Minireviews Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Psychiatry. May 19, 2023; 13(5): 182-190
Published online May 19, 2023. doi: 10.5498/wjp.v13.i5.182
Therapeutic role of psilocybin and 3,4-methylenedioxymethamphetamine in trauma: A literature review
Lakshan N Fonseka, Harvard South Shore-Psychiatry Residency Program, Veteran Affairs Boston Healthcare System, Brockton, MA 02301, United States
Benjamin KP Woo, Chinese American Health Promotion Laboratory, University of California, Los Angeles, Los Angeles, CA 90095, United States
Benjamin KP Woo, Department of Psychiatry and Biobehavioral Sciences, Olive View - University of California, Los Angeles Medical Center, Sylmar, CA 91342, United States
Benjamin KP Woo, Asian American Studies Center, University of California, Los Angeles, Los Angeles, CA 90095, United States
Benjamin KP Woo, Podiatric Medicine and Surgery, Western University of Health Sciences, Pomona, CA 91766, United States
ORCID number: Lakshan N Fonseka (0000-0003-0073-690X); Benjamin KP Woo (0000-0002-0127-4850).
Author contributions: Fonseka LN and Woo BKP both performed the collection of data and contributed to the manuscript drafting; All authors have read and approve the final manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Benjamin KP Woo, MD, Professor, Chinese American Health Promotion Laboratory, University of California, Los Angeles, 3230 Campbell Hall, Los Angeles, CA 90095, United States. bkpwoo@ucla.edu
Received: January 19, 2023
Peer-review started: January 19, 2023
First decision: February 20, 2023
Revised: February 28, 2023
Accepted: April 13, 2023
Article in press: April 13, 2023
Published online: May 19, 2023

Abstract

With the Food and Drug Administration designation in 2017 of 3,4-methylenedioxymethamphetamine (MDMA) as a breakthrough therapy in post-traumatic stress disorder and psilocybin in treatment-resistant depression, psychedelic drugs have continued to garner the attention of researchers and clinicians for their promise of unmatched, rapid improvement in a multitude of psychiatric conditions. Classic psychedelic drugs including psilocybin, lysergic acid diethylamide, and ayahuasca, as well as non-classic drugs such as MDMA and ketamine, are currently being investigated for a potential therapeutic role in trauma, depressive disorders, and other psychopathologies. However, psilocybin and MDMA each have a functional profile well-suited for integration with psychotherapy. The present review focuses on psilocybin and MDMA in psychedelic-assisted therapy (PAT), as these studies compose most of the literature pool. In this review, we discuss the current and future uses of psychedelic drugs, with an emphasis on the role of MDMA and psilocybin in PAT in the setting of trauma and related comorbidities on the efficacy of psychedelic drugs across multiple psychiatric disorders. The article concludes with thoughts for future research, such as incorporating wearables and standardization of symptom scales, therapy styles, and assessment of adverse drug reactions.

Key Words: Psychedelics, Trauma, Depression, Methylenedioxymethamphetamine, Ecstasy, Psilocybin

Core Tip: Psychedelic-assisted therapy with psilocybin or 3,4-methylenedioxymethamphetamine (MDMA) is strongly supportive across psychiatric conditions, especially trauma and related comorbidities, as demonstrated through a pattern of rapid and sustained symptom relief. Both treatments seem to have benefits beyond the Food and Drug Administration breakthrough designations of treatment-resistant depression for psilocybin and post-traumatic stress disorder for MDMA.



INTRODUCTION

The psychedelic renaissance of modern psychiatry can trace its resurgence to 2017-2018, when the Food and Drug Administration (FDA) designated breakthrough therapy status to psilocybin in treatment-resistant depression and 3,4-methylenedioxymethamphetamine (MDMA) in post-traumatic stress disorder (PTSD). Psychedelics can be broadly categorized as classic psychedelics that act as agonists at the 5-HT2A receptor [i.e., psilocybin, lysergic acid diethylamide (LSD), ayahuasca/dimethyltryptamine], empathogens that increase serotonin levels (i.e., MDMA), antagonists at N-methyl-D-aspartate receptors (i.e., ketamine), and atypical hallucinogens with effects across neurotransmitter systems[1,2]. Psychedelic-assisted psychotherapy (PAT) involving MDMA or psilocybin composes most of the literature pool, though studies involving other psychopathologies and compounds have also been investigated to a lesser extent. The predominance of both psilocybin and MDMA in the literature may be attributed to the properties that make each an ideal adjunct to psychotherapy.

Psilocybin and its active metabolite psilocin are derived from psychoactive mushroom species originally used in ceremonial settings to facilitate spiritual experiences in Central and South America, later introduced to Western culture in the 1950s[1,3]. As a classic psychedelic, psilocybin is a 5-HT2A receptor agonist that was found to produce rapid yet enduring improvement in treatment-resistant depression and major depressive disorder (MDD)[4-6]. More recently, psilocybin was compared to escitalopram in a double-blind trial. Both treatments were found to have similar efficacy in depression, which will be discussed in more detail in the present article[7].

