Skip to main content

REVIEW article

Front. Psychiatry, 12 October 2022
Sec. Psychopharmacology
This article is part of the Research Topic Old and New Psychoactive Substances: Pharmacology and Potential Applications View all 8 articles

A proposed mechanism for the MDMA-mediated extinction of traumatic memories in PTSD patients treated with MDMA-assisted therapy

  • Department of Medical Education, Kirk Kerkorian School of Medicine at UNLV, University of Nevada Las Vegas, Las Vegas, NV, United States

Post-traumatic stress disorder (PTSD) is a devastating psychiatric disorder afflicting millions of people around the world. Characterized by severe anxiety, intrusive thoughts, pervasive nightmares, an assortment of somatic symptoms, associations with severe long-term health problems, and an elevated risk of suicide, as much as 40–70% of patients suffer from refractory disease. 3,4-Methylenedioxy-methamphetamine (MDMA), like classic psychedelics such as psilocybin, have been used to enhance the efficacy of psychotherapy almost since their discovery, but due to their perceived potential for abuse and inclusion on USFDA (United States Food and Drug Administration) schedule 1, research into the mechanism by which they produce improvements in PTSD symptomology has been limited. Nevertheless, several compelling rationales have been explored, with the pro-social effects of MDMA thought to enhance therapeutic alliance and thus facilitate therapist-assisted trauma processing. This may be insufficient to fully explain the efficacy of MDMA in the treatment of psychiatric illness. Molecular mechanisms such as the MDMA mediated increase of brain-derived neurotrophic factor (BDNF) availability in the fear memory learning pathways combined with MDMA's pro-social effects may provide a more nuanced explanation for the therapeutic actions of MDMA.

Introduction

3,4-Methylenedioxy-methamphetamine (MDMA), much like classical psychedelics such as psilocybin, has recently received significant attention as a possible novel therapeutic for treatment-resistant psychiatric illnesses such as PTSD (1, 2). The (classical) psychedelics including psilocybin, N,N′-dimethyltryptamine (DMT), and lysergic acid diethylamide (LSD), have been defined as molecules that produce an altered state of consciousness, primarily mediated by activation of the serotonergic 5HT2A receptor (3, 4). MDMA, while also a hallucinogenic, primarily acts through the inhibition of monoamine reuptake and is not considered a classical psychedelic but rather an “entactogen” due to its powerful pro-social effects (3, 5). MDMA, the primary psychoactive compound in the street drug Ecstasy (or Molly), remains a schedule I medication indicating high abuse potential without an approved medical use despite FDA approval as a breakthrough therapy for PTSD in 2017 (6, 7). Designation as a breakthrough therapy, however, does not guarantee that a pharmaceutical will be approved for use, and only indicates that preliminary clinical evidence suggests that the drug may have a substantial benefit in the improvement of a primary endpoint over current therapies in the treatment of a life-threatening disease (6, 8). Substance misuse in the United States is a significant problem, with an estimated annual financial impact of at least $420 billion dollars, $120 billion of the total in healthcare related expenses (9). MDMA can indeed be a drug of abuse, with frequent users developing tolerance and withdrawal symptoms after sudden cessation of use (10). Further, the consequences of MDMA use are complicated by poly-substance abuse and the frequent impurity of street Ecstasy (1012).

Thus, despite growing evidence that MDMA may have a role in treating serious, debilitating mental illness, the drug is still primarily associated with raves, intoxication, and abuse (13, 14). This may be similar to the stigma associated with the treatment of ADHD with dextroamphetamine despite 40 years of evidence supporting the efficacy of stimulant therapy for the disorder (15, 16). It is therefore of some importance that the molecular basis of therapeutic action and overall clinical efficacy of MDMA as an adjutant treatment for mental illness be further elucidated lest MDMA remain a schedule I drug, barred from use as a therapeutic. As noted by Morgan (17), drugs of abuse such as MDMA must be researched and discussed with care and thus what follows will attempt to clearly delineate between what is well-supported by evidence, what is modestly supported by evidence but in need of additional elucidation, and what is largely speculative. It is therefore the purpose of this narrative review to discuss the use of MDMA-assisted psychotherapy as a treatment for PTSD and suggest a primary mechanism of action supporting the purported efficacy of the drug. It is neither the intention of this review to provide a comprehensive accounting of the proposed mechanisms of action of MDMA as they relate to the treatment of mental illness nor dimmish the importance of the pro-social effect of the drug and the role it may play in facilitating therapeutic alliance. Rather we suggest a relationship between the current model of pathological memory encoding and inhibited memory extinction with subsequent reconsolidation in PTSD and the action of MDMA to reverse or ameliorate this defect (18, 19).

