Effects of 3,4-Methylenedioxymethamphetamine on Conditioned Fear Extinction and Retention in a Crossover Study in Healthy Subjects

Patrick Vizeli, Isabelle Straumann, Urs Duthaler, Nimmy Varghese, Anne Eckert, Martin P Paulus, Victoria Risbrough, Matthias E Liechti, Patrick Vizeli, Isabelle Straumann, Urs Duthaler, Nimmy Varghese, Anne Eckert, Martin P Paulus, Victoria Risbrough, Matthias E Liechti

Abstract

Background: 3,4-Methylenedioxymethamphetamine (MDMA) has shown initial promise as an adjunct in psychotherapy to treat posttraumatic stress disorder (PTSD). Its efficacy and safety have been demonstrated across phase I-III studies. However, the mechanism underlying the potential utility of MDMA to treat PTSD in humans has not yet been thoroughly investigated. Preliminary evidence suggests that MDMA may facilitate fear extinction recall, which may be through the release of oxytocin. To test this hypothesis, we examined the efficacy of acute MDMA treatment to enhance fear extinction learning and recall. Methods: We used a two-period, double-blind, randomized, placebo-controlled crossover design in 30 healthy male subjects who received a placebo and a single dose of MDMA (125 mg). Fear extinction was tested using two separate Pavlovian fear conditioning paradigms, one using skin conductance response (SCR), and the other fear-potentiated startle (FPS) to conditioned cues. MDMA treatment occurred after fear conditioning and 2 h before extinction learning. Extinction recall was tested 23 h after MDMA intake. Additional outcome measures included subjective effects, emotion recognition tasks, plasma levels of oxytocin, and pharmacokinetics. Results: Fear conditioning and extinction learning were successful in both fear extinction paradigms (generalized eta-squared [ges] for SCR: 0.08; FPS: 0.07). Compared to placebo treatment, MDMA treatment significantly reduced SCRs to the reinforced conditioned stimulus (CS+) during extinction learning (ges = 0.03) and recall (ges = 0.06). Intensity of the subjective effects of MDMA (good effect, trust, and openness) during extinction learning negatively correlated with the discrimination between CS+ and the safety stimulus (CS-) during recall. MDMA did not influence FPS to conditioned cues. Oxytocin concentration was increased fourfold on average by MDMA during acute effects but was not associated with fear extinction outcomes. Conclusions: MDMA treatment facilitated rapid fear extinction and retention of extinction as measured by SCR to fear cues, in line with animal studies of MDMA facilitation of extinction. However, this effect may be limited to certain forms of learned fear responses, as it was not observed in the extinction model using startle reactivity as the outcome. This study provides further evidence for the facilitation of extinction with MDMA treatment and suggests this may be a component of its efficacy when paired with psychotherapy. Clinical Trial registration: clinicaltrials.gov identifier: NCT03527316.

Keywords: MDMA and fear extinction paradigms; fear extinction; fear-potentiated startle; healthy subjects; oxytocin; skin conductance response.

Conflict of interest statement

ML is a consultant for Mind Medicine, Inc. Knowhow and data associated with this work and owned by the University Hospital Basel were licensed by Mind Medicine, Inc. MP is supported by the William K Warren Foundation and receives grant support from the NationalInstitute on Drug Abuse (U01 DA041089), and the National Institute of General Medical Sciences Center Grant Award Number (1P20GM121312). MP is an advisor to Spring Care, Inc., a behavioral health startup; he has received royalties for an article about methamphetamine in UpToDate. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Vizeli, Straumann, Duthaler, Varghese, Eckert, Paulus, Risbrough and Liechti.

Figures

FIGURE 1
FIGURE 1
Schematic study day. Participants underwent every two sessions with either 125-mg 3,4-methylenedioxymethamphetamine (MDMA) or placebo. The order was randomized but balanced. The washout period between the study days was at least 30 days. Autonomic and subjective effects were assessed throughout the study day. Blood samples for MDMA and oxytocin blood plasma concentrations were collected before and periodically up to 24 h after drug ingestion.