Psilocybin further enhances its synergism with psychotherapy by evoking a sense of unity, ego-dissolution, and awe through “mystical” experiences[8-10]. As described by Vaid et al[11] (2022), PAT may enable the processing of emotional experiences that were previously inaccessible due to trauma blocks. This promotes reconnection with the self and “foundational identity deficit repair,” a re-stabilization of the core ideas that together represent the conscious identity, self-esteem, and other aspects of the self[11]. As will be described later in this article, patients in recent studies report significant reductions in symptom rating scales, with benefits partly attributed to this mystical component-experiences of oneness or ego-dissolution, connection to self-essence, and a broadened perspective beyond self-imposed limits on thought processes and rigid mental frameworks that may once have been adaptive responses to life events[12,13].

MDMA was initially developed by Merck & Co. for hemostasis in 1912, and its psychoactive effects were not published until 1978. After becoming popularized as recreational drug “ecstasy” in the 1980s[1,14,15], the Drug Enforcement Agency cited concerns of abuse potential and neurotoxicity and assigned schedule I status to MDMA in 1985. MDMA is part of a class of psychedelics, termed empathogens or enactogens, that increase empathy and social connection. Its effects are primarily mediated by serotonergic activity, including serotonin/norepinephrine transporter reuptake inhibition and partial agonism of serotonin receptors (5-HT2A, 5-HT1A, 5-HT2C)[1]. By far, MDMA studies formed most of the research elicited by the PubMed search terms. The reasons for the clear bias in the literature and its consequences are varied and discussed in the future directions section. Nevertheless, the research has yielded promising results across psychopathologies, though most research aligns with its FDA designation for PTSD. A recent double-blind, phase 3 clinical trial found that 67% of participants receiving MDMA-assisted psychotherapy no longer met PTSD criteria after 2 mo[16]. These improvements are attributed to several downstream changes in cognition that further complement psychotherapy.

MDMA has been previously shown to decrease amygdala response when patients are presented with angry faces, as well as increase ventral striatum response when viewing happy faces, further supporting that MDMA reduces threat response, enhances reward, and facilitates positive social interactions[1,17,18]. MDMA has the reputation of generating feelings of closeness, connection, and emotional empathy, although similar features also exist in classic psychedelics[19,20]. However, MDMA uniquely offers augmentation of the therapeutic alliance, easefully lowering barriers and enabling the patient to process traumatic memories without feeling overwhelmed[1,21].

MDMA, psilocybin, and other psychedelics appear to induce a temporary period of increased neuroplasticity with associated changes in psychological flexibility[21,22]. Psilocybin studies have demonstrated increased neurogenesis, spinogenesis, and synaptogenesis that facilitate the reconditioning of fear memory and the reversal of stress-induced changes to the prefrontal cortex[23-25]. The plasticity counters the deficits in fear memory extinction seen in PTSD, alleviating distress from the cycle of avoidance and flashbacks associated with persistent traumatic memories[26-28]. Kéri et al[29] suggest that psychedelic-induced serotonin-glutamate interactions affect memory pathways responsible for memory destabilization and reconsolidation[29]. The neuroplastic and fear memory changes enrich the benefits drawn from combination with psychotherapy.

The present focus is on psychedelics involved in PAT, namely MDMA and psilocybin as these studies compose much of the literature pool. A literature review was conducted through PubMed database search of ((psilocybin) OR (MDMA)) AND (trauma). The search was also performed on Reference Citation Analysis (https://www.referencecitationanalysis.com/). The aim is to summarize the literature on psychedelic drugs, with an emphasis on the role of MDMA and psilocybin in PAT in the setting of trauma and related depressive disorders, from 2020 to 2022, bridging the gap from Reiff et al’s review encompassing articles from 2007 to 2019 on the efficacy of psychedelic drugs across multiple psychiatric disorders[1]. The article concludes with thoughts for future research, such as incorporating wearables and standardization of symptom scales, therapy styles, and assessment of adverse drug reactions.

PSILOCYBIN

With its FDA designation for treatment-resistant depression, it follows to begin the discussion of psilocybin with updates in depression, a common comorbidity in trauma-related conditions. Carhart-Harris et al[7] published a clinical trial comparing psilocybin vs escitalopram in 59 patients with MDD[7]. The psylocibin group (n = 30) received 25 mg of psilocybin at the start and a second dose at 3 wk, all while receiving 6 wk of daily placebo. The escitalopram group (n = 29) received 1 mg of psilocybin at the start and a second dose of 1 mg at 3 wk, with daily oral escitalopram 10 mg throughout the 6-wk study. The primary outcome measure was changes from baselines scores (range 0 to 27) on the quick inventory of depressive symptomatology (QIDS-SR-16), with a response defined as reduction in score of at least 50% and remission defined as a score of 5 or less.

A response occurred in 70% of the psilocybin group and 48% of the escitalopram group, while remission occurred in 57% of the psilocybin group and 28% of the escitalopram group. Although these differences do not reach significance, it is notable that both psilocybin and escitalopram appear, at a minimum, to have equivalent impacts. The lack of control group prevents the comparison of each treatment to a baseline population, but it is promising that the efficacy is similar between psilocybin and escitalopram. The authors report that while the initial trial design included a placebo group, this became too practically complex and expensive. Other limitations of the study include the duration, as escitalopram may require more time to display its full efficacy. Additionally, the patients in the trial were not from varying socioeconomic or ethnic backgrounds, limiting external validity. The study also included many secondary outcomes, including scores on other symptom scales, but these were not considered useful as the data were unadjusted for multiple comparisons.

Further analysis of the study was conducted by Murphy et al[30] regarding the influence of therapeutic alliance. It was found that increased strength of therapeutic alliance led to greater emotional breakthrough and mystical experiences across two PAT sessions[30]. Interestingly, the average symptom severity scores at baseline were in the range for moderate depression[7], highlighting an additional application for psilocybin beyond treatment-resistant depression.

In a patient population with moderate to severe MDD, Davis et al[31] performed an 8-wk intervention consisting of two psilocybin dosing sessions less than two weeks apart with supportive psychotherapy. Exclusion criteria illustrate the reduced the severity of depression within the sample population: Selected patients were screened to avoid current antidepressant use, past diagnosis with a psychotic disorder, serious suicide attempts, or prior psychiatric hospitalization. However, enrolled patients required a score of at least 17 on the GRID-Hamilton depression rating scale (GRID-HAMD), the scale used to evaluate outcomes in this study at weeks 1 and 4 post-treatment with psilocybin. A total of 27 patients were selected and randomized into an immediate-treatment group (weeks 1-4, n = 14) and delayed-treatment group (weeks 5-8, n = 12), which served as a waiting list control condition that later received PAT as well. At baseline, the mean GRID-HAMD score was 22.8 with SD 3.9. Patients receiving immediate treatment showed significant reductions at weeks 1 and 4, with mean scores returning at 8.0 (SD 7.1) during week 1 and 8.5 (SD 5.7) at week 4. Overall, 17 patients demonstrated reductions in GRID-HAMD scores of at least 50%, 14 patients went into remission, and 3 patients dropped out before completing the intervention. Although preliminary, the effect sizes seen with psilocybin in this trial are several times larger than that seen in psychotherapy or antidepressant monotherapy[31-33].

The efficacy of psilocybin seen in depression studies likely offers benefits in a trauma-centered approach, due to the widespread comorbidity of trauma with depressive disorders. The role of psilocybin continues to expand outside its FDA designation for treatment-resistant depression. Khan et al[34] reported on an open-label study that provided psilocybin-assisted therapy in traumatized AIDS survivors. The authors noted reductions in PTSD symptoms, attachment anxiety, and demoralization. The intent underlying psychedelic use appears to be important. One survey demonstrated that therapeutic intent behind past psychedelic use in patients with history of child maltreatment showed significant reductions in complex trauma symptoms and internalized shame[35]. This further suggests that psilocybin’s benefits extend beyond depression and into trauma-related pathologies. Though outside the scope of the present article, psilocybin is being explored across several domains including substance use disorders, neurodegenerative diseases such as Alzheimer’s disease[36].

Adverse reactions

In the psilocybin and escitalopram study, the escitalopram group had a higher prevalence of anxiety, dry mouth, sexual dysfunction, and reduced emotional responsiveness. Due to these side effects, four patients self-discontinued the medication and one patient modified regimen to half a daily dose. No patients in the psilocybin group requested dose adjustment or discontinuation[7]. In the trial by Davis et al[31], participants reported mild-to-moderate headache, as well as difficult emotions during in-session time only. No serious adverse events were reported or observed[31].

MDMA

In 2021, Mitchell et al[16] reported data from a double-blind, phase 3 clinical trial found that 67% of participants receiving MDMA-assisted psychotherapy ceased to meet PTSD criteria after 2 mo[16]. The MDMA-treated group (n = 46) showed significant decreases on clinician-administered PTSD Scale for DSM-5 (CAPS-V) compared to inactive placebo with therapy (n = 44). Participants attended three experimental sessions, spaced four weeks apart. The first session started with 80 mg MDMA and the option for a supplemental half-dose of 40 mg, 1.5-2.5 h later. For the next two sessions, the initial dose was increased to 120 mg with supplemental half-dose of 60 mg. No tolerability issues led to participants being withheld the supplemental doses.

MDMA-assisted therapy led to increases in posttraumatic growth, encompassing increased positivity towards self-perception, relationships, or philosophy of life[37]. Scores from 60 participants, pooled from three phase 2 clinical studies meeting PTSD criteria, were assigned to treatment with 75-125 mg MDMA (n = 45) or active control (0-40 mg MDMA, n = 15). The MDMA group had significantly improved scores on the posttraumatic growth inventory and greater reduction in PTSD symptom severity at 12 mo, and 67% of participants no longer met criteria for PTSD. MDMA may promote adaptive stress responses in PTSD that lead to the downstream benefits seen in recent research, such as reduced ratings of PTSD symptom severity by clinicians[38,39].

MDMA has also been studied in the setting of couples therapy, in which one partner has a current diagnosis of PTSD. One study included 6 romantic dyads, in which both partners received MDMA followed by couples’ cognitive behavioral therapy. This led to improved happiness and significant reductions across PTSD symptoms, as unanimously rated by patient, partner, and clinician[40]. Further analysis of this study found enduring improvements in post-traumatic growth, social intimacy, and relational support at 6-mo follow up[41].

These benefits extend to common comorbidities seen in PTSD such as depression[42], substance use[43] and sleep disorders[44], with improvements in Pittsburgh sleep quality index scores that remained significant at one-year follow up. MDMA also shows promise in various other psychopathologies including eating disorders[45] and end of life anxiety associated with life-threatening illness[46], in which patients reported increased ability to cope as they faced illness and existential fears, as well as overall improved quality of life. See Table 1 for a summary of relevant articles discussed in the above psilocybin and MDMA sections.

Table 1 Relevant articles to the discussion with the corresponding treatment investigated.
Ref.
Publication year
Treatment
Psychiatric diagnosis
Carhart-Harris et al[7]2021PsilocybinDepression
Murphy et al[30]2021PsilocybinDepression
Davis et al[31]2021PsilocybinDepression
Khan et al[34]2022PsilocybinTrauma-related disorders in AIDS patients
Healy et al[35]2021PsilocybinComplex Trauma
Kozlowska et al[36]2022PsilocybinNeurodegenerative disorders
Mitchell et al[16]2021MDMAPTSD
Gorman et al[37]2020MDMAPTSD
Hoskins et al[38]2021MDMAPTSD
Arluk et al[39]2022MDMAPTSD
Monson et al[40]2020MDMAPTSD
Wagner et al[41]2021MDMAPTSD
Bird et al[42]2021MDMADepression
Nicholas et al[43]2022MDMASubstance use disorders
Ponte et al[44]2021MDMASleep disorders
Brewerton et al[45]2021MDMAEating disorders
Barone et al[46]2022MDMAEnd-of-life anxiety associated with life-threatening illness
Adverse reactions

Mitchell et al[16] concluded that MDMA was safe and well-tolerated and note that the treatment did not induce abuse potential, suicidality, or QT prolongation[16]. Other studies show that risk factors for developing hyperthermia may include adolescent age and increased alcohol consumption[47-49]. Hyperthermia and rhabdomyolysis are likely context-dependent, occurring at lower frequency when used with therapeutic intent rather than recreational use in the setting of other risk factors[47,50]. Likewise, the “come downs” previously associated following MDMA use may be due to research confounds related to environment and drug sourcing, as clinically administered MDMA has noticeably lacked this side effect[51]. Other reports in the literature include hepatotoxicity that improved with vitamin E[52], and a case of spinal cord injury suspected to be due to the serotonin surge induced by MDMA, as the vasoconstrictive properties of serotonin may have induced ischemia[53].

FUTURE DIRECTIONS
Dual therapy

As the evidence for each treatment builds independently, the future may include the integration of both treatments in sequence. In this scenario, MDMA would likely be used first as it has a larger influence on building therapeutic alliance. The augmented therapeutic alliance would allow for a potentially improved psilocybin experience upon switching at next session, aligning with previously discussed research that strong therapeutic alliances are associated with greater mystical experiences and emotional breakthroughs. Treatment with MDMA has been associated with persistent depressive symptoms, and the antidepressant effects of psilocybin may alleviate this following administration[42]. In a model described by Oehen and Gasser[54], MDMA tended to be used in the first phase to build the therapeutic alliance and increase the patient’s openness to change, leading to enhanced resilience and lowered stress levels. Once further changes were noticed, such as improved self-regulation, less negative self-perception, and increased tolerance to trauma exposure, LSD was introduced to assist psychotherapy. The deepening of the therapeutic process led to improvement per clinical judgment, without adverse events.

