MDMA-assisted therapy for severe PTSD: a randomized, double-blind, placebo-controlled phase 3 study

Jennifer M Mitchell, Michael Bogenschutz, Alia Lilienstein, Charlotte Harrison, Sarah Kleiman, Kelly Parker-Guilbert, Marcela Ot'alora G, Wael Garas, Casey Paleos, Ingmar Gorman, Christopher Nicholas, Michael Mithoefer, Shannon Carlin, Bruce Poulter, Ann Mithoefer, Sylvestre Quevedo, Gregory Wells, Sukhpreet S Klaire, Bessel van der Kolk, Keren Tzarfaty, Revital Amiaz, Ray Worthy, Scott Shannon, Joshua D Woolley, Cole Marta, Yevgeniy Gelfand, Emma Hapke, Simon Amar, Yair Wallach, Randall Brown, Scott Hamilton, Julie B Wang, Allison Coker, Rebecca Matthews, Alberdina de Boer, Berra Yazar-Klosinski, Amy Emerson, Rick Doblin, Jennifer M Mitchell, Michael Bogenschutz, Alia Lilienstein, Charlotte Harrison, Sarah Kleiman, Kelly Parker-Guilbert, Marcela Ot'alora G, Wael Garas, Casey Paleos, Ingmar Gorman, Christopher Nicholas, Michael Mithoefer, Shannon Carlin, Bruce Poulter, Ann Mithoefer, Sylvestre Quevedo, Gregory Wells, Sukhpreet S Klaire, Bessel van der Kolk, Keren Tzarfaty, Revital Amiaz, Ray Worthy, Scott Shannon, Joshua D Woolley, Cole Marta, Yevgeniy Gelfand, Emma Hapke, Simon Amar, Yair Wallach, Randall Brown, Scott Hamilton, Julie B Wang, Allison Coker, Rebecca Matthews, Alberdina de Boer, Berra Yazar-Klosinski, Amy Emerson, Rick Doblin

Abstract

Post-traumatic stress disorder (PTSD) presents a major public health problem for which currently available treatments are modestly effective. We report the findings of a randomized, double-blind, placebo-controlled, multi-site phase 3 clinical trial (NCT03537014) to test the efficacy and safety of 3,4-methylenedioxymethamphetamine (MDMA)-assisted therapy for the treatment of patients with severe PTSD, including those with common comorbidities such as dissociation, depression, a history of alcohol and substance use disorders, and childhood trauma. After psychiatric medication washout, participants (n = 90) were randomized 1:1 to receive manualized therapy with MDMA or with placebo, combined with three preparatory and nine integrative therapy sessions. PTSD symptoms, measured with the Clinician-Administered PTSD Scale for DSM-5 (CAPS-5, the primary endpoint), and functional impairment, measured with the Sheehan Disability Scale (SDS, the secondary endpoint) were assessed at baseline and at 2 months after the last experimental session. Adverse events and suicidality were tracked throughout the study. MDMA was found to induce significant and robust attenuation in CAPS-5 score compared with placebo (P < 0.0001, d = 0.91) and to significantly decrease the SDS total score (P = 0.0116, d = 0.43). The mean change in CAPS-5 scores in participants completing treatment was -24.4 (s.d. 11.6) in the MDMA group and -13.9 (s.d. 11.5) in the placebo group. MDMA did not induce adverse events of abuse potential, suicidality or QT prolongation. These data indicate that, compared with manualized therapy with inactive placebo, MDMA-assisted therapy is highly efficacious in individuals with severe PTSD, and treatment is safe and well-tolerated, even in those with comorbidities. We conclude that MDMA-assisted therapy represents a potential breakthrough treatment that merits expedited clinical evaluation.

Conflict of interest statement

The authors declare the following financial competing interests: A.E., R.M., C.H., A.D.B., S.C., A.C. and J.B.W. received salary support for full-time employment with MAPS PBC for this study and other work; A.L., B.Y.-K. and R.D. received salary support for full-time employment with MAPS for this study and other work; M.M., A.M., M.O.G., B.P. and K.T. received support as contractors from MAPS PBC for training and supervision of research psychotherapists for this study and other work; S.K., K.P.-G. and S.H. received support as contractors of MAPS PBC for their contributions to this study and other work; and study investigators and researchers, J.M.M., M.B., M.O.G., W.G., C.P., I.G., C.N., B.P., S.Q., G.W., S.S.K., B.V.D.K., K.T., R.A., R.W., S.S., J.D.W., C.M., Y.G., E.H., S.A., Y.W. and R.B., received funding from MAPS PBC during the conduct of the study for this study as well as other studies. The following authors disclose receipt of personal fees or grants from companies in the field, but unrelated to the present work: M.B. (Heffter Research Institute, Turnbull Family Foundation, B. More, Mind Medicine, Fournier Family Foundation, Bill Linton, George Sarlo Foundation, RiverStyx Foundation, Dr. Bronners Family Foundation, and National Institutes of Health), C.P. (Fluence and Mindbloom), J.D.W. (Filament Ventures and Silo Pharmaceuticals), and J.M.M. and A.C. (Usona Institute). The following authors disclose non-financial relationships with organizations in the field: M.M. and A.M. serve on the Scientific Advisory Board for Awaken Life Sciences, C.P. co-founded Nautilus Sanctuary and Nautilus Psychiatric Services, S.S. serves on the advisory board for Maya Health, and I.G. serves on the Scientific Advisory Board of Journey Clinical and co-founded Fluence.

Figures

Fig. 1. Procedure timeline and study flow…
Fig. 1. Procedure timeline and study flow diagram.
a, Procedure timeline. Following the screening procedures and medication taper, participants attended a total of three preparatory sessions, three experimental sessions, nine integration sessions and four endpoint assessments (T1–4) over 18 weeks, concluding with a final study-termination visit. IR, independent rater; T, timepoint of endpoint assessment; T1, baseline; T2, after the first experimental session; T3, after the second experimental session; T4, 18 weeks after baseline. b, CONSORT diagram indicating participant numbers and disposition through the course of the trial.
Fig. 2. Measures of MDMA efficacy in…
Fig. 2. Measures of MDMA efficacy in the MDMA-assisted therapy group and the placebo group.
a, Change in CAPS-5 total severity score from T1 to T4 (P < 0.0001, d = 0.91, n = 89 (MDMA n = 46)), as a measure of the primary outcome. Primary analysis was completed using least square means from an MMRM model. b, Change in SDS total score from T1 to T4 (P = 0.0116, d = 0.43, n = 89 (MDMA n = 46)), as a measure of the secondary outcome. Primary analysis was completed using least square means from an MMRM model. c, Change in BDI-II score from T1 to study termination (t = −3.11, P = 0.0026, n = 81 (MDMA n = 42)), as a measure of the exploratory outcome. Data are presented as mean and s.e.m.
Fig. 3. Treatment response and remission for…
Fig. 3. Treatment response and remission for MDMA and placebo groups as a percentage of total participants randomized to each arm (MDMA, n = 46; placebo, n = 44).
Responders (clinically significant improvement, defined as a ≥10-point decrease on CAPS-5), loss of diagnosis (specific diagnostic measure on CAPS-5), and remission (loss of diagnosis and a total CAPS-5 score of ≤11) were tracked in both groups. Non-response is defined as a

Fig. 4. Number of participants reporting the…

Fig. 4. Number of participants reporting the presence of suicidal ideation as measured with the…

Fig. 4. Number of participants reporting the presence of suicidal ideation as measured with the C-SSRS at each visit and separated by treatment group.
C-SSRS ideation scores range from 0 (no ideation) to 5. A C-SSRS ideation score of 4 or 5 is termed ‘serious ideation’. The number of participants endorsing any positive ideation (>0) is shown by the colored bars and noted in the table below the graph. The number of participants endorsing serious ideation is given in parentheses in the table.
Fig. 4. Number of participants reporting the…
Fig. 4. Number of participants reporting the presence of suicidal ideation as measured with the C-SSRS at each visit and separated by treatment group.
C-SSRS ideation scores range from 0 (no ideation) to 5. A C-SSRS ideation score of 4 or 5 is termed ‘serious ideation’. The number of participants endorsing any positive ideation (>0) is shown by the colored bars and noted in the table below the graph. The number of participants endorsing serious ideation is given in parentheses in the table.

References

    1. Yehuda R, et al. Putative biological mechanisms for the association between early life adversity and the subsequent development of PTSD. Psychopharmacology. 2010;212:405–417. doi: 10.1007/s00213-010-1969-6.
    1. Widom CS. Posttraumatic stress disorder in abused and neglected children grown up. Am. J. Psychiatry. 1999;156:1223–1229.
    1. Kessler RC, Sonnega A, Bromet E, Hughes M, Nelson CB. Posttraumatic stress disorder in the National Comorbidity Survey. Arch. Gen. Psychiatry. 1995;52:1048–1060. doi: 10.1001/archpsyc.1995.03950240066012.
    1. Hegel MT, et al. Impact of comorbid panic and posttraumatic stress disorder on outcomes of collaborative care for late-life depression in primary care. Am. J. Geriatr. Psychiatry. 2005;13:48–58. doi: 10.1097/00019442-200501000-00008.
    1. Tarrier N, Taylor K, Gooding P. Cognitive-behavioral interventions to reduce suicide behavior: a systematic review and meta-analysis. Behav. Modif. 2008;32:77–108. doi: 10.1177/0145445507304728.
    1. Wolf EJ, Lunney CA, Schnurr PP. The influence of the dissociative subtype of posttraumatic stress disorder on treatment efficacy in female veterans and active duty service members. J. Consult. Clin. Psychol. 2016;84:95–100. doi: 10.1037/ccp0000036.
    1. Steenkamp MM, Litz BT, Hoge CW, Marmar CR. Psychotherapy for military-related PTSD: a review of randomized clinical trials. JAMA. 2015;314:489–500. doi: 10.1001/jama.2015.8370.
    1. Watkins LE, Sprang KR, Rothbaum BO. Treating PTSD: a review of evidence-based psychotherapy interventions. Front Behav. Neurosci. 2018;12:258. doi: 10.3389/fnbeh.2018.00258.
    1. Bisson JI, Roberts NP, Andrew M, Cooper R, Lewis C. Psychological therapies for chronic post-traumatic stress disorder (PTSD) in adults. Cochrane Database Syst. Rev. 2013;2013(12):CD003388.
    1. Gutner CA, Gallagher MW, Baker AS, Sloan DM, Resick PA. Time course of treatment dropout in cognitive-behavioral therapies for posttraumatic stress disorder. Psychol. Trauma. 2016;8:115–121. doi: 10.1037/tra0000062.
    1. 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. 2020;15:e0239997. doi: 10.1371/journal.pone.0239997.
    1. Rudnick G, Wall SC. The molecular mechanism of “ecstasy” [3,4-methylenedioxy-methamphetamine(MDMA)]: serotonin transporters are targets for MDMA-induced serotonin release. Proc. Natl Acad. Sci. USA. 1992;89:1817–1821. doi: 10.1073/pnas.89.5.1817.
    1. Hake HS, et al. 3,4-Methylenedioxymethamphetamine (MDMA) impairs the extinction and reconsolidation of fear memory in rats. Physiol. Behav. 2019;199:343–350. doi: 10.1016/j.physbeh.2018.12.007.
    1. Nardou R, et al. Oxytocin-dependent reopening of a social reward learning critical period with MDMA. Nature. 2019;569:116–120. doi: 10.1038/s41586-019-1075-9.
    1. Mithoefer MC, 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;236:2735–2745. doi: 10.1007/s00213-019-05249-5.
    1. Feduccia AA, et al. Breakthrough for trauma treatment: safety and efficacy of MDMA-assisted psychotherapy compared to paroxetine and sertraline. Front. Psychiatry. 2019;10:650. doi: 10.3389/fpsyt.2019.00650.
    1. Lee DJ, et al. Psychotherapy versus pharmacotherapy for posttraumatic stress disorder: systematic review and meta-analyses to determine first-line treatments. Depress. Anxiety. 2016;33:792–806. doi: 10.1002/da.22511.
    1. Feduccia, A. A., Jerome, L., Mithoefer, M. C. & Holland, J. Discontinuation of medications classified as reuptake inhibitors affects treatment response of MDMA-assisted psychotherapy. Psychopharmacology (Berl.) 238, 581–588 (2021).
    1. Bauer EP. Serotonin in fear conditioning processes. Behav. Brain Res. 2015;277:68–77. doi: 10.1016/j.bbr.2014.07.028.
    1. Johnson PL, et al. Assessment of fear and anxiety associated behaviors, physiology and neural circuits in rats with reduced serotonin transporter (SERT) levels. Transl. Psychiatry. 2019;9:33. doi: 10.1038/s41398-019-0368-y.
    1. Lee HJ, et al. Influence of the serotonin transporter promoter gene polymorphism on susceptibility to posttraumatic stress disorder. Depress. Anxiety. 2005;21:135–139. doi: 10.1002/da.20064.
    1. Lesch KP, et al. Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science. 1996;274:1527–1531. doi: 10.1126/science.274.5292.1527.
    1. Hariri AR, et al. Serotonin transporter genetic variation and the response of the human amygdala. Science. 2002;297:400–403. doi: 10.1126/science.1071829.
    1. Kawahara H, Yoshida M, Yokoo H, Nishi M, Tanaka M. Psychological stress increases serotonin release in the rat amygdala and prefrontal cortex assessed by in vivo microdialysis. Neurosci. Lett. 1993;162:81–84. doi: 10.1016/0304-3940(93)90565-3.
    1. Bedi G, Phan KL, Angstadt M, de Wit H. Effects of MDMA on sociability and neural response to social threat and social reward. Psychopharmacology. 2009;207:73–83. doi: 10.1007/s00213-009-1635-z.
    1. Frewen PA, Lanius RA. Toward a psychobiology of posttraumatic self-dysregulation: reexperiencing, hyperarousal, dissociation, and emotional numbing. Ann. NY Acad. Sci. 2006;1071:110–124. doi: 10.1196/annals.1364.010.
    1. Kamboj SK, et al. Recreational 3,4-methylenedioxy-N-methylamphetamine (MDMA) or ‘ecstasy’ and self-focused compassion: preliminary steps in the development of a therapeutic psychopharmacology of contemplative practices. J. Psychopharmacol. 2015;29:961–970. doi: 10.1177/0269881115587143.
    1. Dewey D, Schuldberg D, Madathil R. Do peritraumatic emotions differentially predict PTSD symptom clusters? Initial evidence for emotion specificity. Psychol. Rep. 2014;115:1–12. doi: 10.2466/16.02.PR0.115c11z7.
    1. Hysek CM, et al. MDMA enhances emotional empathy and prosocial behavior. Soc. Cogn. Affect. Neurosci. 2014;9:1645–1652. doi: 10.1093/scan/nst161.
    1. Keller SM, Zoellner LA, Feeny NC. Understanding factors associated with early therapeutic alliance in PTSD treatment: adherence, childhood sexual abuse history, and social support. J. Consult. Clin. Psychol. 2010;78:974–979. doi: 10.1037/a0020758.
    1. Imel ZE, Laska K, Jakupcak M, Simpson TL. Meta-analysis of dropout in treatments for posttraumatic stress disorder. J. Consult. Clin. Psychol. 2013;81:394–404. doi: 10.1037/a0031474.
    1. Mithoefer M. MDMA-assisted psychotherapy; promising treatment for PTSD. American Psychiatric Association Annual Meeting, Toronto, Ontario, Canada (2015).
    1. Fortenbaugh FC, et al. The impact of common psychiatric and behavioral comorbidities on functional disability across time and individuals in post-9/11 veterans. J. Trauma. Stress. 2020;33:750–761. doi: 10.1002/jts.22501.
    1. Bureau of Labor and Statistics. Employment Situation of Veterans—2020. News release, 18 March 2021;
    1. Congressional Budget Office. Possible Higher Spending Paths for Veterans’ Benefits (2018);
    1. Cloitre M, Petkova E, Wang J, Lu Lassell F. An examination of the influence of a sequential treatment on the course and impact of dissociation among women with PTSD related to childhood abuse. Depress. Anxiety. 2012;29:709–717. doi: 10.1002/da.21920.
    1. Vizeli P, Liechti ME. Safety pharmacology of acute MDMA administration in healthy subjects. J. Psychopharmacol. 2017;31:576–588. doi: 10.1177/0269881117691569.
    1. Funk KA, Bostwick JR. A comparison of the risk of QT prolongation among SSRIs. Ann. Pharmacother. 2013;47:1330–1341. doi: 10.1177/1060028013501994.
    1. Bershad AK, Miller MA, Baggott MJ, de Wit H. The effects of MDMA on socio-emotional processing: does MDMA differ from other stimulants? J. Psychopharmacol. 2016;30:1248–1258. doi: 10.1177/0269881116663120.
    1. Weathers FW, et al. The Clinician-Administered PTSD Scale for DSM-5 (CAPS-5): development and initial psychometric evaluation in military veterans. Psychol. Assess. 2018;30:383–395. doi: 10.1037/pas0000486.

Source: PubMed

3
Subscribe