FIGURE 2
FIGURE 2
MDMA treatment reduces CS+/CS− discrimination during fear extinction learning and enhances extinction recall. Standardized values (z-scores) of both trial types (conditioned fear stimulus [CS+] and conditioned safety stimulus [CS−]) of the skin conductance response task. (A) Subjects showed differentiation between CS+ and CS− in acquisition. (B) In the extinction learning phase, subjects after a placebo displayed discrimination of the conditioned stimuli in the early phase and extinction over time, but already early nondiscrimination of the conditioned stimuli after MDMA. (C) In the recall phase, subjects after the placebo displayed an early return of fear memory by discrimination of the conditioned stimuli. There was no discrimination of the conditioned stimuli after MDMA, indicating extinction retention. The acquisition was before drug intake. MDMA, 3,4-methylenedioxymethamphetamine. Values are mean ± standard error of the mean (SEM). Stats in (A) are main effects, *p < 0.05, ***p < 0.001. Stats in (B) and (C) are from Tukey posthoc tests, #p < 0.05, ##p < 0.01, ###p < 0.001.
FIGURE 3
FIGURE 3
MDMA promotes extinction recall. The difference in standardized values (Δz-scores) between the trial types (conditioned fear stimulus [CS+] minus conditioned safety stimulus [CS−]) of the skin conductance response task in the recall. Subjects after MDMA showed significantly less discrimination between CS+ and CS− compared to the placebo session. Stats are from Tukey posthoc tests following an interaction of drug × time in delta values [(F1,22) = 6.74, p = 0.017]. *p < 0.05. MDMA, 3,4-methylenedioxymethamphetamine.
FIGURE 4
FIGURE 4
No changes induced by MDMA in the fear-potentiated startle task. Difference between the standardized values of both conditioned trial types (conditioned fear stimulus [CS+] and conditioned safety stimulus [CS−]) and the pulse alone (NA) during the fear-potentiated startle task. (A) Subjects showed a clear differentiation between CS+ and CS− in acquisition. (B) In the extinction learning phase, subjects in both sessions displayed discrimination of the conditioned stimuli in the early trial blocks (#CS+ vs. CS− of block 1 and 2, Tukey posthoc: p < 0.001 and p < 0.01, respectively) and extinction over time (trial blocks). (C) In the recall extinction phase, subjects in both sessions showed a return of fear of the conditioned stimulus in the early trial blocks and renewed extinction over time. The acquisition was before drug intake. MDMA, 3,4-methylenedioxymethamphetamine. Values are mean ± SEM. Stats in (A) are main effects. *p < 0.05, **p < 0.01, ***p < 0.001.
FIGURE 5
FIGURE 5
MDMA characteristic effects, such as trust and openness, correlate with extinction recall. Correlation matrix between MDMA and oxytocin concentrations and subjective effects of MDMA with extinction recall parameters. Characteristic MDMA effects during extinction learning correlated with the CS+ and subsequently ΔCS in the early phase of extinction recall. Pearson correlation coefficient was used. p-values in white numbers. *p < 0.05; **p < 0.01. Not corrected for multiple testing. CS+, conditioned fear stimulus; CS−, conditioned safety stimulus; ΔCS, CS+–CS−; AMRS, Adjective Mood Rating Scale; VAS, visual analog scale; FPS, fear-potentiated startle; SCR, skin conductance response; MDMA, 3,4-methylenedioxymethamphetamine. Concentrations at 2.5 h were generated as the mean of the respective 2 and 3 h time points. Data is from MDMA sessions only.
FIGURE 6
FIGURE 6
MDMA inhibits the recognition of negative emotion and promotes misclassification as happy. Results of the facial emotion recognition task. (A) Correctly identified emotions. (B) Emotion misclassification if not correctly recognized. Stats are from paired t-tests. *p < 0.05; **p < 0.01. Bonferroni corrected for multiple testing. MDMA, 3,4-methylenedioxymethamphetamine. Values are mean ± standard error of the mean. The task was performed 3 h after drug intake.