Standardization

Although variation in research methodology is needed to enrich the literature pool, standardization in several aspects may allow for more informed comparisons between treatment modalities. Symptom rating scales for depression (i.e, QIDS-SR, GRID-HAMD) and PTSD (i.e, CAPS-IV, CAPS-V) varied, with some studies using subjective reports of PTSD diagnostic criteria. Similarly, the therapy modalities that accompanied psychedelic administration were diverse, including uniquely developed forms of therapy as in the accept-connect-embody manualized therapy developed in a psilocybin trial[7,30], supportive therapy[31], and couples’ cognitive behavioral therapy[40]. The future may look more like the manualized MDMA-assisted therapy provided through public benefit corporation Multidisciplinary Association for Psychedelic Studies (MAPS). MAPS also has an ethics code, a vital addition to future protocols as patients are placed in a state of increased suggestibility and affective instability during and after the treatment[55]. This critical period requires a trauma-informed approach to care, with support for transgender and gender diverse patients[56]. Lastly, the method of eliciting adverse drug reactions may benefit from standardization. Studies varied in terms of asking open-ended questions, prompting for specific symptoms, and documenting clinical observations. It is comforting that current studies have not shown any serious adverse events, and future studies may benefit by drawing upon a standardized set of common symptoms.

Wearables

Wearable technology has previously demonstrated efficacy as a bridge between patients and providers in mental health, such as in MDD, dementia, schizophrenia[57-59]. Wearables are likewise being used in non-psychiatric contexts, including medical monitoring in oncology and gastroenterology[60,61]. It follows that wearables may have clinical utility in monitoring patient symptoms during and after PAT. Daily functioning, including wearable-derived sleep and activity data, is incorporated in a protocol examining the effects of microdosed psychedelics and may be a useful metric to track long term changes following psychedelic treatment[62].

CONCLUSION

The literature on PAT using psilocybin or MDMA is strongly supportive across psychiatric conditions, especially trauma and related comorbidities. Both treatments seem to have benefits beyond the FDA breakthrough designations of treatment-resistant depression for psilocybin and PTSD for MDMA. As adjuncts to psychotherapy, psilocybin and MDMA show a pattern of rapid and sustained symptom relief. Future studies may consider the advantages of a standardized approach to symptom rating scales, therapy styles, and assessment of adverse drug reactions. Wearables may also offer additional metrics to examine the long-term trends in activity and sleep. As clinical trials continue to show positive results, providers and patients become closer to seeing the effects translate to the clinic and the community.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Psychiatry

Country/Territory of origin: United States

Peer-review report’s scientific quality classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: He L, China; Pantelis AG, Greece S-Editor: Li L L-Editor: A P-Editor: Chen YX

References
1.  Reiff CM, Richman EE, Nemeroff CB, Carpenter LL, Widge AS, Rodriguez CI, Kalin NH, McDonald WM; the Work Group on Biomarkers and Novel Treatments, a Division of the American Psychiatric Association Council of Research. Psychedelics and Psychedelic-Assisted Psychotherapy. Am J Psychiatry. 2020;177:391-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 238]  [Article Influence: 59.5]  [Reference Citation Analysis (0)]
2.  Garcia-Romeu A, Kersgaard B, Addy PH. Clinical applications of hallucinogens: A review. Exp Clin Psychopharmacol. 2016;24:229-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 82]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
3.  Wasson GR  The Wondrous Mushroom: Mycolatry in Meso- America. New York: McGraw-Hill, 1980.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Carhart-Harris RL, Bolstridge M, Rucker J, Day CM, Erritzoe D, Kaelen M, Bloomfield M, Rickard JA, Forbes B, Feilding A, Taylor D, Pilling S, Curran VH, Nutt DJ. Psilocybin with psychological support for treatment-resistant depression: an open-label feasibility study. Lancet Psychiatry. 2016;3:619-627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 691]  [Cited by in F6Publishing: 759]  [Article Influence: 94.9]  [Reference Citation Analysis (0)]
5.  Carhart-Harris RL, Bolstridge M, Day CMJ, Rucker J, Watts R, Erritzoe DE, Kaelen M, Giribaldi B, Bloomfield M, Pilling S, Rickard JA, Forbes B, Feilding A, Taylor D, Curran HV, Nutt DJ. Psilocybin with psychological support for treatment-resistant depression: six-month follow-up. Psychopharmacology (Berl). 2018;235:399-408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 361]  [Cited by in F6Publishing: 433]  [Article Influence: 72.2]  [Reference Citation Analysis (0)]
6.  Woo BK, Chen W. Substance misuse among older patients in psychiatric emergency service. Gen Hosp Psychiatry. 2010;32:99-101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
7.  Carhart-Harris R, Giribaldi B, Watts R, Baker-Jones M, Murphy-Beiner A, Murphy R, Martell J, Blemings A, Erritzoe D, Nutt DJ. Trial of Psilocybin versus Escitalopram for Depression. N Engl J Med. 2021;384:1402-1411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 362]  [Cited by in F6Publishing: 505]  [Article Influence: 168.3]  [Reference Citation Analysis (0)]
8.  Argento E, Christie D, Mackay L, Callon C, Walsh Z. Psychedelic-Assisted Psychotherapy After COVID-19: The Therapeutic Uses of Psilocybin and MDMA for Pandemic-Related Mental Health Problems. Front Psychiatry. 2021;12:716593.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
9.  Griffiths RR, Richards WA, McCann U, Jesse R. Psilocybin can occasion mystical-type experiences having substantial and sustained personal meaning and spiritual significance. Psychopharmacology (Berl). 2006;187:268-83; discussion 284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 728]  [Cited by in F6Publishing: 668]  [Article Influence: 37.1]  [Reference Citation Analysis (0)]
10.  Hendricks PS. Awe: a putative mechanism underlying the effects of classic psychedelic-assisted psychotherapy. Int Rev Psychiatry. 2018;30:331-342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 74]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
11.  Vaid G, Walker B. Psychedelic Psychotherapy: Building Wholeness Through Connection. Glob Adv Health Med. 2022;11:2164957X221081113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 4]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
12.  Roseman L, Nutt DJ, Carhart-Harris RL. Quality of Acute Psychedelic Experience Predicts Therapeutic Efficacy of Psilocybin for Treatment-Resistant Depression. Front Pharmacol. 2017;8:974.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 332]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
13.  Griffiths RR, Johnson MW, Richards WA, Richards BD, Jesse R, MacLean KA, Barrett FS, Cosimano MP, Klinedinst MA. Psilocybin-occasioned mystical-type experience in combination with meditation and other spiritual practices produces enduring positive changes in psychological functioning and in trait measures of prosocial attitudes and behaviors. J Psychopharmacol. 2018;32:49-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 205]  [Article Influence: 34.2]  [Reference Citation Analysis (0)]
14.  Danforth AL, Struble CM, Yazar-Klosinski B, Grob CS. MDMA-assisted therapy: A new treatment model for social anxiety in autistic adults. Prog Neuropsychopharmacol Biol Psychiatry. 2016;64:237-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 55]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
15.  Holland J, Weil A, Metzner R, Doblin R, Rushkoff D, Shulgin SA, Schachter RZ.   Ecstasy: The Complete Guide: A comprehensive look at the risks and benefits of MDMA. Holland J, Editor. Rochester, Vermont: Park Street Press, 2001.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Mitchell JM, Bogenschutz M, Lilienstein A, Harrison C, Kleiman S, Parker-Guilbert K, Ot'alora G M, Garas W, Paleos C, Gorman I, Nicholas C, Mithoefer M, Carlin S, Poulter B, Mithoefer A, Quevedo S, Wells G, Klaire SS, van der Kolk B, Tzarfaty K, Amiaz R, Worthy R, Shannon S, Woolley JD, Marta C, Gelfand Y, Hapke E, Amar S, Wallach Y, Brown R, Hamilton S, Wang JB, Coker A, Matthews R, de Boer A, Yazar-Klosinski B, Emerson A, Doblin R. MDMA-assisted therapy for severe PTSD: a randomized, double-blind, placebo-controlled phase 3 study. Nat Med. 2021;27:1025-1033.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 337]  [Article Influence: 112.3]  [Reference Citation Analysis (0)]
17.  Bedi G, Phan KL, Angstadt M, de Wit H. Effects of MDMA on sociability and neural response to social threat and social reward. Psychopharmacology (Berl). 2009;207:73-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 126]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
18.  Wardle MC, de Wit H. MDMA alters emotional processing and facilitates positive social interaction. Psychopharmacology (Berl). 2014;231:4219-4229.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 54]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
19.  Feduccia AA, Holland J, Mithoefer MC. Progress and promise for the MDMA drug development program. Psychopharmacology (Berl). 2018;235:561-571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
20.  Liechti ME. Modern Clinical Research on LSD. Neuropsychopharmacology. 2017;42:2114-2127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 113]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
21.  Gravitz L. Hope that psychedelic drugs can erase trauma. Nature. 2022;609:S83-S85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (2)]
22.  Carhart-Harris RL, Erritzoe D, Williams T, Stone JM, Reed LJ, Colasanti A, Tyacke RJ, Leech R, Malizia AL, Murphy K, Hobden P, Evans J, Feilding A, Wise RG, Nutt DJ. Neural correlates of the psychedelic state as determined by fMRI studies with psilocybin. Proc Natl Acad Sci U S A. 2012;109:2138-2143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 517]  [Cited by in F6Publishing: 548]  [Article Influence: 45.7]  [Reference Citation Analysis (0)]
23.  Ly C, Greb AC, Cameron LP, Wong JM, Barragan EV, Wilson PC, Burbach KF, Soltanzadeh Zarandi S, Sood A, Paddy MR, Duim WC, Dennis MY, McAllister AK, Ori-McKenney KM, Gray JA, Olson DE. Psychedelics Promote Structural and Functional Neural Plasticity. Cell Rep. 2018;23:3170-3182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 360]  [Cited by in F6Publishing: 461]  [Article Influence: 76.8]  [Reference Citation Analysis (0)]
24.  Catlow BJ, Song S, Paredes DA, Kirstein CL, Sanchez-Ramos J. Effects of psilocybin on hippocampal neurogenesis and extinction of trace fear conditioning. Exp Brain Res. 2013;228:481-491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 147]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
25.  Arnsten AF. Stress signalling pathways that impair prefrontal cortex structure and function. Nat Rev Neurosci. 2009;10:410-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1674]  [Cited by in F6Publishing: 1704]  [Article Influence: 113.6]  [Reference Citation Analysis (0)]
26.  Andero R, Ressler KJ. Fear extinction and BDNF: translating animal models of PTSD to the clinic. Genes Brain Behav. 2012;11:503-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 184]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
27.  Guthrie RM, Bryant RA. Extinction learning before trauma and subsequent posttraumatic stress. Psychosom Med. 2006;68:307-311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 140]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
28.  Raut SB, Marathe PA, van Eijk L, Eri R, Ravindran M, Benedek DM, Ursano RJ, Canales JJ, Johnson LR. Diverse therapeutic developments for post-traumatic stress disorder (PTSD) indicate common mechanisms of memory modulation. Pharmacol Ther. 2022;239:108195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
29.  Kéri S. Trauma and Remembering: From Neuronal Circuits to Molecules. Life (Basel). 2022;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
30.  Murphy R, Kettner H, Zeifman R, Giribaldi B, Kartner L, Martell J, Read T, Murphy-Beiner A, Baker-Jones M, Nutt D, Erritzoe D, Watts R, Carhart-Harris R. Therapeutic Alliance and Rapport Modulate Responses to Psilocybin Assisted Therapy for Depression. Front Pharmacol. 2021;12:788155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 47]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
31.  Davis AK, Barrett FS, May DG, Cosimano MP, Sepeda ND, Johnson MW, Finan PH, Griffiths RR. Effects of Psilocybin-Assisted Therapy on Major Depressive Disorder: A Randomized Clinical Trial. JAMA Psychiatry. 2021;78:481-489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 337]  [Cited by in F6Publishing: 496]  [Article Influence: 165.3]  [Reference Citation Analysis (0)]
32.  Rubin A, Yu M. Within-group effect size benchmarks for cognitive–behavioral therapy in the treatment of adult depression. Soc Work Res. 2017;41:135-144.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Fournier JC, DeRubeis RJ, Hollon SD, Dimidjian S, Amsterdam JD, Shelton RC, Fawcett J. Antidepressant drug effects and depression severity: a patient-level meta-analysis. JAMA. 2010;303:47-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1318]  [Cited by in F6Publishing: 1198]  [Article Influence: 85.6]  [Reference Citation Analysis (0)]
34.  Khan AJ, Bradley E, O'Donovan A, Woolley J. Psilocybin for Trauma-Related Disorders. Curr Top Behav Neurosci. 2022;56:319-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
35.  Healy CJ, Lee KA, D'Andrea W. Using Psychedelics With Therapeutic Intent Is Associated With Lower Shame and Complex Trauma Symptoms in Adults With Histories of Child Maltreatment. Chronic Stress (Thousand Oaks). 2021;5:24705470211029881.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
36.  Kozlowska U, Nichols C, Wiatr K, Figiel M. From psychiatry to neurology: Psychedelics as prospective therapeutics for neurodegenerative disorders. J Neurochem. 2022;162:89-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
37.  Gorman I, Belser AB, Jerome L, Hennigan C, Shechet B, Hamilton S, Yazar-Klosinski B, Emerson A, Feduccia AA. Posttraumatic Growth After MDMA-Assisted Psychotherapy for Posttraumatic Stress Disorder. J Trauma Stress. 2020;33:161-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
38.  Hoskins MD, Sinnerton R, Nakamura A, Underwood JFG, Slater A, Lewis C, Roberts NP, Bisson JI, Lee M, Clarke L. Pharmacological-assisted Psychotherapy for Post-Traumatic Stress Disorder: a systematic review and meta-analysis. Eur J Psychotraumatol. 2021;12:1853379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
39.  Arluk S, Matar MA, Carmi L, Arbel O, Zohar J, Todder D, Cohen H. MDMA treatment paired with a trauma-cue promotes adaptive stress responses in a translational model of PTSD in rats. Transl Psychiatry. 2022;12:181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
40.  Monson CM, Wagner AC, Mithoefer AT, Liebman RE, Feduccia AA, Jerome L, Yazar-Klosinski B, Emerson A, Doblin R, Mithoefer MC. MDMA-facilitated cognitive-behavioural conjoint therapy for posttraumatic stress disorder: an uncontrolled trial. Eur J Psychotraumatol. 2020;11:1840123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
41.  Wagner AC, Liebman RE, Mithoefer AT, Mithoefer MC, Monson CM. Relational and Growth Outcomes Following Couples Therapy With MDMA for PTSD. Front Psychiatry. 2021;12:702838.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
42.  Bird CIV, Modlin NL, Rucker JJH. Psilocybin and MDMA for the treatment of trauma-related psychopathology. Int Rev Psychiatry. 2021;33:229-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 21]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
43.  Nicholas CR, Wang JB, Coker A, Mitchell JM, Klaire SS, Yazar-Klosinski B, Emerson A, Brown RT, Doblin R. The effects of MDMA-assisted therapy on alcohol and substance use in a phase 3 trial for treatment of severe PTSD. Drug Alcohol Depend. 2022;233:109356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 15]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
44.  Ponte L, Jerome L, Hamilton S, Mithoefer MC, Yazar-Klosinski BB, Vermetten E, Feduccia AA. Sleep Quality Improvements After MDMA-Assisted Psychotherapy for the Treatment of Posttraumatic Stress Disorder. J Trauma Stress. 2021;34:851-863.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
45.  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. Med Hypotheses. 2021;146:110367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
46.  Barone W, Mitsunaga-Whitten M, Blaustein LO, Perl P, Swank M, Swift TC. Facing death, returning to life: A qualitative analysis of MDMA-assisted therapy for anxiety associated with life-threatening illness. Front Psychiatry. 2022;13:944849.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 4]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
47.  Reddi S, Friedman MS. Recreational drug toxicity with severe hyperthermia: Rapid onsite treatment and clinical course. Am J Emerg Med. 2022;62:144.e5-144.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
48.  Chitre NM, Bagwell MS, Murnane KS. The acute toxic and neurotoxic effects of 3,4-methylenedioxymethamphetamine are more pronounced in adolescent than adult mice. Behav Brain Res. 2020;380:112413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
49.  van Amsterdam J, Brunt TM, Pierce M, van den Brink W. Hard Boiled: Alcohol Use as a Risk Factor for MDMA-Induced Hyperthermia: a Systematic Review. Neurotox Res. 2021;39:2120-2133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
50.  Richards JR, Wang CG, Fontenette RW, Stuart RP, McMahon KF, Turnipseed SD. Rhabdomyolysis, Methamphetamine, Amphetamine and MDMA Use: Associated Factors and Risks. J Dual Diagn. 2020;16:429-437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
51.  Sessa B, Aday JS, O'Brien S, Curran HV, Measham F, Higbed L, Nutt DJ. Debunking the myth of 'Blue Mondays': No evidence of affect drop after taking clinical MDMA. J Psychopharmacol. 2022;36:360-367.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
52.  Javanmard MZ, Meghrazi K, Ghafori SS, Karimipour M. The ameliorating effects of Vitamin E on hepatotoxicity of ecstasy. J Res Med Sci. 2020;25:91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
53.  Kastaneer ME, Maijers MC, Martina JD. Spinal cord injury after the use of ecstasy in a young male. Spinal Cord Ser Cases. 2020;6:29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
54.  Oehen P, Gasser P. Using a MDMA- and LSD-Group Therapy Model in Clinical Practice in Switzerland and Highlighting the Treatment of Trauma-Related Disorders. Front Psychiatry. 2022;13:863552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
55.  Anderson BT, Danforth AL, Grob CS. Psychedelic medicine: safety and ethical concerns. Lancet Psychiatry. 2020;7:829-830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 37]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
56.  Stauffer CS, Brown MR, Adams D, Cassity M, Sevelius J. MDMA-assisted psychotherapy; Inclusion of transgender and gender diverse people in the frontiers of PTSD treatment trials. Front Psychiatry. 2022;13:932605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Reference Citation Analysis (0)]
57.  Woo BKP, Chung JOP, Shu S, Zhu P. Wearable technology for symptom measurement in major depressive disorder. J Affect Disord. 2022;317:5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
58.  Shu S, Woo BK. Use of technology and social media in dementia care: Current and future directions. World J Psychiatry. 2021;11:109-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 27]  [Cited by in F6Publishing: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (1)]
59.  Fonseka LN, Woo BKP. Wearables in Schizophrenia: Update on Current and Future Clinical Applications. JMIR Mhealth Uhealth. 2022;10:e35600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
60.  Fonseka LN, Woo BK. Consumer Wearables and the Integration of New Objective Measures in Oncology: Patient and Provider Perspectives. JMIR Mhealth Uhealth. 2021;9:e28664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
61.  Chong KP, Woo BK. Emerging wearable technology applications in gastroenterology: A review of the literature. World J Gastroenterol. 2021;27:1149-1160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
62.  Murphy RJ, Sumner RL, Evans W, Menkes D, Lambrecht I, Ponton R, Sundram F, Hoeh N, Ram S, Reynolds L, Muthukumaraswamy S. MDLSD: study protocol for a randomised, double-masked, placebo-controlled trial of repeated microdoses of LSD in healthy volunteers. Trials. 2021;22:302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]