PTSD

Post-Traumatic Stress Disorder (PTSD) is a debilitating, often chronic illness triggered by exposure to a single or a series of traumatic events and is characterized by impaired functioning in multiple functional domains (20). The World Health Organization (WHO) developed the International Classification of Functioning, Disability, and Health (ICF) to provide a common language for use when discussing disability and health which described functional “domains,” or activities and tasks where impairment is disruptive of quality of life (21, 22). A systematic review conducted by Jellestad et al. (23) found PTSD has been associated with impairment in the ICF domains of “General Tasks and Demands, Mobility, Self-Care, Domestic Life, Interpersonal Interactions and Relationships, Major Life Areas and Community, Social and Civic Life” (p. 14). The DSM III criteria for PTSD was based on dysfunctions in three domains: hypervigilance, the re-experiencing of the original trauma, and avoidant behavior of stimuli associated with the original trauma (24). In 1994, the DSM IV revised and expanded the diagnostic criteria for PTSD, which were subsequently widely criticized for reasons as disparate and contradictory as being too narrow in defining trauma to being over-diagnosed and thus too sensitive in populations recently exposed to traumatic events (25, 26). The DSM, published in 2013, again significantly revised the diagnostic criteria for PTSD. Most notably, the disorder was reclassified from an anxiety disorder to a new category dedicated to disorders associated with traumatic events. Briefly, a diagnosis of PTSD can be made when: (a) a person has been exposed to a single traumatic or to repeated traumatic events either directly or indirectly; (b) the person must experience at least one intrusive symptom such as flashbacks; (c) two or more negative mood or cognitive alterations; and (d) two or more symptoms of increased arousal such as hypervigilance (27, 28). Diagnosis can be complicated, however, because the ICD-11 defines PTSD more narrowly; in trauma survivors, it has been suggested that only 42% met both the ICD-11 and DSM-V criteria for PTSD (29). PTSD is also associated with a significant increase in physical/medical morbidity and premature mortality (3032). Patients with a diagnosis of PTSD are at greater risk disorders of multiple organ systems including the musculoskeletal and cardiovascular system and frequently suffer from a variety of comorbid psychiatric illnesses. For patients with recurrent symptoms, PTSD increases the risk of accelerated cognitive decline (dementia) (33). Suicide risk is also thought to be elevated in this patient population, although the high rate of comorbid psychiatric diagnoses has been confounding (34, 35). One recent study, however, reported suicide rates that were 6.74 times higher for men with PTSD and remained elevated at 2.61 times the basal rate in the studied population after controlling for comorbid psychiatric illness (36).

Once diagnosed, the treatment of PTSD is complicated and often ineffective, with between 40 and 70% of patients diagnosed with the disorder suffering recurrent symptoms despite treatment (1, 37, 38). The treatment modality—pharmacological or psychotherapeutic—seems to matter little; even with large effect sizes in RCTs. First line psychotherapies like cognitive processing therapy (CPT) and prolonged exposure therapy (PET) only induce complete remission in 28–40% of patients (39). The lack of agreement between the multiple treatment guidelines produced by organizations as varied as the American Psychiatric Association (APA), the Veterans Health Administration (VA), the American Academy of Family Physicians (AAFP), and several other public and private organizations is problematic. This also creates a dilemma for providers attempting to select appropriate, evidence-based therapies for their patients (40). The VA (41) and APA guidelines, for instance, do not agree on first-line treatment for PTSD, with the APA recommending psychotherapy and pharmacotherapy (42) as equally effective options while the VA considers traumafocused therapies superior to pharmacotherapy (43). It is beyond the scope of this article to describe and compare the many modalities that have been studied for the treatment of PTSD; a summary is provided below.

PTSD treatment

Pharmacological

As of 2019, only two medications– the selective serotonin reuptake inhibitors (SSRI) paroxetine (Paxil®) and sertraline (Zoloft®) have full FDA approval for the treatment of PTSD (43). Although FDA approval has not been conferred, sufficient evidence from randomly controlled trials suggests that Fluoxetine (Prozac®) and the serotonin and norepinephrine reuptake inhibitor (SSNI) venlafaxine may also be effective in the treatment of PTSD. The atypical antipsychotic quetiapine (Seroquel®) may also be superior to placebo as a monotherapy (44, 45). Besides SSRIs, other classes of medications trialed for PTSD treatment include α-adrenoreceptor antagonists (prazosin), atypical antipsychotics (risperidone, olanzapine), atypical antidepressants (trazodone, nefazodone, mirtazapine), MAOIs (brofaromine, phenelzine), tricyclic antidepressants (imipramine), anticonvulsants (topiramate, valproic acid, tiagabine), β-blockers (propranolol), and antihistamines (hydroxyzine) (4446). Recently, molecules with hallucinogenic and/or dissociative properties like ketamine, psilocybin, and MDMA have generated considerable interest as therapeutics for a variety of psychiatric illnesses including PTSD (47, 48). Failure to separate from placebo has been of significant concern in many of the drugs trialed as PTSD treatments; in one study for example, citalopram did not show benefit over placebo but qt prolongation, a known side effect, was seen in the experimental arm of the trial (43). In general, most of the medications trialed for PTSD treatment demonstrated improvements in clinician or patient rated symptom scales but did not show benefit over placebo (46). Huang et al. (46) and Hoskins et al. (45) conducted thorough meta-analyses of the efficacy of pharmacotherapy for PTSD and are excellent sources for addition information regarding the medical treatment of the disorder.

Psychotherapies

As noted above, psychotherapy is a first-line treatment for PTSD. Broadly, psychotherapies for PTSD can be divided into two categories: Trauma-focused treatments and non-trauma-focused treatments (40). In trauma-focused therapy, the traumatic event and accompanying negative emotions are directly confronted while non-trauma-focused therapies instead target PTSD symptomatology and maladaptive coping mechanisms without addressing the underlying insult (49, 50). Although it has been suggested that trauma-focused therapy might be less well-tolerated by PTSD patients and therefore lead to increased treatment drop out, patients prematurely terminate treatment with both trauma-focused and non-trauma-focused at equal rates (51, 52). Numerous psychotherapeutic modalities have been tested for the treatment of PTSD; a recent systematic review and meta-analysis by Lewis et al. (50) included studies of 29 discreet interventions. Of the modalities reviewed, Cognitive Processing Therapy (CPT), Prolonged Exposure Therapy (PE), Cognitive Therapy (CT), and Eye Movement Desensitization Therapy (EMDT) had the most robust data supporting their efficacy with several additional modalities demonstrating promising efficacy, but requiring additional study (28, 40, 50). Analysis of long-term psychotherapeutic treatment for PTSD supports the efficacy of the same treatment modalities (49). Mindfulness-based therapies such as Mindfulness-Based Stress Reduction (MBSR) are becoming of interest as an alternative to the techniques above, particularly for patients who prefer a non-trauma-focused approach; the majority of highly effective psychotherapeutic modalities for PTSD are trauma-focused (39).

Pharmacological-assisted psychotherapy

The use of psychedelics to enhance the clinical benefits of psychotherapy is not a new idea, with experiments in the 1950–1970's trialing “classic” psychedelics like LSD in the treatment of several psychiatric illnesses (14, 53). The scheduling of LSD, MDMA, and other psychedelics has made research more difficult, but these molecules have recently become of increased interest due to the need for more effective treatments for psychiatric illnesses like PTSD (54, 55). A simple search of PubMed using the term “psychedelics” returned 1,510 possible matches published in the past 12 months alone. Psychotherapy “enhanced” or assisted by classical psychedelics like psilocybin or non-classical hallucinogenic molecules such as Ketamine and MDMA (referred to hereafter as “psychedelic-assisted therapy” for simplicity) may be the most promising use of these medications (45, 5557). In psychedelic-assisted psychotherapy, patients usually attend a limited number of “preparatory” psychotherapy sessions followed by administration of a fixed dose of a psychedelic in a supportive setting where therapists are present throughout the experience; often the psychedelic-enhanced session may last as long as 8–12 h and involve two or more therapists working in shifts, acting as facilitators of the session and not active directors (17). Patients then attend follow-up sessions after their psychedelic experience (58, 59). Although randomly controlled studies of psychedelic-assisted therapy have generally been small, the results suggest that the treatment modality produces significant and durable improvements in psychiatric symptomology. For example, one 2011 quasi-crossover randomly controlled trial of 20 patients diagnosed with treatment resistant PTSD treated with MDMA-assisted psychotherapy found a clinically significant decrease in Clinician-Administered PTSD Scale scores in 83% in the treatment arm vs. 25% in the placebo arm. Long-term follow up found that 14/19 patients enrolled in the trial had a durable improvement of PTSD symptoms three and half years after treatment (60, 61). Although still potentially under-powered, a meta-analysis of six stage-two clinical trials found that 53% of subjects in the active treatment groups no longer met the clinical criteria of PTSD as much as 6 years after treatment with MDMA-assisted psychotherapy as compared to 23% in controls (17, 18). Based on the pooled results of similar trials, the Food and Drug Administration (FDA) approved MDMA-assisted therapy as a break-through treatment for PTSD (6). Studies are ongoing, investigating both the overall efficacy of psychedelics in the treatment of PTSD as well as their effect on specific symptoms such as disturbed sleep and persistent nightmares (62).

MDMA

History and chemistry

3,4-methylenedioxymethamphetamine (MDMA) is a phenethylamine amphetamine derivative with structural similarity to classic psychedelics like mescaline (6365). MDMA was synthesized for the first time in 1912 by chemist Arthur Kollisch and patented by Merck as a potential hemostatic agent (66). Alexander T. Shulgin “rediscovered” MDMA in 1965 after synthesizing the related compounds 3,4-methylenedioxyethylamphetamine (MDE), 3-methoxy-4,5-methylenedioxyamphetamine (MMDA), and 3,4-methylenedioxyamphetamine (MDA) in the early 1960's. MDMA can be synthesized via a variety of processes that generally result in a racemic mixture of products. More recently, the production of enantiopure MDMA has increased to explore the differential pharmacology of the (S) and (R) forms. Differential binding of the (S) and (R) isomers occurs at the 5HT1 and 5HT2 receptors but the psychopharmacological effects of the stereospecific binding of MDMA are unclear. The (S) isomer may be associated with the empathic and psychostimulant of MDMA and possibly the (R) isomer with the hallucinogenic effects of the drug (63); Lyon et al. (67, 68). Shulgin synthesized and investigated the psychoactive properties of over 200 compounds from 1960 to 1993, including MDMA and was the first to note the unique properties of MDMA intoxication. Historical reconstruction suggests that MDMA was first used as a street drug in the late 1960's, and increasingly detected in seized illicit substances by forensic laboratories throughout the 1970's (66). Like LSD before it, in the 1980's MDMA was explored as a possible pharmacotherapy for a variety of psychiatric disorders and as an adjunctive to talking therapy. Unfortunately, MDMA rebranded as “Ecstasy,” became popular as a recreational drug in the club scene of the 1980's and by 1985 had been designated as a Schedule I compound with high abuse potential without any known medical use (14). As of 2004, a reported 11 million people in the United States had experimented with MDMA at some point in their lives (69). By 2011, the number of Americans reporting experimentation with MDMA increased to 14.5 million (10).

Physiological and psychological effects

As noted above, the effects of MDMA consumption are unique, with stimulatory, hallucinogenic, and pro-social components that defy the easy categorization and inclusion of MDMA in other classes of drugs such as the “classical” psychedelics or other amphetamine-based, psycho-stimulant molecules (13, 64). The pro-social effects of MDMA have resulted in its description as a novel drug termed an “Entactogen” or “empathogen” (14, 64, 70). The subjective effects of MDMA have been amply described in numerous studies, with sexual arousal, euphoria, relaxation, sensory enhancement, and mild visual hallucinations frequently described (10, 71, 72). Importantly, particularly in the setting of its use as an adjunctive to talking theory, is the increased sociability induced by MDMA (71). As noted by Vegting et al. (73), ecstasy comes from the Greek “εκστασις” (ekstasis) which roughly translates as “standing outside of yourself” (p. 3,473). The pro-social effects of MDMA have also been well-described and include an enhanced sense of trust, improved empathy, an increased sense of closeness and inclusion, and a general increase in social confidence/perceived social competence (11, 12, 70). Kamilar-Britt and Bedi (70) provide an excellent review of lab and clinical research regarding the pro-social effects on MDMA. Physiologically, not all the effects of MDMA are reported as positive by its users, with undesirable effects such as headache, nausea, bruxism, trismus, agitation, and dry mouth, racing heart, fever, chills, and insomnia sometimes experienced (10, 12, 74). Further, long-term use of MDMA has been associated with multiple sequelae including depressed mood and anxiety; cognitive deficits in working memory, attention, and verbal processing; and a reduction in serotonin transporters (SERTs) resulting in escalating tolerance (75). Multiple studies have explored the potential neurotoxicity of MDMA in the setting of long-time use (24, 69, 76). Thus, it is important to balance the therapeutic benefits of MDMA against the negative effects of chronic use– in general; the use of MDMA to treat mental illness is limited to two or three carefully controlled administrations thus alleviating concern for the long-term consequences sometimes experienced by recreational users (14). Recently, Mitchell et al. (77) reported the results of a randomized, double blind, multi-site, placebo-controlled stage three clinical trial (n = 90) of MDMA for the treatment of severe PTSD. The authors concluded that MDMA as compared to placebo produced statistically significant improvement in PTSD symptom serenity as measured by the Clinician-Administered PTSD Scale for DSM-V (CAPS-5), the primary endpoint of the study (77).

Biochemistry and pharmacology

The psychological and physiological effects of MDMA are a result of a complex set of only partially understood interactions between the drug and multiple neurotransmitter pathways (69). MDMA readily crosses the blood-brain barrier and binds to multiple targets with varying affinity, producing its effects in a dose-dependent manner (78). Like other psychostimulants, MDMA (the [+] enantiomer) binds with high affinity to monoamine transporters; the drug has the greatest affinity for the norepinephrine transporter (NET; KM = 225 ± 113 nM), followed closely by the dopamine transporter (DAT; KM = 444 ± 227 nM) and the serotonin transporter (SERT; KM = 447 ± 197 nM) (79). These transporters clear monoamines from the synaptic cleft and move them across the neuronal membrane where they are subsequently repackaged into storage vesicles by the vesicular monoamine transporter (VMAT) for later release (80). Verrico et al. (79) noted, however, that binding site affinity and subsequent transporter blockade do not necessarily have a linear correlation, suggesting that full inhibition was inducible regardless of affinity. However, as opposed to stimulants like cocaine that only blockade monoamine transporters, MDMA is a full substrate of SERT, DAT, and NET; it not only competes with endogenous monoamine substrates, upon binding it reverses the direction of transport and induces non-exocytic/non-vesicular, transporter-mediated release of serotonin, dopamine, and norepinephrine into the synaptic cleft (76, 81, 82). Additionally, MDMA binds to the VMAT2 transporter and by reversing the direction of transport and reducing the pH gradient across the vesicular membrane, induces the release of monoamines into the cytosolic pool of the neuron (83). The combination of inhibition and reversal of reuptake and the suppression of vesicular sequestration of monoamines inside the neuron results in a rapid increase in neurotransmitter levels at the synaptic cleft, producing many of the acute effects of MDMA (81). MDMA is also thought to bind directly to the dopamine D1 and D2, β-adrenergic, α2-adrenergic, serotonin 5HT2A, serotonin 5HT2B, and H1-histaminergic receptors. Activity at these receptors is relatively weak however, and it is not yet clear how interaction at these receptors contributes to the effects of the drug (78, 84, 85). Recent studies suggest that MDMA may also be active at the trace-amine associated receptor (TAAR1) and the Sigma-1 receptor as well (63). Significant increase in serum levels of oxytocin, prolactin, dehydroepiandrosterone (DHEA), vasopressin, and cortisol are also seen after MDMA ingestion, but these increases may be a result of increased serotonergic activity and not a direct effect of the drug itself. This conclusion is supported by a recent study that found that oxytocin release and the pro-social behavior resulting from increased plasma levels of oxytocin was extinguished by administering a 5HT1A antagonist in a murine model (63). These acute physiological effects account for the transient euphoria, increased sociability, and hallucinations induced by MDMA, but they do not provide an immediate explanation for the efficacy of MDMA-assisted therapy as a treatment for PTSD (18).

Fear learning and extinction

Although it is beyond the scope of this review to extensively explore the complex bi-directional interaction between emotion, physiology, and memory, decades of research support the theory that emotional and physiological state determine in part what is remembered (8688). Importantly, emotional state at the time of memory formation may be retrieved and re-experienced when external stimuli similar to those present when the memory was encoded are encountered again (89, 90). As noted by Engen and Anderson (89), exogenously stimulated emotional states and the episodic memories that often accompany these states can be disruptive, with perturbations of equilibrium inversely correlated with an individual's ability to self-regulate their emotional state. Fear is an essentially universal response to perceived threats that allows animals to adopt an appropriate defensive posture preparatory to fighting or fleeing without the time lag associated with conscious cognitive processing. Fear learning, or the association of an unconditioned stimulus (US) with a conditioned stimulus (CS), is an adaptative process that allows organisms—including humans—to reflexively respond to an initially novel environmental threat should exogenous cues similar to those present at the original encounter be encountered again (90, 91). These learned responses can persist for a lifetime (92).

Fear learning is thought to be regulated by a neural network connecting the amygdala, the medial pre-frontal cortex (mPFC), and the hippocampus (93, 94). The amygdala, a part of the limbic system, is a complex collection of nuclei that participates in memory processing, emotional expression, and the management of sensory input. It is located superior to the brainstem, and is an evolutionarily older structure (95). The pre-frontal cortex is a “younger” part of the brain, located deep to the frontal bone in the anterior-most part of the brain, and is thought to be the part of the brain from which cognition, decision-making, and other complex, higher-function processes arise (96). The hippocampi are two mirrored structures consisting of three parts: the cornu ammonis, dentate gyrus, and subiculum (97). The role of these structures in fear memory learning has been elucidated primarily using disruptive pharmaceuticals and surgical lesioning techniques to demonstrate the inhibition of the development of a persistent fear response when any of these pathways are interrupted (90). Much simplified, the fear learning process integrates sensory information coding for both the US and CS, sent to the lateral nucleus (LA) of the amygdala both directly from the primary sensory cortices and thalamus and indirectly through the cortico-amygdala pathway. The overlapping US and CS signals strengthen neuronal connections in these pathways, potentiating the fear-learning process; neuronal plasticity at the molecular level is thought to account for this effect (98102). The signals arising in the LA are then relayed to the basolateral (BLA) and central nucleus of the amygdala, the later triggering the physiological fear response through projections to the hypothalamus and other targets in the brainstem. The BLA connects to the ventral hippocampus and plays a critical role in attaching context-related cueing to the fear response (90, 91, 103). The medial pre-frontal cortex also connects directly to the BLA, modulating behavioral responses to fear and plays a critical role in the extinction of fear memory learning (90). In molecular terms, multiple pathways and neuromodulators are implicated in the process, including brain-derived neurotrophic factor (BDNF) and its associated receptor tropomyosin-related kinase B (Trk-B), multiple gene transcription factors, and a diverse set of metabotropic and ionotropic receptors (100, 104). Cahil and Milton (104) have provided a comprehensive summary of these molecular mechanisms.

As noted above, although fear memory learning is often adaptive, the conditioned response to benign stimuli can be extremely disruptive; once a threat is no longer present, it is maladaptive to retain an associated fear response (101, 105). Thus, it is also adaptive that fear memory learning can be “unlearned,” a process termed extinction (106, 107). Fear extinction has been defined as “…a lessening of conditioned fear responses following extinction training, during which subjects are exposed to repetitive presentations of conditioned stimuli (CS) alone” [(110), p. 1]. Although the process is not entirely understood, fear extinction has been thought to involve either the uncoupling of the response from the fear memory, the selective erasure of the fear memory, or perhaps both (92). Memory retrieval is an active process, in which the recalled memory is destabilized transiently, restabilized in a process called reconsolidation, then returned to quiescent, long-term storage state (108). Recently, Kida (108, 109) has presented data that suggested that fear memory extinction is better characterized as a learning event in which an inhibitory memory is encoded by the same process, in the same brain structures, as the original fear memory. Supporting this hypothesis, a study of the role of dopamine in fear memory extinction concluded that dopaminergic neurons projecting from the BLA and a set of contiguous structures (intercalated cell masses), combined with glutaminergic neurons in the mPFC, specifically the infra-limbic subregion, were all highly active during fear memory extinction. Selectively ablating these neurons suppressed the extinction of fear memories in a murine model (105, 110, 111). Further, multiple studies have suggested that synaptic plasticity in the mPFC, the hippocampus, and the BLA is as essential in extinguishing fear memories as it is in forming them (99101).

Pathological fear learning and extinction in PTSD

The pathophysiology of PTSD, while well-studied, remains incompletely understood (112). As noted by Krystal et al. (113), defining PTSD solely as a deficit in fear memory learning extinction is likely too narrow. Nevertheless, the fear memory learning pathways discussed above demonstrate significant, apparently pathological changes leading to dysfunctional fear memory learning defects in patients suffering from the illness (93, 114, 115). It has been suggested that the intrusive symptoms of PTSD—disturbing nightmares, upsetting memories, and uncontrollable flashbacks—have been correlated with impairment of fear memory extinction and may also underly additional symptoms such as pervasive feelings of guilt and shame, anxiety, and predilections for uncontrollable anger and substance abuse (38, 109, 115118). Several review articles have explored the central role of impaired fear memory learning extinction in the pathogenesis of PTSD (93, 113, 116, 118). These studies found moderately consistent evidence that there are pathological changes in the amygdala, hippocampus, and pre-frontal cortex at the functional, structural, and molecular levels.

Functionally, PTSD patients have displayed increased amygdala activation as assessed by fMRI during fear conditioning as opposed to healthy, non-trauma exposed controls (93, 115, 117119). Hyperactivity in the BLA in particular may be associated with increased fear response to environmental conditions and a corresponding tendency to generalize threat responses to benign stimuli (93, 115, 118, 120, 121). It has also been suggested that in PTSD patients, the amygdala shows increased responsiveness to negative stimuli in general (120). Several studies have also found increased activity during fear conditioning in the dorsal anterior cingulate cortex (dACC) and reduced responsiveness in the ventral anterior cingulate cortex during fear memory extinction (114, 115, 122). Reduced activity in the ventral-medial pre-frontal cortex (vmPFC) has been reported in several fMRI studies, particularly during extinction training recall (93, 115, 117). It has been suggested that in healthy patients, the vmPFC suppresses the amygdala's response to negative stimuli, and regulates fear memory extinction and extinction recall (93, 114, 118, 123). Hippocampal hypoactivity has also been found in PTSD patients, possibly indicating impairment of context-dependent memory encoding during both fear memory learning and extinction. Further, like the vmPFC, the hippocampus is thought to regulate activity in the amygdala and thus hippocampal dysfunction may also play a role in amygdala hyperactivity (115117, 124). Additionally, functional impairment in connectivity between these brain regions and others may significantly contribute to the symptomology of PTSD (101, 113, 122, 123). It is important to note that there has been variability in fMRI results, measuring brain activity in PTSD patients. The divergence in findings may be due to atypical vs. typical PTSD presentations, genetic variations resulting in baseline differences between patients, the stimuli used to elicit fear responses in the patients, and variances in experimental setting (93). Likewise, structural analysis of the brain regions involved in fear memory learning produced mixed and sometimes conflicting results (93, 121, 125). Regardless of specific pathology, functional dysregulation of fear memory learning is consistent in patients with PTSD.

In PTSD, pathological changes at the neuronal/molecular level are likewise complex and have been extensively explored in murine models (112, 118, 126). Further, alterations in gene expression, receptor populations, neurotrophic molecule levels, neurotransmitter levels, and neuro-remodeling via changes in dendritic spine density have been found to be exposure phase-dependent; upregulations seen shortly after exposure to trauma may be transient and later resolve (109). Additionally, the population of receptors, ligands, and intracellular signaling pathways varies depending on the mechanism in question. For example, in the amygdala, the mTOR signaling pathway, the β-adrenergic receptor, the glucocorticoid receptor, β-arrestin-2, and altered glutaminergic signaling have been associated with hyperresponsiveness to negative stimuli and other PTSD-associated symptoms. An entirely different list including dopamine receptors D1 and D5 have been associated with the memory destabilization necessary for extinction and reconsolidation (109, 119). A loss of synaptic plasticity secondary to chronic inflammation, reductions in the absolute number of α-amino-3-hydroxy-5-methyl-4-isoxazloepropionic acid receptors (AMPARs) and N-methyl-D-aspartate receptors (NMDARs), reduced activity of neurotrophic molecules, increased excitotoxicity, and reduced dendritic spine density and arborization has been associated with a number of pathologies in PTSD (101, 109, 112, 118).

Brain-derived neurotrophic factor (BDNF) and its associated receptor, tropomyosin receptor kinase B (TrkB), have come under increasing scrutiny for their role in several psychiatric disorders including PTSD (100, 104, 107, 113, 127131). The synthesis of BDNF precursor pro-BDNF occurs in microglia, astrocytes, and glutaminergic neurons which is secreted along with the mature form of the neurotrophin, both of which are active (127). BDNF signaling is associated with multiple aspects of neuroplasticity including the upregulation and phosphorylation of NMDARs and increases in the complexity, size, and number of dendritic spines at the synapse and is necessary for long-term potentiation and memory consolidation (132, 133). In PTSD, BDNF in the hippocampus is essential for fear memory extinction, and it has been suggested that a deficit of BDNF may underly hippocampal synaptic loss and the subsequent inhibition of the extinction process (113, 127, 129). The impact of BDNF activity in the amygdala (and the dACC) is less clear in PTSD and is locus dependent. BDNF in the BLA is active during the formation of a fear memory and thus an excess could be associated with worsening of PTSD symptomatology (128). Conversely, several studies have demonstrated that much like in the hippocampus, BDNF availability in the amygdala is critical for fear memory extinction and when blocked, disrupts extinction (134). Likewise, decreased BDNF activity in the vmPFC has been associated with defective fear memory extinction– in murine model, mice with BDNF Val66Met mutation that downregulates the activity-dependent secretion of the neurotrophin had reduced activity in the vmPFC during dear extinction processing (128). Additional mouse studies have demonstrated that reduced expression of BDNF has been correlated with dendritic arbor deficiencies in the hippocampus and vmPFC, inducing PTSD-associated behavioral patterns (135). Interestingly, there is conflicting data as to whether serum and/or plasma levels of BNDF are increased or decreased in PTSD, with several studies supporting both positions (128, 130). Regardless, the evidence strongly supports the assertion that aberrant BDNF signaling is a part of the pathology of PTSD; the neuroplasticity required to form new fear memory associations requires the presence of BDNF as does the neuroplasticity required for extinction learning and memory reconsolidation. In essence, the reduction of BDNF availability after trauma-induced fear memory learning “locks” the conditioned response in place, preventing extinction and memory reconsolidation.

Traumatic memory extinction and reconsolidation mediated by MDMA

As discussed above, the efficacy of MDMA as an adjunctive enhancer of psychotherapy in the treatment of PTSD is mediated by multiple mechanisms, some of which likely remain unexplored. Many mechanisms have been postulated. Smith et al. (136), for instance suggest that MDMA bilaterally suppresses amygdala activation and thus dampens negative emotions while simultaneously enhancing responsiveness to positive emotion thus facilitating psychotherapy. Wagner et al. (57) similarly support the idea that immediate and long-term potentiation of (interpersonal) openness and repressed neuroticism mediated by serotonin and oxytocin release may explain MDMA's effect in improving PTSD symptoms. Vermetten and Yehuda (137) likewise suggest that the altered state of consciousness induced by MDMA “can facilitate a deeper psychotherapeutic process” (p. 231). The pro-social effects of MDMA are well-known and likely contribute, perhaps significantly, to efficacy of MDMA-assisted psychotherapy (138, 139). In this model, the primary effect of MDMA in the treatment of PTSD is to enhance the strength of the therapeutic alliance between patient and provider (139). This effect alone, however, does not comfortably account for the effect of MDMA as a therapeutic in light of the functional, structural, and molecular neuronal deficits found in PTSD, particularly in the fear memory leaning and extinction pathways.

However, like other psychedelics such as psilocybin, MDMA has been demonstrated to induce increases in neural plasticity (63, 140144)). Ly et al. (145) and Olsen (146) suggested that the term “psychoplastogen” might be an appropriate label for molecules such as MDMA, psilocybin, and ketamine because of their profound neurogenic effects. The authors further noted that the neuroplastic effects of the “psychoplastogens” like MDMA are BDNF-dependent; even some studies discussing the addictive and neurotoxic effects of MDMA have noted the alterations in BDNF expression in many regions of the brain after administration of the drug in murine models (140, 145148). Despite some evidence that serotonergic signaling and transport plays a vital if not necessary role in MDMA's ability to enhance (or at least dis-inhibit) fear memory extinction, BDNF signaling was the necessary component for the effect to occur (107, 140, 145). Multiple studies found increased BDNF protein levels or increased BDNF mRNA transcripts in regions of the brain critical to fear learning such as the hippocampus and the vmPFC after MDMA treatment. Dunlap et al. (63) for instance noted that significantly increased levels of BDNF mRNA transcripts were found in the prefrontal cortex of rats after MDMA administration, with increases of BDNF's target receptor TrkB also noted 24 h after drug treatment. Although Martinez-Turrillas (149) conversely found decreased levels of BDNF in the hippocampus in their study, other authors have found the opposite and report increased Hippocampal BDNF in the dentate gyrus and CA3 specifically (150). In these areas of the brain where it has been suggested that the pathology of PTSD induces neuronal loss and degraded synaptic connections, upregulation of BDNF mediated by MDMA has resulted in increased synaptic connectivity and growth in dendritic spine density and number (145, 150). Further supporting the importance of BDNF in the neuroplastic and therefore therapeutic effects of MDMA, the inhibition of BDNF, its receptor TrkB, or TrkB's downstream signaling molecule mTOR resulted in complete inhibition of psychedelic-induced neuronal plasticity (107, 140, 143, 145, 146). Thus, MDMA, by upregulating or at least restoring normal levels of BDNF in the amygdala, hippocampus, and vmPFC, may “unlock” the trauma-induced fear memory association and allows for extinction learning and subsequent reconsolidation of the memory. This mechanism, in addition to the pro-social effects that allow for a deeper therapeutic alliance between patient and provider may account for a significant part of the efficacy of MDMA in the treatment of PTSD.

Discussion and conclusion

MDMA-assisted psychotherapy, though approved as a breakthrough treatment by the FDA, will require significant additional research before it can be accepted as a first-line option for refractory PTSD (6, 17). Nevertheless, over the past two decades, enough evidence has accumulated to postulate empirically supported rationale for the reported efficacy of the adjunctive use of MDMA. While the pro-social effects of MDMA are profound and likely substantially improve therapeutic alliance, without an additional mechanism of action working at the molecular level, it is difficult to account for the large effect size reported for MDMA-assisted psychotherapy. BDNF-facilitated neuroplasticity aligns a key pathogenic mechanism in PTSD (dysfunction in the fear memory pathways) with a known drug effect for MDMA and other psychedelics. As is often the case, the evidence here to date only demonstrates correlation and not causation and thus it is not possible to state categorically that BDNF and subsequent increases in neuronal connectivity and dendritic spine density are the motive force behind the positive effects of the treatment. Further, the direct effects of MDMA on various receptors and the massive increase in neurotransmitter availability at the synapse has not been addressed here and cannot be discounted as another explanatory mechanism for treatment efficacy (151). The difficulty in correlating molecular studies in murine models with human behavior is likewise a problem as it often is in the treatment of psychiatric illness. These limitations aside, it seems likely that BDNF modulation by MDMA accounts for at least a part of the theoretic benefits associated with MDMA-assisted psychotherapy for the treatment of PTSD. Future studies will continue to sharpen the model presented here, or suggest an alternative model; either way, MDMA-assisted therapy provides another option for patients suffering from PTSD that have failed multiple treatment regimens.

Author contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Conflict of interest

The 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.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Krediet E, Bostoen T, Breeksema J, van Schagen A, Passie T, Vermetten E. Reviewing the potential of psychedelics for the treatment of PTSD. Int J Neuropsychopharmacol. (2020) 23:385–400. doi: 10.1093/ijnp/pyaa018

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Sessa B. MDMA and PTSD treatment: PTSD: from novel pathophysiology to innovative therapeutics. Neurosci Lett. (2017) 649:176–80. doi: 10.1016/j.neulet.2016.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Dos Santos RG, Bouso JC, Rocha JM, Rossi GN, Hallak JE. The use of classic hallucinogens/psychedelics in a therapeutic context: healthcare policy opportunities and challenges. Risk Manag Healthc Policy. (2021) 14:901–10. doi: 10.2147/RMHP.S300656

PubMed Abstract | CrossRef Full Text | Google Scholar

4. McClure-Begley, Roth BL. The promises and perils of psychedelic pharmacology for psychiatry. Nat Rev Drug Discov. (2022) 21:463–73. doi: 10.1038/s41573-022-00421-7

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Nichols DE. Entactogens: how the name for a novel class of psychoactive agents originated. Front Psychiatry. (2022) 13:863088. doi: 10.3389/fpsyt.2022.863088

PubMed Abstract | CrossRef Full Text | Google Scholar

6. 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. Front Psychiatry. (2019) 10:650. doi: 10.3389/fpsyt.2019.00650

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Kenny BJ, Zito PM. Controlled Substance Schedules. In StatPearls. (2021). Available online at: https://www.ncbi.nlm.nih.gov/books/NBK538457/ (accessed Jul 29, 2021).

Google Scholar

8. Food US and Drug Administration (2018). Breakthrough Therapy. Available online at: https://www.fda.gov/patients/fast-track-breakthrough-therapy-accelerated-approval-priority-review/breakthrough-therapy

Google Scholar

9. McLellan AT. Substance misuse and substance use disorders: why do they matter in healthcare? Trans Am Clin Climatol Assoc. (2017) 128:112–30.

PubMed Abstract | Google Scholar

10. Meyer JS. 3,4-methylenedioxymethamphetamine (MDMA): current perspectives. Subst Abuse Rehabil. (2013) 4:83–99. doi: 10.2147/SAR.S37258

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Dolan SB, Chen Z, Huang R, Gatch MB. “Ecstasy” to addiction: Mechanisms and reinforcing effects of three synthetic cathinone analogs of MDMA. Neuropharmacology. (2018) 133:171–80. doi: 10.1016/j.neuropharm.2018.01.020

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Kirkpatrick MG, de Wit H. MDMA: a social drug in a social context. Psychopharmacology. (2015) 232:1155–63. doi: 10.1007/s00213-014-3752-6

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Kalant H. The pharmacology and toxicology of “ecstasy” (MDMA) and related drugs. CMAJ. (2001) 165:917–28. Available online at: https://www.cmaj.ca/content/cmaj/165/7/917.full.pdf

PubMed Abstract | Google Scholar

14. Sessa B, Higbed L, Nutt D. A review of 3,4-methylenedioxymethamphetamine (MDMA)-assisted psychotherapy. Front Psychiatry. (2019) 10:138. doi: 10.3389/fpsyt.2019.00138

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Mueller AK, Fuermaier AB, Koerts J, Tucha L. Stigma in attention deficit hyperactivity disorder. Atten Defic Hyperact Disord. (2012) 4:101–14. doi: 10.1007/s12402-012-0085-3

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Osland ST, Steeves TD, Pringsheim T. Pharmacological treatment for attention deficit hyperactivity disorder (ADHD) in children with comorbid tic disorders. Cochrane Database Syst Rev. (2018) 6:CD007990. doi: 10.1002/14651858.CD007990.pub3

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Morgan L. MDMA-assisted psychotherapy for people diagnosed with treatment-resistant PTSD: what it is and what it isn't. Ann Gen Psychiatry. (2020) 19:33. doi: 10.1186/s12991-020-00283-6

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Feduccia AA, Mithoefer MC. MDMA-assisted psychotherapy for PTSD: are memory reconsolidation and fear extinction underlying mechanisms? Prog Neuropsychopharmacol Biol Psychiatry. (2018) 84:221–8. doi: 10.1016/j.pnpbp.2018.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Luoma JB, Shahar B, Kati Lear M, Pilecki B, Wagner A. Potential processes of change in MDMA-Assisted therapy for social anxiety disorder: enhanced memory reconsolidation, self-transcendence, therapeutic relationships. Hum Psychopharmacol. (2022) 37:e2824. doi: 10.1002/hup.2824

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Sherin JE, Nemeroff CB. Post-traumatic stress disorder: the neurobiological impact of psychological trauma. Dialogues Clin Neurosci. (2011) 13:263–78. doi: 10.31887/DCNS.2011.13.2/jsherin

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Ustün TB, Chatterji S, Kostansjek N, Bickenbach J. WHO's ICF and functional status information in health records. Health Care Financ Rev. (2003) 24:77–88.

PubMed Abstract | Google Scholar

22. Vargus-Adams JN, Majnemer A. International Classification of Functioning, Disability and Health (ICF) as a framework for change: revolutionizing rehabilitation. J Child Neurol. (2014) 29:1030–5. doi: 10.1177/0883073814533595

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Jellestad L, