investigating psychedelic drugs for psychiatric disorders. Journal of Psychiatric Research. 2021;139:71–81. [PubMed: 34048997] E8
Skolnick P. Ibogaine as a glutamate antagonist: relevance to its putative antiaddictive properties. The Alkaloids Chemistry & Biology. 2001;56:55–62. [PubMed: 11705116] E7
Smith CG. Gilles De La Tourette syndrome treated with LSD. Irish Journal of Medical Science. 1969;8(6):269–271. [PubMed: 5258762] E6
Smith CM. A new adjunct to the treatment of alcoholism: the hallucinogenic drugs. Quarterly Journal of Studies on Alcohol. 1958;19(3):406–417. [PubMed: 13579169] E4
Smith CM. Some reflections on the possible therapeutic effects of the hallucinogens; with special reference to alcoholism. Quarterly Journal of Studies on Alcohol. 1959;20(2):292–301. [PubMed: 13668045] E7
Smith KW, Sicignano DJ, Hernandez AV, White CM. MDMA-Assisted Psychotherapy for Treatment of Posttraumatic Stress Disorder: A Systematic Review With Meta-Analysis. Journal of Clinical Pharmacology. 2022;62(4):463–471. [PubMed: 34708874] E8
Sola EM. MDMA-assisted psychotherapy for PTSD: A thematic analysis of transformation in combat veterans. Dissertation Abstracts International: Section B: The Sciences and Engineering. 2018;79(10-B(E). E6
Solursh LP. The use of LSD-25 in psychotherapy: An evaluation. International Journal of Neuropsychiatry. 1966;2(6):651–656. E4
Soskin RA. Personality and attitude change after two alcoholism treatment programs. Comparative contributions of lysergide and Human Relations Training. Quarterly Journal of Studies on Alcohol. 1970;31(4):920–931. [PubMed: 4395263] E4
Soskin RA. The use of LSD in time-limited psychotherapy. Journal of Nervous & Mental Disease. 1973;157(6):410–419. [PubMed: 4148471] E1
Soskin RA. Dipropyltryptamine in psychotherapy. Current Psychiatric Therapies. 1975;15:147–156. [PubMed: 1181120] E7
Spriggs MJ, Kettner H, Carhart-Harris RL. Positive effects of psychedelics on depression and wellbeing scores in individuals reporting an eating disorder. Eating & Weight Disorders: EWD. 2021;26(4):1265–1270. [PubMed: 32895801] E1
Strauss N. Psilocybin-assisted therapy for anxiety and depression: implications for euthanasia. Medical Journal of Australia. 2017;206(11):468–469. [PubMed: 28918722] E7
Suzuki J, Dekker MA, Valenti ES, et al. Toxicities associated with NBOMe ingestion-A novel class of potent hallucinogens: A review of the literature. Psychosomatics: Journal of Consultation and Liaison Psychiatry. 2015;56(2):129–139. [PMC free article: PMC4355190] [PubMed: 25659919] E7
Teixeira PJ, Johnson MW, Timmermann C, et al. Psychedelics and health behaviour change. Journal of Psychopharmacology. 2022;36(1):12–19. [PMC free article: PMC8801670] [PubMed: 34053342] E7
Thomas K, Malcolm B, Lastra D. Psilocybin-Assisted Therapy: A Review of a Novel Treatment for Psychiatric Disorders. Journal of Psychoactive Drugs. 2017;49(5):446–455. [PubMed: 28481178] E7
Tomsovic M, Edwards RV. Lysergide treatment of schizophrenic and nonschizophrenic alcoholics: a controlled evaluation. Quarterly Journal of Studies on Alcohol. 1970;31(4):932–949. [PubMed: 5490826] E11
Tupper KW, Wood E, Yensen R, Johnson MW. Psychedelic medicine: a re-emerging therapeutic paradigm. CMAJ Canadian Medical Association Journal. 2015;187(14):1054–1059. [PMC free article: PMC4592297] [PubMed: 26350908] E7
Uthaug MV, Mason NL, Toennes SW, et al. A placebo-controlled study of the effects of ayahuasca, set and setting on mental health of participants in ayahuasca group retreats. Psychopharmacology. 2021;238(7):1899–1910. [PMC free article: PMC8233273] [PubMed: 33694031] E1
Van Dusen W, Wilson W, Miners W, Hook H. Treatment of alcoholism with lysergide. Quarterly Journal of Studies on Alcohol. 1967;28(2):295–304. [PubMed: 6049171] E11
Vargas-Perez H, Doblin R. The potential of psychedelics as a preventative and auxiliary therapy for drug abuse. Current Drug Abuse Reviews. 2013;6(1):1. [PubMed: 23773089] E7
Varker T, Watson L, Gibson K, Forbes D, O’Donnell ML. Efficacy of Psychoactive Drugs for the Treatment of Posttraumatic Stress Disorder: A Systematic Review of MDMA, Ketamine, LSD and Psilocybin. Journal of Psychoactive Drugs. 2021;53(1):85–95. [PubMed: 32931403] E8
Vastag B. Addiction research. Ibogaine therapy: a ‘vast, uncontrolled experiment’. Science. 2005;308(5720):345–346. [PubMed: 15831735] E7
Vermetten E, Yehuda R. MDMA-assisted psychotherapy for posttraumatic stress disorder: A promising novel approach to treatment. Neuropsychopharmacology. 2020;45(1):231–232. [PMC free article: PMC6879520] [PubMed: 31455855] E7
Viamontes JA. Review of drug effectiveness in the treatment of alcoholism. American Journal of Psychiatry. 1972;128(12):1570–1571. [PubMed: 4554403] E7
Voineskos D, Daskalakis ZJ, Blumberger DM. Management of treatment-resistant depression: Challenges and strategies. Neuropsychiatric Disease and Treatment. 2020;16:221–234. [PMC free article: PMC6982454] [PubMed: 32021216] E7
Wagner AC, Mithoefer MC, Mithoefer AT, Monson CM. Combining Cognitive-Behavioral Conjoint Therapy for PTSD with 3,4-Methylenedioxymethamphetamine (MDMA): A Case Example. Journal of Psychoactive Drugs. 2019;51(2):166–173. [PubMed: 30890035] E6
Wagner MT, Mithoefer MC, Mithoefer AT, et al. Therapeutic effect of increased openness: Investigating mechanism of action in MDMA-assisted psychotherapy. Journal of Psychopharmacology. 2017;31(8):967–974. [PMC free article: PMC5544120] [PubMed: 28635375] E4
Watts R, Day C, Krzanowski J, Nutt D, Carhart-Harris R. Patients’ accounts of increased “connectedness” and “acceptance” after psilocybin for treatment-resistant depression. Journal of Humanistic Psychology. 2017;57(5):520–564. E6
Weston NM, Gibbs D, Bird CIV, et al. Historic psychedelic drug trials and the treatment of anxiety disorders. Depression & Anxiety. 2020;37(12):1261–1279. [PubMed: 32627308] E8
Wheeler SW, Dyer NL. A systematic review of psychedelic-assisted psychotherapy for mental health: An evaluation of the current wave of research and suggestions for the future. Psychology of Consciousness: Theory, Research, and Practice. 2020;7(3):279–315. E8
Williams MT, Reed S, Aggarwal R. Culturally informed research design issues in a study for MDMA-assisted psychotherapy for posttraumatic stress disorder. Journal of Psychedelic Studies. 2020;4(1):40–50. E7
Yaden DB, Yaden ME, Griffiths RR. Psychedelics in Psychiatry-Keeping the Renaissance From Going Off the Rails. JAMA Psychiatry. 2021;78(5):469–470. [PMC free article: PMC8102315] [PubMed: 33263720] E7
Yensen R, Di Leo FB, Rhead JC, et al. MDA-assisted psychotherapy with neurotic outpatients: A pilot study. Journal of Nervous and Mental Disease. 1976;163(4):233–245. [PubMed: 972325] E1
Young AH. The age of psychedelics. Journal of Psychopharmacology. 2022;36(1):3–5. [PubMed: 35090362] E7
Zeifman RJ, Yu D, Singhal N, Wang G, Nayak SM, Weissman CR. Decreases in Suicidality Following Psychedelic Therapy: A Meta-Analysis of Individual Patient Data Across Clinical Trials. Journal of Clinical Psychiatry. 2022;83(2):18. [PubMed: 35044730] E8

APPENDIX C. EVIDENCE TABLES

CHARACTERISTICS OF INCLUDED PRIMARY STUDIES

RCTs

Download PDF (218K)

Observational Studies

Download PDF (225K)

SETTING CHARACTERISTICS OF INCLUDED PRIMARY STUDIES

RCTs

Download PDF (211K)

Observational Studies

Download PDF (213K)

OUTCOME DATA OF INCLUDED PRIMARY STUDIES

Disorder-specific Outcomes

Download PDF (274K)

Studies Used in Pooled Analysis (MDMA for PTSD)

Download PDF (182K)

Harms

Ayahuasca (PDF, 184K)

Ibogaine (PDF, 182K)

MDMA (PDF, 301K)

Psilocybin (PDF, 202K)

QUALITY ASSESSMENT OF INCLUDED PRIMARY STUDIES

RCTs – RoB 2 Tool

Download PDF (217K)

Observational Studies – ROBINS-I Tool for Cohort Studies

Download PDF (182K)

Observational Studies – ROBINS-I Tool for Uncontrolled Pre-post Studies

Download PDF (221K)

STRENGTH OF EVIDENCE FOR INCLUDED STUDIES

Download PDF (206K)

APPENDIX D. PEER REVIEW DISPOSITION

Comment #Reviewer #CommentAuthor Response
Are the objectives, scope, and methods for this review clearly described?
11Yes None
23Yes None
34Yes None
45Yes None
56Yes None
67Yes None
78Yes None
Is there any indication of bias in our synthesis of the evidence?
8 1No None
92No None
103Yes - Pg22, 5 & Pg32, 21: “but whether effects are primarily due to psilocybin exposure or intensive psychotherapy remains unclear”, can you expand on the inclusion of this statement? This doesn’t seem to address a key question of this evidence review. This statement perhaps presents a false dichotomy unnecessarily, I suggest more framing or removing.Thank you for this comment. We agree that this wording is problematic without additional context and have removed it. In the section pertaining to PTSD, we now discuss in more detail our assessment of what the evidence can and cannot tell us about the relative contribution of the intervention components.
114No None
125No None
136No None
147No None
158No None
Are there any published or unpublished studies that we may have overlooked?
161No
172No
183Yes - Anderson BT, et al. (2020). Psilocybin-assisted group therapy for demoralized older long-term AIDS survivor men: an open-label safety and feasibility pilot study. EClinicalMedicine. 100538. Thank you for identifying this study, which we did not find in our search. This study does not meet our inclusion criteria as we excluded studies of adults with serious medical illness (eg terminal cancer, end-stage renal disease, and in this case HIV/AIDS) experiencing secondary mental health symptoms.
194No None
205No None
216No None
227No None
238No None
Additional suggestions or comments can be provided below. If applicable, please indicate the page and line numbers from the draft report.
24 1Overall I think this is a good draft. I found it helpful to see the data summarized this way. My overarching concern is the lack of attention to the adequacy of placebo control in this research. It is a known challenge because it is difficult to plausibly mimic the effects of psychedelic drugs, e.g., https://doi​.org/10.1038​/s41591-021-01524-1. The limited use of active placebos and reduced adequacy of blinding tempers the evidence of effectiveness. I am commenting on the MDMA-assisted psychotherapy studies for PTSD but the issue is broadly relevant.Thank you for your comments regarding the use of different placebos. In the Results section pertaining to MDMA-assisted psychotherapy, we have expanded our discussion on the use of inactive (0mg) and low-dose placebos. We comment that low-dose placebos are meant to produce some of the sensations of having ingested a psychedelic but are thought to have a subtherapeutic effect that is distinct from psychedelic doses used in intervention groups. We revised Figure 3 (forest plot) to separate the studies using inactive and low-dose placebos and now provide pooled estimates for the study sub-groups. We comment that use of inactive placebos with inadequate blinding is a potential source of bias in the Results section and in the Limitations sections.
251In MDMA-assisted psychotherapy for PTSD, 2 of the 5 published studies used inactive placebo (Mithoefer et al., 2011; Mitchell et al., 2021), whereas the other studies used lose-dose MDMA: 25 mg (Oehen et al., 2013), 30 mg (Mithoefer et al., 2018), and 40 mg (Ot’alora, 2018). This is a particular problem for interventions like psychedelics, for which there is a lot of hype and public-facing “evidence” on the efficacy of these interventions.As stated above, we now explicitly discuss the use of 2 placebo types – 0mg placebo and low-dose placebo.
261Blinding was inadequate in the studies that used inactive placebo. In the Mithoefer et al. (2011) study, 95% of participants correctly guessed their assigned condition, and in the Mitchell study, 84%/96% guessed correctly in the inactive/MDMA groups, respectively. Guessing was less accurate in studies that used active placebo, but still incomplete: 46% overall in Mithoefer (2018); 46% vs. 66% in 25mg vs. 125 mg in Oehen (2013); and 73% vs. 58% in 40mg vs. 100/125 mg in Ot’alora (2018). Even if the effect sizes in the ESP report do not vary as a function of blinding—and for MDMA I think they probably do not due to the Mitchell study having a relatively small effect and the Mithoefer 2018 study having a large effect—this is a key issue that should be addressed in the report to caution readers.Thank you for your comments regarding the potential for ineffective blinding. As stated above, we comment that use of inactive placebos with inadequate blinding is a potential source of bias in the Results section and in the Limitations sections. We also recommend use of low-dose placebo in the Future Research section and recommend that studies report data on blinding effectiveness.
271Another concern is the limited number of total as well as Veteran participants that have been studied in MDMA-assisted psychotherapy for PTSD. Looking at the 5 published studies, these trials have included a total of only 176 participants in the ITT samples.* All except one study (Mitchell et al., 2021, N = 90) included fewer than 30 participants. Almost 2 out of 3 participants (64%) have been female and almost no participants have experienced combat trauma or were military veterans, with the exception of the study by Mithoefer et al. (2018), which enrolled 22 Veterans. The existing data therefore have limited generalizability to VA patients. The ongoing MAPS study (https:​//clinicaltrials​.gov/ct2/show/NCT04077437) may mitigate this limitation, but it is unlikely that many Veterans will be enrolled given that the protocol does not focus specifically on Veterans. Effect sizes in PTSD medication and psychotherapy studies are smaller in Veteran and active duty samples relative to non-military samples, so the lack of Veteran data is likely to cause the effects on Veterans to be overestimated. Thank you for this comment. We added a sentence to the Key Findings in the Executive Summary to highlight the low inclusion rates of Veterans. In the Discussion and Limitations section, we comment on concerns related to generalizing study findings to VHA populations. We have also recommended inclusion of Veteran populations as a priority in the Future Research section
281*I was surprised to see the sample sizes listed for these trials because they are smaller than those reported in the studies. I looked at one trial, Mitchell, in which the sample size is 90 overall and 89 in the primary MMRM analysis, even though data are missing for some participants in later assessments. Figure 3 in the ESP report lists the N as 79; however, the authors say the N for T4, the final endpoint, was 82. So even if the ESP report is using the T4 N, it is difficult to understand how the total was 79. I assume that the language on the calculation of effect sizes is standard, but in this and future reports, it would be helpful to clarify how the Ns were determined.Thank you for this comment. In this particular study, the outcome of change from baseline in PTSD symptoms was only reported for participants who completed the intervention, which was 79. We reviewed the report Tables and Figures for consistency and clarified when we were referencing the “analytic N” rather than the study’s total N.
291

Pg 1 Line 11-12: It is extremely important to present the effect sizes for the active and placebo controlled studies separately.

Also, one of the major issues in this literature is the problem of unblinding in placebo control. IT is a key issue that undermines the interpretation of findings. Having just reviewed this literature carefully, I don’t think this interpretation is valid given the serious limitations in the literature.

As stated above, we now explicitly discuss the use of 2 placebo types – 0mg placebo and low-dose placebo. We revised Figure 3 (forest plot) to separate the studies using inactive and low-dose placebos and now provide pooled estimates for the study sub-groups.
301Pg 1 Line 33: What about the challenges of placebo blinding--finding credible active placebo is challenging, especially for long-acting medications like MDMA and psilocybinPlease see responses to Comments #24-26.
311Pg 2 Line 18-19: Please see prior comments about the importance of presenting effect sizes for active and placebo control separately. This is analagous to presenting separate effect sizes for in psychotherapy studies for active and waitlist, for example. And placebo control is inadequate, so interpretation needs to be tempered.Please see responses to Comments #24-26.
321Pg 31 Line 48-49: This section [Discussion] needs to address the issue of placebo plinding and the need for adequate control. The possibility of plcebo effects is especially concerning for threatments like MDMA that have been promoted in the popular pressPlease see responses to Comments #24-26.
331Pg 31 Line 48-49: please see: Caution at psychiatry’s psychedelic frontier. Matthew J. Burke1,2,3,4 ✉ and Daniel M. Blumberger1,5 arising from J. M. Mitchell et al. Nature Medicine https://doi​.org/10.1038​/s41591-021-01336-3 (2021)Thank you for highlighting this commentary. In the Results section pertaining to MDMA-assisted psychotherapy for PTSD, we discuss the use of different placebo types and specifically highlight that Mitchell 2021 used an inactive placebo. As above, we recommend use of low-dose placebo in the Future Research section and recommend that studies report data on blinding effectiveness.
341Pg 31 Line 57: Two of the 5 published MDMA studies used inactive placebo (Mithoefer et al., 2011; Mitchell et al., 2021), whereas the other studies used lose-dose MDMA: 25 mg (Oehen et al., 2013), 30 mg (Mithoefer et al., 2018), and 40 mg (Ot’alora, 2018). Blinding was inadequate in the studies that used inactive placebo. In the Mithoefer et al. (2011) study, 95% of participants correctly guessed their assigned condition, and in the Mitchell study, 84%/96% guessed correctly in the inactive/MDMA groups, respectively. Guessing was less accurate in studies that used active placebo: 46% overall in Mithoefer (2018); 46% vs. 66% in 25mg vs. 125 mg in Oehen (2013); and 73% vs. 58% in 40mg vs. 100/125 mg in Ot’alora (2018). The data indicate that active placebo is needed in order to achieve adequate blinding, which is especially important given the novelty of MDMA and the media’s portrayal of its efficacy.Please see responses to Comments #24-26.
351Pg. 32 Line 16-17: Limitations of findings: Thesetrials have included a total of only 176 participants. All except one study (Mitchell et al., 2021, N = 90) included fewer than 30 participants. Almost 2 out of 3 participants (64%) have been female and almost no participants have experienced combat trauma or were military veterans, with the exception of the study by Mithoefer et al. (2018), which enrolled 22 Veterans. The existing data therefore have limited generalizability to VA patients. The ongoing MAPS study (https:​//clinicaltrials​.gov/ct2/show/NCT04077437) may mitigate this limitation, but it is unlikely that many Veterans will be enrolled given that the protocol does not focus specifically on Veterans.Please see response to Comment #27.
361Pg 33 Line 39-40: active placebo-controlled studiesPlease see responses to Comments #24-26.
371Pg 33 Line 44-45: include veteransPlease see response to Comment #27.
382This review is thorough and comprehensive and provides a detailed overview of the full landscape of psychedelic use in MH and SUD conditions, which is much needed and appreciated. However, I am concerned that the summary of findings for MDMA for PTSD is overstating the strength of the evidence. The limitations of studies were not as fully explored in this section as I think is warranted. For example, there was little mention of the problematic nature of the 2 studies that are placebo controlled, especially given in those studies the participants had very high rates of identifying whether they received treatment or placebo (hence very high risk of being unblinded). Lumping these studies together with the active comparator studies is problematic. Also, the total N across all MDMA studies is only 176, with all but the 2021 Mitchell study having N<30. Also - there is little note of the study population of greatest interest to VA - Veterans. Most of these psychedelic studies are lacking in veteran participants, especially combat veterans (of particularly important note for PTSD studies). This is something that should be added to the discussion as well as an area needed to expand upon findings with future research.Thank you for these comments. Please see our responses to Comments #24-27 above, which address similar concerns raised by Reviewer 1. We reassessed our Strength of Evidence (SOE) rating for MDMA-assisted psychotherapy for PTSD (now rated as low SOE) accounting for the limitations that you and other reviewers highlighted.
393Pg2, Ln58: long-term, cite long-term MDMA studies clearlyWe have removed this sentence from the Executive Summary to improve clarity. In our discussion of evidence gaps and areas for future research, we include the durability of treatment effects as a gap.
403Pg6, 41: “expectations” should maybe be changed to “intentions”? (a tenet of this intervention is “intention without expectation”, i.e., expectation/expectancy is a component of ‘set’ that is mitigated)Change made.
413P6,58: comma after “feasibility” Change made
423Pg11, 29: Mitchell used CAPS-5, which has a different score rangeWe have corrected the text regarding CAPS-5 scoring.
433Pg13, 53: consider making it clear that participants were able to continue psychotherapy with outside provider(s) during study participationThank you for this comment. In the Results section, we now provide more detail (when reported by individual studies) on how many study participants continued or restarted other treatments during the study period or follow-up period.
443Pg24, 51: “3-4 integration sessions”; the protocol from Mitchell (2021) included 9 integration sessions total and many Phase 2 MDMA studies included 6-9 integration sessionsWe had originally counted only the sessions that were not aligned with the experimental sessions. We have revised the wording to reflect the total number of sessions.
453Pg32, 27: edit “estimate draw conclusions” Removed “estimate”
464Thank you for the opportunity to review this ESP. I found the summary to be thorough in highlighting the limitations of current psychedelic research (which includes small size of trials, inconsistency in intervention and co-intervention, risk of bias, limitations in blinding, and lack of data on long-term durability). The recommendations for future research are prudent, as preliminary data is positive. The clinical implication of discontinuing current treatment is certainly relevant as more people seek these compounds worldwide and standard treatments are lacking.Thank you for your comments.
475The authors conducted a systematic review of studies examining the effects of psychedelic medications for mental health and substance use disorders. They included 38 studies. They found moderate strength evidence indicating that MDMA likely improves PTSD symptoms. The strength of evidence for the remaining effects were deemed either low or insufficient. None
485This is a very well written and comprehensive review of the literature. In my view, the authors provided a balanced portrayal and contextualized their findings as applied to the veteran population nicely. I have a few suggestions they might consider addressing.Thank you for your comments. Suggestions are addressed below.
495I wonder if it would be helpful to spend a bit more time discussing the content of the psychotherapy component of the psychedelic-assisted psychotherapies. A brief description of this treatment component is included in Table 2 but was discussed only briefly in the text. A better understanding of what this psychotherapy component entails may be helpful for stakeholders, particularly given issues related to accessibility and scalability which were highlighted. For example, how equipped might VA mental health providers / systems be to deliver the interventions that are being tested in these trials?We agree and have expanded our discussion of the psychotherapy component of the interventions for PTSD and depression in particular.
505I may have missed this, but I was curious what sample size cutoff was used to evaluate whether a trial was small or not for the purposes of grading the strength of evidence. In particular, I was curious why the Carhart-Harris et al. (2021) psilocybin for depression trial (n = 59) provided insufficient evidence. In comparison, the authors graded evidence from the Davis et al. (2020) trial (n = 27) when combined with 4 pre-post studies as providing low strength of evidence (i.e., not insufficient).Thank you for this comment. We reassessed our Strength of Evidence (SOE) ratings for the outcomes related to depression to be consistent with the algorithm we presented in the Methods section (“low strength evidence consisted of a single trial or multiple small trials and/or multiple observational studies, with unclear to high risk of bias, consistent findings, and clinically relevant outcomes”). In this case, we rated the SOE as low for the outcomes informed by the Carhart-Harris 2021 trial and the Davis 2020 trial.
515I realize this may not be typically done for VA ESP reviews, but I wondered whether it would be helpful to calculate standardized effect sizes for those studies that reported mean differences (e.g., MDMA for PTSD, Carhart-Harris et al., 2020). From my perspectives, effect sizes provide another helpful metric of potential magnitude of effects beyond the grading of the strength of evidence.Thank you for this comment. Given this and other reviewers’ feedback, we have calculated SMDs for CAPS outcomes of MDMA for PTSD studies.
525I wondered about the authors’ choice to combine MDMA trials that included placebo and active controls. In general, I think it is much preferred to avoid combining across control condition types. At the very least, the authors might discuss why this is appropriate (e.g., active control MDMA dose was too low to be considered therapeutic and/or unlikely to produce a perceptible effect). Might it be possible to examine whether control condition type moderated treatment effects?Thank you for these comments. Please see our responses to Comments #24-25 above, which address similar concerns raised by Reviewer 1. We revised Figure 3 (forest plot) to separate the studies using 0mg and low-dose placebos and now provide pooled estimates for the study sub-groups.
535Two potential typos to correct: p. 2, line 60 (“PTSD-assisted psychotherapy -> “psychedelic-assisted psychotherapy”), p. 11, line 35 (“PI” -> “CI”).“PTSD-assisted” has been changed to “psychedelicassisted”. “PI” is correct as it is referring to the prediction interval, not the confidence interval.
546None None
557This is a concisely written report summarizing the evidence to date on the use of psychedelic pharmacotherapy for the treatment of mental health and substance use disorder. The overall report was informative, accurate and portrayed a non-biased review of classic and non-classic psychedelics, with their evolution, history, structure, and proposed mechanism of action. A comprehensive review of the evidence to date on efficacy, risks and benefits was well summarized. As stated in the review, the evidence on psychedelics for the treatment of mental health and substance use disorders is insufficient or low strength in evidence for most of the psychedelics. The one caveat to this is the evidence for MDMA-assisted therapy for PTSD where research according to this report is moderately strong in favor of the benefits of MDMA for improving PSTD. While the evidence does in fact support a strong signal for the efficacy of MDMA for treating PTSD with associated serious risks being rare, there are several practical, methodological and generalizability concerns with these studies that should be made more explicit - as they potentially mitigate this favorable rating. These concerns are summarized below.Thank you for your comments. Suggestions are addressed below.
567The “psychotherapy” component across all the MDMA-assisted therapy studies for PTSD is vague and varies significantly in session number and likely content, except Monson et al., 2020. It is unclear if any of the strides made in PTSD psychotherapy research over the past 2 decades have been integrated into the “therapy” component of the MAPS MDMA-assisted therapy protocol for PTSD. Current MAPS protocol for preparation and integration is loosely structured and not in line with current best practice for PTSD therapy. This has significant implications for replication and implementation in settings such as VHA. Future studies examining MDMA-assisted evidence-based psychotherapy for PTSD are needed.Thank you for this comment. We added a paragraph to the Limitations section to discuss the lack of clarify on to what extent, if any, protocols such as those used by MAPS overlap with recommended VA/DoD psychotherapy approaches.
577A significant methodological concern across the MDMA studies is the placebo comparator. Given the subjective experience of MDMA there is a clear placebo problem with many of these studies and this should temper enthusiasm of findings. Although this is highlighted in the review, this point should be made more explicit.Thank you for these comments. Please see our responses to Comments #24-25 above, which address similar concerns raised by Reviewer 1.
587Most of the published MDMA studies are small. The largest study to date is Mitchell and colleagues (2021) which was only an N=90. This is a relatively small sample size for a phase III trial and raises concern with replication in a larger more diverse samples and settings, as noted. The moderate rating would be more compelling if there were larger more diverse samples.Thank you for this comment. We reassessed our Strength of Evidence (SOE) rating for MDMA-assisted psychotherapy for PTSD (now rated as low SOE) accounting for the limitations that you and other reviewers highlighted.
597All MDMA studies were funded/sponsored by MAPS raising concern re bias.In the Result section, we state that all studies were funded or sponsored by MAPS.
607The lack of current MDMA evidence with military veterans’ seeking mental health treatment in a VA setting is perhaps the largest concern. Given the medical and psychiatric complexity of the veteran mental health treatment seeking population, this is a significant gap. This current evidence cannot be generalized to this important population.Thank you for these comments. Please see our responses to Comment #27 above, which address similar concerns raised by Reviewer 1.
617Given these important concerns I believe a strong cautionary statement should be included regarding the moderate favorable evidence for MDMA-assisted therapy for PTSD.Thank you for this comment. As stated above, we reassessed our Strength of Evidence (SOE) rating for MDMA-assisted psychotherapy for PTSD (now rated as low SOE) accounting for the limitations that you and other reviewers highlighted. We also provide more context regarding generalizability and emphasize that findings are preliminary.
628Minor edit needed on page 2 - I believe that “PTSD-assisted psychotherapy” should be changed to “MDMA-assisted psychotherapy for PTSD”Change made.

APPENDIX E. RESEARCH IN PROGRESS

StatusStudy TitleStudy DesignInformation Resources
Clinical Trials
Substance Use Disorder
Not yet recruitingSafety and Feasibility of Psilocybin in Methamphetamine Use Disorder in a Community-Based SamplePre-post study ClinicalTrials.gov Identifier: NCT05322954
Not yet recruitingA Randomized, Double-Blind Study of Psilocybin for Opioid Use Disorder in Patients on Methadone Maintenance with Ongoing Opioid UseCrossover RCT ClinicalTrials.gov Identifier: NCT05242029
Not yet recruitingPsilocybin-Enhanced Psychotherapy for Methamphetamine Use DisorderRCT ClinicalTrials.gov Identifier: NCT04982796
Not yet recruitingPsilocybin for Treatment of Alcohol Use Disorder: a Feasibility StudyPre-post study ClinicalTrials.gov Identifier: NCT04718792
Active, not recruitingPilot Trial of Visual Healing®, a Nature-themed Virtual Immersive Experience, to Optimize Set and Setting in Psilocybin-assisted Therapy for Alcohol Use DisorderRCT ClinicalTrials.gov Identifier: NCT04410913
RecruitingPhase I Study of the Safety and Adjunctive Effects of Psilocybin in Adults with Opioid Use Disorder Maintained on Buprenorphine/NaloxonePre-post study ClinicalTrials.gov Identifier: NCT04161066
UnknownOpen-Label Proof of Concept Feasibility Study to Explore the Safety, Tolerability and Potential Role of MDMA-Assisted Psychotherapy for the Treatment of Detoxified Patients with Alcohol Use DisorderPre-post study ClinicalTrials.gov Identifier: NCT04158778
RecruitingPhase II, Randomized, Double Blind, Placebo Controlled, Parallel Group, Single Center Study of Psilocybin Efficacy and Mechanism in Alcohol Use DisorderRCT ClinicalTrials.gov Identifier: NCT04141501
RecruitingPreliminary Efficacy and Safety of Ibogaine in the Treatment of MethadoneRCT ClinicalTrials.gov Identifier: NCT04003948
Not yet recruitingTolerability and Efficacy of Ibogaine in the Treatment of Alcoholism: The First Randomized, Double-blind, Placebo-controlled, Escalating-dose, Phase 2 TrialRCT ClinicalTrials.gov Identifier: NCT03380728
RecruitingPsilocybin-facilitated Treatment for Cocaine Use: A Pilot StudyRCT ClinicalTrials.gov Identifier: NCT02037126
RecruitingPsilocybin-facilitated Smoking Cessation Treatment: A Pilot StudyRCT ClinicalTrials.gov Identifier: NCT01943994
UnknownEffects and Therapeutic Potential of Psilocybin in Alcohol DependencePre-post study ClinicalTrials.gov Identifier: NCT01534494
RecruitingIbogaine to Determine Maximum Tolerated Dose (MTD) or Treat-to-Target Dose (TTD) for the Evaluation of Efficacy and SafetyRCT ClinicalTrials.gov Identifier: NCT05029401
Anxiety Disorders
Not yet recruitingEffects of Repeated Dosing of Psilocybin on Obsessive-Compulsive Disorder: A Randomized, Waitlist-Controlled StudyRCT ClinicalTrials.gov Identifier: NCT05370911
RecruitingSocial Anxiety MDMA-Assisted Therapy Investigation (SAMATI): A Randomized, Delayed Treatment Control Phase 2 Study of the Safety and Effectiveness of Manualized MDMA-Assisted Therapy for the Treatment of Social Anxiety DisorderRCT ClinicalTrials.gov Identifier: NCT05138068
Not yet recruitingOpen Label, Phase 1 Study for Evaluating the Feasibility, Safety and Efficacy of Psychotherapy Assisted Psilocybin for Treatment of Severe OCDPre-post study ClinicalTrials.gov Identifier: NCT04882839
RecruitingPsilocybin Treatment in Obsessive-Compulsive Disorder: a Preliminary Efficacy Study and Exploratory Investigation of Neural Correlates.RCT ClinicalTrials.gov Identifier: NCT03356483
CompletedLSD Treatment in Persons Suffering from Anxiety Symptoms in Severe Somatic Diseases or in Psychiatric Anxiety Disorders: a Randomized, Double-blind, Placebo-controlled Phase II StudyCrossover RCT ClinicalTrials.gov Identifier: NCT03153579
Mood Disorders
Not yet recruitingAn Open Label Study of Single-Dose Psilocybin for Major Depressive Disorder with Co-occurring Borderline Personality DisorderPre-post study ClinicalTrials.gov Identifier: NCT05399498
Not yet recruitingA Phase I/IIa, Randomized, Double-Blind, Placebo-Controlled Study to Assess Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of Multiple Ascending Oral Doses of CYB003 in Participants with Major Depressive DisorderRCT ClinicalTrials.gov Identifier: NCT05385783
RecruitingPsilocybin Versus Ketamine - Fast Acting Antidepressant Strategies in Treatment-resistant DepressionRCT ClinicalTrials.gov Identifier: NCT05383313
Not yet recruitingThe Effects of Psilocybin on Self-Focus and Self-Related Processing in Treatment Resistant MDDPre-post study ClinicalTrials.gov Identifier: NCT05381974
Not yet recruitingMicrodosing Psychedelics to Improve MoodCrossover RCT ClinicalTrials.gov Identifier: NCT05259943
Not yet recruitingPsilocybin-assisted Cognitive Behavioral Therapy for DepressionPre-post study ClinicalTrials.gov Identifier: NCT05227612
RecruitingAn Open-Label Pilot Study Examining the Feasibility, Safety, and Effectiveness of Psilocybin Therapy for Depression in Bipolar II DisorderPre-post study ClinicalTrials.gov Identifier: NCT05065294
Active, not recruitingThe Efficacy and Tolerability of Psilocybin in Participants With Treatment-Resistant Depression: a Phase 2, Randomized Feasibility StudyRCT ClinicalTrials.gov Identifier: NCT05029466
Not yet recruitingRandomized Double Blind Placebo Controlled Assessing the Efficacy of Micro-dosed Psilocybin in Reducing Anxiety and or Depression Levels in AdultsRCT ClinicalTrials.gov Identifier: NCT04989972
CompletedThe Safety and Efficacy Of Psilocybin as an Adjunctive Therapy in Participants With Treatment Resistant DepressionPre-post study ClinicalTrials.gov Identifier: NCT04739865
Active, not recruitingFixed Order, Open-Label, Dose-Escalation Study of DMT in HumansNon-randomized crossover trial ClinicalTrials.gov Identifier: NCT04711915
CompletedA Phase 1/2 Study of GH001 in Patients With Treatment-Resistant DepressionNon-randomized sequential trial ClinicalTrials.gov Identifier: NCT04698603
RecruitingA Double-blind, Randomised, Placebo-controlled Study of Intravenous Doses of SPL026 (DMT Fumarate), a Serotonergic Psychedelic, in Healthy Subjects (Part A) and Patients With Major Depressive Disorder (Part B)RCT ClinicalTrials.gov Identifier: NCT04673383
RecruitingA Phase II Randomized, Double-blind, Active Placebo-controlled Parallel Group Trial to Examine the Efficacy and Safety of Psilocybin in Treatment-resistant Major DepressionRCT ClinicalTrials.gov Identifier: NCT04670081
RecruitingThe Effect of Psilocybin on MDD Symptom Severity and Synaptic Density - A Single Dose Randomized, Double Blind, Placebo-Controlled Phase 2 Positron Emission Tomography StudyRCT ClinicalTrials.gov Identifier: NCT04630964
RecruitingPsilocybin Treatment of Major Depressive Disorder With Co-occurring Alcohol Use DisorderRCT ClinicalTrials.gov Identifier: NCT04620759
RecruitingMulticentre Study To Assess Safety And Efficacy Of Psilocybin In Patients With Treatment-Resistant Depression Following Completion Of COMP 001 And COMP 003 Trials (P-TRD LTFU)Prospective cohort ClinicalTrials.gov Identifier: NCT04519957
RecruitingAn Open Label Study of the Safety and Efficacy of Psilocybin in Participants With Treatment-Resistant Depression (P-TRD)Pre-post study ClinicalTrials.gov Identifier: NCT04433858
RecruitingThe Safety and Efficacy of Psilocybin in Participants With Type 2 Bipolar Disorder (BP-II) DepressionPre-post study ClinicalTrials.gov Identifier: NCT04433845
Enrolling by invitationA Two-Year Observational Follow-up Study of Subjects With Major Depressive Disorder Following a Randomized, Double-Blind Single-Dose of Psilocybin or Niacin-ControlCohort (long-term follow-up) ClinicalTrials.gov Identifier: NCT04353921
Active, not recruitingLSD Therapy for Persons Suffering From Major Depression: A Randomised, Double-blind, Active-placebo Controlled Phase II StudyRCT ClinicalTrials.gov Identifier: NCT03866252
CompletedThe Safety and Efficacy of Psilocybin in Participants With Treatment Resistant DepressionRCT ClinicalTrials.gov Identifier: NCT03775200
CompletedPhase II, Randomized, Double Blind, Placebo Controlled, Parallel Group, Single Center Study of Psilocybin Efficacy in Major DepressionRCT ClinicalTrials.gov Identifier: NCT03715127
Active, not recruitingPsilocybin - Induced Neuroplasticity in the Treatment of Major Depressive DisorderCrossover RCT ClinicalTrials.gov Identifier: NCT03554174
UnknownPsilocybin and Depression - Assessing the Long-term Effects of a Single Administration of Psilocybin on the Psychiatric Symptoms and Brain Activity of Patients With Severe DepressionRCT ClinicalTrials.gov Identifier: NCT03380442
PTSD
RecruitingThe Safety and Tolerability of COMP360 in Participants With Post-traumatic Stress DisorderPre-post study ClinicalTrials.gov Identifier: NCT05312151
RecruitingSustaining Remission From Posttraumatic Stress Disorder (PTSD) Using Tuned Vibroacoustic Stimulation (TVS) Following MDMA-Assisted PsychotherapyPre-post study ClinicalTrials.gov Identifier: NCT05274230
Not yet recruitingInvestigating the Therapeutic Effects of Psilocybin in Treatment-Resistant Post-TraumaticPre-post study ClinicalTrials.gov Identifier: NCT05243329
Not yet recruitingMDMA-Assisted Therapy for Postpartum People With Opioid Use Disorder and Coexisting Post Traumatic Stress DisorderPre-post study ClinicalTrials.gov Identifier: NCT05219175
Not yet recruitingAn Open-Label Feasibility and Safety Study of MDMA-Assisted Group Therapy for the Treatment of Posttraumatic Stress Disorder in VeteransPre-post study ClinicalTrials.gov Identifier: NCT05173831
SuspendedA Phase 2 Open-Label Treatment Development Study of MDMA-Assisted Cognitive Processing Therapy (CPT) for Posttraumatic Stress Disorder (PTSD)Pre-post study ClinicalTrials.gov Identifier: NCT05067244
Enrolling by InvitationLong-Term Safety and Persistence of Effectiveness of Manualized MDMA-Assisted Therapy for the Treatment of Posttraumatic Stress DisorderRetrospective cohort ClinicalTrials.gov Identifier: NCT05066282
WithdrawnA Phase 2, Open Label Study of the Safety and Effectiveness of MDMA-assisted Therapy for Participants With Posttraumatic Stress DisorderPre-post study ClinicalTrials.gov Identifier: NCT04968938
RecruitingA Phase 2, Open-Label, Randomized Comparative Effectiveness Study for MDMA-Assisted Psychotherapy in U.S. Veterans With Chronic PTSDRCT ClinicalTrials.gov Identifier: NCT04784143
Enrolling by invitationA Multi-Site Open-Label Safety Extension Study of Manualized MDMA-Assisted Psychotherapy for the Treatment of Participants With Posttraumatic Stress DisorderPre-post study ClinicalTrials.gov Identifier: NCT04714359
AvailableA Multi-site Expanded Access Program for MDMA-assisted Psychotherapy for Patients With Treatment-resistant PTSDExpanded access protocol ClinicalTrials.gov Identifier: NCT04438512
RecruitingOpen-label Phase 2 Study of MDMA-Assisted Psychotherapy in Veterans With Combat-Related, Refractory PTSDPre-post study ClinicalTrials.gov Identifier: NCT04264026
Active, not recruitingA Randomized, Double-Blind, Placebo-Controlled, Multi-Site Phase 3 Study of the Efficacy and Safety of Manualized MDMA-Assisted Psychotherapy for the Treatment of Posttraumatic Stress Disorder of Moderate or Greater SeverityRCT ClinicalTrials.gov Identifier: NCT04077437
RecruitingAn Open-Label, Phase 2, Multicenter Feasibility Study of Manualized MDMA-Assisted Psychotherapy With an Optional fMRI Sub-Study Assessing Changes in Brain Activity in Subjects With Posttraumatic Stress DisorderPre-post study ClinicalTrials.gov Identifier: NCT04030169
CompletedAn Open-Label, Multi-Site Phase 2 Study of the Safety and Effect of Manualized MDMA-Assisted Psychotherapy for the Treatment of Severe Posttraumatic Stress Disorder (Canada)Pre-post study ClinicalTrials.gov Identifier: NCT03485287
CompletedAn Open-Label, Multi-Site Phase 2 Study of the Safety and Effect of Manualized MDMA-Assisted Therapy for the Treatment of Severe Posttraumatic Stress DisorderPre-post study ClinicalTrials.gov Identifier: NCT03282123
CompletedA Phase 1/2 Open-Label Treatment Development Study of Methylenedioxymethamphetamine (MDMA)-Assisted Cognitive-Behavioral Conjoint Therapy (CBCT) in Dyads in Which 1 Member Has Chronic Posttraumatic Stress Disorder (PTSD)Pre-post study ClinicalTrials.gov Identifier: NCT02876172
CompletedAn Open-Label Proof-of-Principle Study Testing the Use of an Additional MDMA-Assisted Therapy Session in People Who Relapsed After Participating in a Phase 2 Clinical Trial of MDMA-Assisted Therapy to Treat Chronic, Treatment-Resistant Posttraumatic Stress Disorder (PTSD)Pre-post study ClinicalTrials.gov Identifier: NCT01458327
TerminatedMDMA-assisted Therapy in Twelve People With War and Terrorism-related Posttraumatic Stress Disorder (PTSD)RCT ClinicalTrials.gov Identifier: NCT00402298
Eating Disorders
RecruitingA Phase 2a Safety and Feasibility Study Evaluating Psilocybin (TRP-8802) Administration in Concert With Psychotherapy in the Treatment of Binge Eating DisorderPre-post study ClinicalTrials.gov Identifier: NCT05035927
Active, not recruitingEvaluation of Psilocybin in Anorexia Nervosa: Safety and EfficacyPre-post study ClinicalTrials.gov Identifier: NCT04661514
Active, not recruitingSafety and Efficacy of Psilocybin for Body Dysmorphic DisorderPre-post study ClinicalTrials.gov Identifier: NCT04656301
Not yet recruitingAn Open-Label, Multi-Site Phase 2 Study of the Safety and Feasibility of MDMA-Assisted Psychotherapy for Eating DisordersNon-randomized controlled trial ClinicalTrials.gov Identifier: NCT04454684
RecruitingEffects of Psilocybin in Anorexia NervosaPre-post study ClinicalTrials.gov Identifier: NCT04052568
Other
RecruitingSafety and Efficacy of Repeated Low Dose MM-120 as Treatment for Attention Deficit Disorder (ADHD) in Adults: a Multi-center, Randomized, Double-blind, Placebo-controlled Phase 2a Proof of Concept TrialRCT ClinicalTrials.gov Identifier: NCT05200936
RecruitingThe Safety and Efficacy of Psilocybin in Patients With Treatment-resistant Depression and Chronic Suicidal IdeationPre-post study ClinicalTrials.gov Identifier: NCT05220410

Notes. MM-120 is a form of LSD. SPL026 is also known as DMT Fumarate.

Abbreviations. ADHD=attention-deficit/hyperactivity disorder; BP-II=bipolar disorder II; CPT=cognitive processing therapy; DMT=5-methoxy-N,N-dimethyltryptamine; fMRI=functional magnetic resonance imaging; GH001=5 methoxy N,N dimethyltryptamine; LSD=lysergic acid diethylamide; LTFU=long-term follow up; MDD=major depressive disorder; MDMA=3,4-methylenedioxy-methamphetamine; MTD=maximum tolerated dose; OCD=obsessive compulsive disorder; RCT=randomized controlled trial; SAMATI=social anxiety MDMA-assisted therapy investigation; TRD=treatment resistant depression; TTD=treat-to-target dose; TVS=tuned vibroacoustic stimulation.

REFERENCES

1.
Bogenschutz MP, Ross S, Bhatt S, et al. Percentage of Heavy Drinking Days Following Psilocybin-Assisted Psychotherapy vs Placebo in the Treatment of Adult Patients With Alcohol Use Disorder: A Randomized Clinical Trial. JAMA Psychiatry. 2022. [PMC free article: PMC9403854] [PubMed: 36001306]
2.
Carhart-Harris R, Giribaldi B, Watts R, et al. Trial of Psilocybin versus Escitalopram for Depression. New England Journal of Medicine. 2021;384(15):1402–1411. [PubMed: 33852780]
3.
Davis AK, Barrett FS, May DG, et al. Effects of Psilocybin-Assisted Therapy on Major Depressive Disorder: A Randomized Clinical Trial. JAMA Psychiatry. 2021;78(5):481–489. [PMC free article: PMC7643046] [PubMed: 33146667]
4.
Gukasyan N, Davis AK, Barrett FS, et al. Efficacy and safety of psilocybin-assisted treatment for major depressive disorder: Prospective 12-month follow-up. Journal of Psychopharmacology. 2022;36(2):151–158. [PMC free article: PMC8864328] [PubMed: 35166158]
5.
Palhano-Fontes F, Barreto D, Onias H, et al. Rapid antidepressant effects of the psychedelic ayahuasca in treatment-resistant depression: a randomized placebo-controlled trial. Psychological Medicine. 2019;49(4):655–663. [PMC free article: PMC6378413] [PubMed: 29903051]
6.
Zeifman RJ, Palhano-Fontes F, Hallak J, Arcoverde E, Maia-Oliveira JP, Araujo DB. The Impact of Ayahuasca on Suicidality: Results From a Randomized Controlled Trial. Frontiers in Pharmacology. 2019;10. [PMC free article: PMC6878725] [PubMed: 31798447]
7.
Glue P, Cape G, Tunnicliff D, et al. Ascending Single-Dose, Double-Blind, Placebo-Controlled Safety Study of Noribogaine in Opioid-Dependent Patients. Clinical Pharmacology in Drug Development. 2016;5(6):460–468. [PubMed: 27870477]
8.
Bouso JC, Doblin R, Farre M, Alcazar MA, Gomez-Jarabo G. MDMA-assisted psychotherapy using low doses in a small sample of women with chronic posttraumatic stress disorder. Journal of Psychoactive Drugs. 2008;40(3):225–236. [PubMed: 19004414]
9.
Mitchell JM, Bogenschutz M, Lilienstein A, et al. MDMA-assisted therapy for severe PTSD: a randomized, double-blind, placebo-controlled phase 3 study. Nature Medicine. 2021;27(6):1025–1033. [PMC free article: PMC8205851] [PubMed: 33972795]
10.
Mithoefer MC, Wagner MT, Mithoefer AT, Jerome L, Doblin R. The safety and efficacy of {+/-}3,4-methylenedioxymethamphetamine-assisted psychotherapy in subjects with chronic, treatment-resistant posttraumatic stress disorder: the first randomized controlled pilot study. Journal of Psychopharmacology. 2011;25(4):439–452. [PMC free article: PMC3122379] [PubMed: 20643699]
11.
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. Journal of Psychopharmacology. 2013;27(1):28–39. [PMC free article: PMC3573678] [PubMed: 23172889]
12.
Mithoefer MC, Mithoefer AT, Feduccia AA, et al. 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy for post-traumatic stress disorder in military veterans, firefighters, and police officers: a randomised, double-blind, dose-response, phase 2 clinical trial. The Lancet Psychiatry. 2018;5(6):486–497. [PubMed: 29728331]
13.
Mithoefer MC, Feduccia AA, Jerome L, et al. MDMA-assisted psychotherapy for treatment of PTSD: study design and rationale for phase 3 trials based on pooled analysis of six phase 2 randomized controlled trials. Psychopharmacology. 2019;236(9):2735–2745. [PMC free article: PMC6695343] [PubMed: 31065731]
14.
Oehen P, Traber R, Widmer V, Schnyder U. A randomized, controlled pilot study of MDMA (+/− 3,4-Methylenedioxymethamphetamine)-assisted psychotherapy for treatment of resistant, chronic Post-Traumatic Stress Disorder (PTSD). Journal of Psychopharmacology. 2013;27(1):40–52. [PubMed: 23118021]
15.
Ot’alora GM, Grigsby J, Poulter B, et al. 3,4-Methylenedioxymethamphetamine-assisted psychotherapy for treatment of chronic posttraumatic stress disorder: A randomized phase 2 controlled trial. Journal of Psychopharmacology. 2018;32(12):1295–1307. [PMC free article: PMC6247454] [PubMed: 30371148]
16.
Dos Santos RG, Osorio FL, Rocha JM, et al. Ayahuasca Improves Self-perception of Speech Performance in Subjects With Social Anxiety Disorder: A Pilot, Proof-of-Concept, Randomized, Placebo-Controlled Trial. Journal of Clinical Psychopharmacology. 2021;41(5):540–550. [PubMed: 34166299]
17.
Bogenschutz MP, Forcehimes AA, Pommy JA, Wilcox CE, Barbosa PC, Strassman RJ. Psilocybin-assisted treatment for alcohol dependence: a proof-of-concept study. Journal of Psychopharmacology. 2015;29(3):289–299. [PubMed: 25586396]
18.
Sessa B, Aday JS, O’Brien S, et al. Debunking the myth of ‘Blue Mondays’: No evidence of affect drop after taking clinical MDMA. Journal of Psychopharmacology. 2022;36(3):360–367. [PubMed: 34894842]
19.
Carhart-Harris RL, Bolstridge M, Rucker J, et al. Psilocybin with psychological support for treatment-resistant depression: an open-label feasibility study. The Lancet Psychiatry. 2016;3(7):619–627. [PubMed: 27210031]
20.
Carhart-Harris RL, Bolstridge M, Day CMJ, et al. Psilocybin with psychological support for treatment-resistant depression: six-month follow-up. Psychopharmacology. 2018;235(2):399–408. [PMC free article: PMC5813086] [PubMed: 29119217]
21.
Lyons T, Carhart-Harris RL. More realistic forecasting of future life events after psilocybin for treatment-resistant depression. Frontiers in Psychology. 2018;9. [PMC free article: PMC6194345] [PubMed: 30369890]
22.
Osorio L, Sanches RF, Macedo LR, et al. Antidepressant effects of a single dose of ayahuasca in patients with recurrent depression: a preliminary report. Revista Brasileira de Psiquiatria. 2015;37(1):13–20. [PubMed: 25806551]
23.
Sanches RF, de Lima Osorio F, Dos Santos RG, et al. Antidepressant Effects of a Single Dose of Ayahuasca in Patients With Recurrent Depression: A SPECT Study. Journal of Clinical Psychopharmacology. 2016;36(1):77–81. [PubMed: 26650973]
24.
Zeifman RJ, Singhal N, Dos Santos RG, et al. Rapid and sustained decreases in suicidality following a single dose of ayahuasca among individuals with recurrent major depressive disorder: results from an open-label trial. Psychopharmacology. 2021;238(2):453–459. [PubMed: 33118052]
25.
Stroud JB, Freeman TP, Leech R, et al. Psilocybin with psychological support improves emotional face recognition in treatment-resistant depression. Psychopharmacology. 2018;235(2):459–466. [PMC free article: PMC5813058] [PubMed: 29085980]
26.
Moreno FA, Wiegand CB, Taitano EK, Delgado PL. Safety, tolerability, and efficacy of psilocybin in 9 patients with obsessive-compulsive disorder. Journal of Clinical Psychiatry. 2006;67(11):1735–1740. [PubMed: 17196053]
27.
Brown TK, Alper K. Treatment of opioid use disorder with ibogaine: detoxification and drug use outcomes. American Journal of Drug & Alcohol Abuse. 2018;44(1):24–36. [PubMed: 28541119]
28.
Davis AK, Barsuglia JP, Windham-Herman A-M, Lynch M, Polanco M. Subjective effectiveness of ibogaine treatment for problematic opioid consumption: Short- and long-term outcomes and current psychological functioning. Journal of Psychedelic Studies. 2017;1(2):65–73. [PMC free article: PMC6157925] [PubMed: 30272050]
29.
Knuijver T, Schellekens A, Belgers M, et al. Safety of ibogaine administration in detoxification of opioid-dependent individuals: a descriptive open-label observational study. Addiction. 2022;117(1):118–128. [PMC free article: PMC9292417] [PubMed: 33620733]
30.
Malcolm BJ, Polanco M, Barsuglia JP. Changes in Withdrawal and Craving Scores in Participants Undergoing Opioid Detoxification Utilizing Ibogaine. Journal of Psychoactive Drugs. 2018;50(3):256–265. [PubMed: 29608409]
31.
Noller GE, Frampton CM, Yazar-Klosinski B. Ibogaine treatment outcomes for opioid dependence from a twelve-month follow-up observational study. American Journal of Drug & Alcohol Abuse. 2018;44(1):37–46. [PubMed: 28402682]
32.
Jardim AV, Jardim DV, Chaves BR, et al. 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy for victims of sexual abuse with severe post-traumatic stress disorder: an open label pilot study in Brazil. Revista Brasileira de Psiquiatria. 2021;43(2):181–185. [PMC free article: PMC8023155] [PubMed: 32638920]
33.
Monson CM, Wagner AC, Mithoefer AT, et al. MDMA-facilitated cognitive-behavioural conjoint therapy for posttraumatic stress disorder: An uncontrolled trial. European Journal of Psychotraumatology. 2020;11(1):1840123. [PMC free article: PMC7748057] [PubMed: 33408811]
34.
Wang JB, Lin J, Bedrosian L, et al. Scaling Up: Multisite Open-Label Clinical Trials of MDMA-Assisted Therapy for Severe Posttraumatic Stress Disorder. Journal of Humanistic Psychology. 2021;0(0):00221678211023663.
35.
Thomas G, Lucas P, Capler NR, Tupper KW, Martin G. Ayahuasca-assisted therapy for addiction: results from a preliminary observational study in Canada. Current Drug Abuse Reviews. 2013;6(1):30–42. [PubMed: 23627784]
36.
Mash DC, Kovera CA, Pablo J, et al. Ibogaine: complex pharmacokinetics, concerns for safety, and preliminary efficacy measures. Annals of the New York Academy of Sciences. 2000;914:394–401. [PubMed: 11085338]
37.
Mash DC, Kovera CA, Pablo J, et al. Ibogaine in the treatment of heroin withdrawal. The Alkaloids Chemistry & Biology. 2001;56:155–171. [PubMed: 11705106]
38.
Mash DC, Duque L, Page B, Allen-Ferdinand K. Ibogaine Detoxification Transitions Opioid and Cocaine Abusers Between Dependence and Abstinence: Clinical Observations and Treatment Outcomes. Front Pharmacol. 2018;9:529. [PMC free article: PMC5996271] [PubMed: 29922156]
39.
Schenberg EE, de Castro Comis MA, Chaves BR, da Silveira DX. Treating drug dependence with the aid of ibogaine: a retrospective study. Journal of Psychopharmacology. 2014;28(11):993–1000. [PubMed: 25271214]
40.
Johnson MW, Garcia-Romeu A, Cosimano MP, Griffiths RR. Pilot study of the 5-HT2AR agonist psilocybin in the treatment of tobacco addiction. Journal of Psychopharmacology. 2014;28(11):983–992. [PMC free article: PMC4286320] [PubMed: 25213996]
41.
Johnson MW, Garcia-Romeu A, Griffiths RR. Long-term follow-up of psilocybin-facilitated smoking cessation. The American Journal of Drug and Alcohol Abuse. 2017;43(1):55–60. [PMC free article: PMC5641975] [PubMed: 27441452]
42.
Davis AK, Averill LA, Sepeda ND, Barsuglia JP, Amoroso T. Psychedelic treatment for trauma-related psychological and cognitive impairment among US Special Operations Forces Veterans. Chronic Stress. 2020;4:2470547020939564. [PMC free article: PMC7359647] [PubMed: 32704581]
43.
Sessa B, Higbed L, O’Brien S, et al. First study of safety and tolerability of 3,4-methylenedioxymethamphetamine-assisted psychotherapy in patients with alcohol use disorder. Journal of Psychopharmacology. 2021;35(4):375–383. [PubMed: 33601929]
44.
Kotler M. A Randomized, Double-Blind, Active Placebo-Controlled Phase 2 Pilot Study of MDMA-assisted Psychotherapy in People With Chronic, Treatment-Resistant Posttraumatic Stress Disorder (PTSD). NCT01689740. Multidisplinary Association for Psychedelic Studies. https:​//clinicaltrials​.gov/ct2/show/record​/NCT01689740?term=NCT01689740&draw​=2&rank=1. Published 2017. Accessed August 22, 2022.
45.
Pacey I. A Randomized, Double-Blind, Controlled Phase 2 Pilot Study of Manualized 3,4-methylenedioxymethamphetamine (MDMA)-Assisted Psychotherapy in 12 Subjects With Treatment-Resistant Posttraumatic Stress Disorder (PTSD) - Canada. NCT01958593. Multidisciplinary Association for Psychedelic Studies. https:​//clinicaltrials​.gov/ct2/show/record​/NCT01958593?term=NCT01958593&draw​=2&rank=1. Published 2017. Accessed August 22, 2022.

Suggested citation:

Mackey KM, Anderson JK, Williams BE, Ward RM, Parr NJ. Evidence Brief: Psychedelic Medications for Mental Health and Substance Use Disorders. Washington, DC: Evidence Synthesis Program, Health Services Research and Development Service, Office of Research and Development, Department of Veterans Affairs. VA ESP Project #09-199; 2022.

This report was prepared by the Evidence Synthesis Program Coordinating Center located at the VA Portland Health Care System, directed by Mark Helfand, MD, MPH, MS and funded by the Department of Veterans Affairs, Veterans Health Administration, Health Services Research and Development.

The findings and conclusions in this document are those of the author(s) who are responsible for its contents and do not necessarily represent the views of the Department of Veterans Affairs or the United States government. Therefore, no statement in this article should be construed as an official position of the Department of Veterans Affairs. No investigators have any affiliations or financial involvement (eg, employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties) that conflict with material presented in the report.

Copyright Notice

This publication is in the public domain and is therefore without copyright. All text from this work may be reprinted freely. Use of these materials should be acknowledged.

Bookshelf ID: NBK586533PMID: 36413606

Views

Other titles in this collection

Related information

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...