References

    1. Acheson D., Feifel D., de Wilde S., McKinney R., Lohr J., Risbrough V. (2013). The Effect of Intranasal Oxytocin Treatment on Conditioned Fear Extinction and Recall in a Healthy Human Sample. Psychopharmacol. Berl. 229 (1), 199–208. 10.1007/s00213-013-3099-4
    1. Acheson D. T., Geyer M. A., Baker D. G., Nievergelt C. M., Yurgil K., Risbrough V. B., et al. (2015). Conditioned Fear and Extinction Learning Performance and its Association with Psychiatric Symptoms in Active Duty Marines. Psychoneuroendocrinology 51, 495–505. 10.1016/j.psyneuen.2014.09.030
    1. Ball T. M., Knapp S. E., Paulus M. P., Stein M. B. (2017). Brain Activation during Fear Extinction Predicts Exposure Success. Depress Anxiety 34 (3), 257–266. 10.1002/da.22583
    1. Bedi G., Phan K. L., Angstadt M., de Wit H. (2009). Effects of MDMA on Sociability and Neural Response to Social Threat and Social Reward. Psychopharmacol. Berl. 207 (1), 73–83. 10.1007/s00213-009-1635-z
    1. Bershad A. K., Mayo L. M., Van Hedger K., McGlone F., Walker S. C., de Wit H. (2019). Effects of MDMA on Attention to Positive Social Cues and Pleasantness of Affective Touch. Neuropsychopharmacology 44, 1698–1705. 10.1038/s41386-019-0402-z
    1. Breslau N., Davis G. C., Andreski P., Peterson E. L., Schultz L. R. (1997). Sex Differences in Posttraumatic Stress Disorder. Arch. Gen. Psychiatry 54 (11), 1044–1048. 10.1001/archpsyc.1997.01830230082012
    1. Brignell C. M., Curran H. V. (2006). Drugs, Sweat, and Fears: a Comparison of the Effects of Diazepam and Methylphenidate on Fear Conditioning. Psychopharmacol. Berl. 186 (4), 504–516. 10.1007/s00213-006-0363-x
    1. Brown H., Pollard K. A. (2021). Drugs of Abuse: Sympathomimetics. Crit. Care Clin. 37 (3), 487–499. 10.1016/j.ccc.2021.03.002
    1. Carhart-Harris R. L., Murphy K., Leech R., Erritzoe D., Wall M. B., Ferguson B., et al. (2015). The Effects of Acutely Administered 3,4-Methylenedioxymethamphetamine on Spontaneous Brain Function in Healthy Volunteers Measured with Arterial Spin Labeling and Blood Oxygen Level-Dependent Resting State Functional Connectivity. Biol. Psychiatry 78 (8), 554–562. 10.1016/j.biopsych.2013.12.015
    1. Connolly H. L., Lefevre C. E., Young A. W., Lewis G. J. (2019). Sex Differences in Emotion Recognition: Evidence for a Small Overall Female Superiority on Facial Disgust. Emotion 19 (3), 455–464. 10.1037/emo0000446
    1. Curry D. W., Young M. B., Tran A. N., Daoud G. E., Howell L. L. (2018). Separating the Agony from Ecstasy: R(-)-3,4-methylenedioxymethamphetamine Has Prosocial and Therapeutic-like Effects without Signs of Neurotoxicity in Mice. Neuropharmacology 128, 196–206. 10.1016/j.neuropharm.2017.10.003
    1. Danforth A. L., Struble C. M., Yazar-Klosinski B., Grob C. S. (2016). MDMA-assisted Therapy: A New Treatment Model for Social Anxiety in Autistic Adults. Prog. Neuropsychopharmacol. Biol. Psychiatry 64, 237–249. 10.1016/j.pnpbp.2015.03.011
    1. Dolder P. C., Müller F., Schmid Y., Borgwardt S. J., Liechti M. E. (2018). Direct Comparison of the Acute Subjective, Emotional, Autonomic, and Endocrine Effects of MDMA, Methylphenidate, and Modafinil in Healthy Subjects. Psychopharmacol. Berl. 235 (2), 467–479. 10.1007/s00213-017-4650-5
    1. Domes G., Heinrichs M., Gläscher J., Büchel C., Braus D. F., Herpertz S. C. (2007). Oxytocin Attenuates Amygdala Responses to Emotional Faces Regardless of Valence. Biol. Psychiatry 62 (10), 1187–1190. 10.1016/j.biopsych.2007.03.025
    1. Domes G., Lischke A., Berger C., Grossmann A., Hauenstein K., Heinrichs M., et al. (2010). Effects of Intranasal Oxytocin on Emotional Face Processing in Women. Psychoneuroendocrinology 35 (1), 83–93. 10.1016/j.psyneuen.2009.06.016
    1. Dumont G. J., Sweep F. C., van der Steen R., Hermsen R., Donders A. R., Touw D. J., et al. (2009). Increased Oxytocin Concentrations and Prosocial Feelings in Humans after Ecstasy (3,4-methylenedioxymethamphetamine) Administration. Soc. Neurosci. 4 (4), 359–366. 10.1080/17470910802649470
    1. Eckstein M., Becker B., Scheele D., Scholz C., Preckel K., Schlaepfer T. E., et al. (2015). Oxytocin Facilitates the Extinction of Conditioned Fear in Humans. Biol. Psychiatry 78 (3), 194–202. 10.1016/j.biopsych.2014.10.015
    1. Ekman P., Friesen W. V. (1976). Pictures of Facial Affect. Palo Alto: Consulting Psychologists.
    1. Farre M., Abanades S., Roset P. N., Peiró A. M., Torrens M., O'Mathúna B., et al. (2007). Pharmacological Interaction between 3,4-methylenedioxymethamphetamine (Ecstasy) and Paroxetine: Pharmacological Effects and Pharmacokinetics. J. Pharmacol. Exp. Ther. 323 (3), 954–962. 10.1124/jpet.107.129056
    1. Feduccia A. A., Mithoefer M. C. (2018). MDMA-assisted Psychotherapy for PTSD: Are Memory Reconsolidation and Fear Extinction Underlying Mechanisms? Prog. Neuropsychopharmacol. Biol. Psychiatry 84 (Pt), 221–228. 10.1016/j.pnpbp.2018.03.003
    1. Feduccia A. A., Jerome L., Mithoefer M. C., Holland J. (2020). Discontinuation of Medications Classified as Reuptake Inhibitors Affects Treatment Response of MDMA-Assisted Psychotherapy. Psychopharmacology 238, 581–588. 10.1007/s00213-020-05710-w
    1. Glenn C. R., Klein D. N., Lissek S., Britton J. C., Pine D. S., Hajcak G. (2012). The Development of Fear Learning and Generalization in 8-13 Year-Olds. Dev. Psychobiol. 54 (7), 675–684. 10.1002/dev.20616
    1. Glover E. M., Phifer J. E., Crain D. F., Norrholm S. D., Davis M., Bradley B., et al. (2011). Tools for Translational Neuroscience: PTSD Is Associated with Heightened Fear Responses Using Acoustic Startle but Not Skin Conductance Measures. Depress Anxiety 28 (12), 1058–1066. 10.1002/da.20880
    1. Hake H. S., Davis J. K. P., Wood R. R., Tanner M. K., Loetz E. C., Sanchez A., et al. (2019). 3,4-methylenedioxymethamphetamine (MDMA) Impairs the Extinction and Reconsolidation of Fear Memory in Rats. Physiol. Behav. 199, 343–350. 10.1016/j.physbeh.2018.12.007
    1. Holze F., Vizeli P., Müller F., Ley L., Duerig R., Varghese N., et al. (2020). Distinct Acute Effects of LSD, MDMA, and D-Amphetamine in Healthy Subjects. Neuropsychopharmacology 45 (3), 462–471. 10.1038/s41386-019-0569-3
    1. Holze F., Avedisian I., Varghese N., Eckert A., Liechti M. E. (2021). Role of the 5-HT2A Receptor in Acute Effects of LSD on Empathy and Circulating Oxytocin. Front. Pharmacol. 12, 711255. 10.3389/fphar.2021.711255
    1. Hurlemann R., Patin A., Onur O. A., Cohen M. X., Baumgartner T., Metzler S., et al. (2010). Oxytocin Enhances Amygdala-dependent, Socially Reinforced Learning and Emotional Empathy in Humans. J. Neurosci. 30 (14), 4999–5007. 10.1523/JNEUROSCI.5538-09.2010
    1. Hysek C. M., Vollenweider F. X., Liechti M. E. (2010). Effects of a Beta-Blocker on the Cardiovascular Response to MDMA (Ecstasy). Emerg. Med. J. 27, 586–589. 10.1136/emj.2009.079905
    1. Hysek C. M., Domes G., Liechti M. E. (2012a). MDMA Enhances "mind Reading" of Positive Emotions and Impairs "mind Reading" of Negative Emotions. Psychopharmacol. Berl. 222, 293–302. 10.1007/s00213-012-2645-9
    1. Hysek C. M., Simmler L. D., Nicola V. G., Vischer N., Donzelli M., Krähenbühl S., et al. (2012b). Duloxetine Inhibits Effects of MDMA ("ecstasy") In Vitro and in Humans in a Randomized Placebo-Controlled Laboratory Study. PLoS One 7, e36476. 10.1371/journal.pone.0036476
    1. Hysek C. M., Fink A. E., Simmler L. D., Donzelli M., Grouzmann E., Liechti M. E. (2013). α1-Adrenergic Receptors Contribute to the Acute Effects of 3,4-Methylenedioxymethamphetamine in Humans. J. Clin. Psychopharmacol. 33, 658–666. 10.1097/jcp.0b013e3182979d32
    1. Hysek C. M., Schmid Y., Simmler L. D., Domes G., Heinrichs M., Eisenegger C., et al. (2014). MDMA Enhances Emotional Empathy and Prosocial Behavior. Soc. Cogn. Affect Neurosci. 9, 1645–1652. 10.1093/scan/nst161
    1. Janke W., Debus G. (1978). Die Eigenschaftswörterliste. Göttingen: Hogrefe.
    1. JASP Team (2022). JASP (Version 0.16.1) (Amsterdam: University of Amsterdam; ).
    1. Jerome L., Feduccia A. A., Wang J. B., Hamilton S., Yazar-Klosinski B., Emerson A., et al. (2020). Long-term Follow-Up Outcomes of MDMA-Assisted Psychotherapy for Treatment of PTSD: a Longitudinal Pooled Analysis of Six Phase 2 Trials. Psychopharmacol. Berl. 237 (8), 2485–2497. 10.1007/s00213-020-05548-2
    1. Kirkpatrick M. G., Lee R., Wardle M. C., Jacob S., de Wit H. (2014). Effects of MDMA and Intranasal Oxytocin on Social and Emotional Processing. Neuropsychopharmacology 39, 1654–1663. 10.1038/npp.2014.12
    1. Kirsch P., Esslinger C., Chen Q., Mier D., Lis S., Siddhanti S., et al. (2005). Oxytocin Modulates Neural Circuitry for Social Cognition and Fear in Humans. J. Neurosci. 25 (49), 11489–11493. 10.1523/JNEUROSCI.3984-05.2005
    1. Kuypers K. P., de la Torre R., Farre M., Yubero-Lahoz S., Dziobek I., Van den Bos W., et al. (2014). No Evidence that MDMA-Induced Enhancement of Emotional Empathy Is Related to Peripheral Oxytocin Levels or 5-HT1a Receptor Activation. PLoS One 9 (6), e100719. 10.1371/journal.pone.0100719
    1. Lang P. J., Bradley M. M., Cuthbert B. N. (1998). Emotion, Motivation, and Anxiety: Brain Mechanisms and Psychophysiology. Biol. Psychiatry 44 (12), 1248–1263. 10.1016/s0006-3223(98)00275-3
    1. Liechti M. E., Vollenweider F. X. (2000). The Serotonin Uptake Inhibitor Citalopram Reduces Acute Cardiovascular and Vegetative Effects of 3,4-methylenedioxymethamphetamine ('Ecstasy') in Healthy Volunteers. J. Psychopharmacol. 14 (3), 269–274. 10.1177/026988110001400313
    1. Liechti M. E., Baumann C., Gamma A., Vollenweider F. X. (2000). Acute Psychological Effects of 3,4-methylenedioxymethamphetamine (MDMA, "Ecstasy") Are Attenuated by the Serotonin Uptake Inhibitor Citalopram. Neuropsychopharmacology 22 (5), 513–521. 10.1016/S0893-133X(99)00148-7
    1. Maples-Keller J., Watkins L. E., Nylocks K. M., Yasinski C., Coghlan C., Black K., et al. (2022a). Acquisition, Extinction, and Return of Fear in Veterans in Intensive Outpatient Prolonged Exposure Therapy: A Fear-Potentiated Startle Study. Behav. Res. Ther. 154, 104124. 10.1016/j.brat.2022.104124
    1. Maples-Keller J. L., Norrholm S. D., Burton M., Reiff C., Coghlan C., Jovanovic T., et al. (2022b). A Randomized Controlled Trial of 3,4-methylenedioxymethamphetamine (MDMA) and Fear Extinction Retention in Healthy Adults. J. Psychopharmacol. 36 (3), 368–377. 10.1177/02698811211069124
    1. Marsh A. A., Yu H. H., Pine D. S., Blair R. J. (2010). Oxytocin Improves Specific Recognition of Positive Facial Expressions. Psychopharmacol. Berl. 209 (3), 225–232. 10.1007/s00213-010-1780-4
    1. Martins D., Gabay A. S., Mehta M., Paloyelis Y. (2020). Salivary and Plasmatic Oxytocin Are Not Reliable Trait Markers of the Physiology of the Oxytocin System in Humans. Elife 9, e62456. 10.7554/eLife.62456
    1. Mitchell J. M., Bogenschutz M., Lilienstein A., Harrison C., Kleiman S., Parker-Guilbert K., et al. (2021). MDMA-assisted Therapy for Severe PTSD: a Randomized, Double-Blind, Placebo-Controlled Phase 3 Study. Nat. Med. 27 (6), 1025–1033. 10.1038/s41591-021-01336-3
    1. Mithoefer M. C., Wagner M. T., Mithoefer A. T., Jerome L., Doblin R. (2011). 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. J. Psychopharmacol. 25 (4), 439–452. 10.1177/0269881110378371
    1. Mithoefer M. C., Wagner M. T., Mithoefer A. T., Jerome L., Martin S. F., Yazar-Klosinski B., et al. (2013). Durability of Improvement in Post-traumatic Stress Disorder Symptoms and Absence of Harmful Effects or Drug Dependency after 3,4-Methylenedioxymethamphetamine-Assisted Psychotherapy: a Prospective Long-Term Follow-Up Study. J. Psychopharmacol. 27 (1), 28–39. 10.1177/0269881112456611
    1. Mithoefer M. C., Feduccia A. A., Jerome L., Mithoefer A., Wagner M., Walsh Z., et al. (2019). 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 236, 2735–2745. 10.1007/s00213-019-05249-5
    1. Neumann I. D., Maloumby R., Beiderbeck D. I., Lukas M., Landgraf R. (2013). Increased Brain and Plasma Oxytocin after Nasal and Peripheral Administration in Rats and Mice. Psychoneuroendocrinology 38, 1985–1993. 10.1016/j.psyneuen.2013.03.003
    1. Norrholm S. D., Jovanovic T., Olin I. W., Sands L. A., Karapanou I., Bradley B., et al. (2011). Fear Extinction in Traumatized Civilians with Posttraumatic Stress Disorder: Relation to Symptom Severity. Biol. Psychiatry 69 (6), 556–563. 10.1016/j.biopsych.2010.09.013
    1. Oehen P., Traber R., Widmer V., Schnyder U. (2013). A Randomized, Controlled Pilot Study of MDMA (±3,4-Methylenedioxymethamphetamine)-Assisted Psychotherapy for Treatment of Resistant, Chronic Post-traumatic Stress Disorder (PTSD). J. Psychopharmacol. 27 (1), 40–52. 10.1177/0269881112464827
    1. Oeri H. E. (2020). Beyond Ecstasy: Alternative Entactogens to 3,4-methylenedioxymethamphetamine with Potential Applications in Psychotherapy. J. Psychopharmacol. 35, 512–536. 10.1177/0269881120920420
    1. Parrott A. C. (2016). Oxytocin, Cortisol and 3,4-methylenedioxymethamphetamine: Neurohormonal Aspects of Recreational 'ecstasy'. Behav. Pharmacol. 27 (8), 649–658. 10.1097/FBP.0000000000000262
    1. Pineles S. L., Nillni Y. I., King M. W., Patton S. C., Bauer M. R., Mostoufi S. M., et al. (2016). Extinction Retention and the Menstrual Cycle: Different Associations for Women with Posttraumatic Stress Disorder. J. Abnorm Psychol. 125 (3), 349–355. 10.1037/abn0000138
    1. R Core Team (2021). R: A Language and Environment for Statistical Computing (Vienna, Austria: R Foundation for Statistical Computing; ).
    1. Richards A., Inslicht S. S., Yack L. M., Metzler T. J., Russell Huie J., Straus L. D., et al. (2022). The Relationship of Fear-Potentiated Startle and Polysomnography-Measured Sleep in Trauma-Exposed Men and Women with and without PTSD: Testing REM Sleep Effects and Exploring the Roles of an Integrative Measure of Sleep, PTSD Symptoms, and Biological Sex. Sleep 45 (1), zsab271. 10.1093/sleep/zsab271
    1. Risbrough V. B., Glenn D. E., Baker D. G. (2016). On the Road to Translation for PTSD Treatment: Theoretical and Practical Considerations of the Use of Human Models of Conditioned Fear for Drug Development. Curr. Top. Behav. Neurosci. 28, 173–196. 10.1007/7854_2015_5010
    1. Schmid Y., Hysek C. M., Simmler L. D., Crockett M. J., Quednow B. B., Liechti M. E. (2014). Differential Effects of MDMA and Methylphenidate on Social Cognition. J. Psychopharmacol. 28, 847–856. 10.1177/0269881114542454
    1. Sehlmeyer C., Dannlowski U., Schöning S., Kugel H., Pyka M., Pfleiderer B., et al. (2011). Neural Correlates of Trait Anxiety in Fear Extinction. Psychol. Med. 41 (4), 789–798. 10.1017/S0033291710001248
    1. Shin L. M., Rauch S. L., Pitman R. K. (2006). Amygdala, Medial Prefrontal Cortex, and Hippocampal Function in PTSD. Ann. N. Y. Acad. Sci. 1071, 67–79. 10.1196/annals.1364.007
    1. Spielberger C. D., Gorsuch R. C., Lusheme R. E. (1970). Manual for the Stait Trait Anxiety Inventory. Palo Alto: Consulting Psychologists Press.
    1. Stockhorst U., Antov M. I. (2015). Modulation of Fear Extinction by Stress, Stress Hormones and Estradiol: A Review. Front. Behav. Neurosci. 9, 359. 10.3389/fnbeh.2015.00359
    1. Straus L. D., Acheson D. T., Risbrough V. B., Drummond S. P. A. (2017). Sleep Deprivation Disrupts Recall of Conditioned Fear Extinction. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2 (2), 123–129. 10.1016/j.bpsc.2016.05.004
    1. Studerus E., Vizeli P., Harder S., Ley L., Liechti M. E. (2021). Prediction of MDMA Response in Healthy Humans: a Pooled Analysis of Placebo-Controlled Studies. J. Psychopharmacol. 35 (5), 556–565. 10.1177/0269881121998322
    1. Vizeli P., Liechti M. E. (2017). Safety Pharmacology of Acute MDMA Administration in Healthy Subjects. J. Psychopharmacol. 31 (5), 576–588. 10.1177/0269881117691569
    1. Vizeli P., Liechti M. E. (2018). Oxytocin Receptor Gene Variations and Socio-Emotional Effects of MDMA: A Pooled Analysis of Controlled Studies in Healthy Subjects. PLoS One 13 (6), e0199384. 10.1371/journal.pone.0199384
    1. Young M. B., Andero R., Ressler K. J., Howell L. L. (2015). 3,4-Methylenedioxymethamphetamine Facilitates Fear Extinction Learning. Transl. Psychiatry 5, e634. 10.1038/tp.2015.138
    1. Young M. B., Norrholm S. D., Khoury L. M., Jovanovic T., Rauch S. A. M., Reiff C. M., et al. (2017). Inhibition of Serotonin Transporters Disrupts the Enhancement of Fear Memory Extinction by 3,4-methylenedioxymethamphetamine (MDMA). Psychopharmacology 234, 2883–2895. 10.1007/s00213-017-4684-8

Source: PubMed

3
Se inscrever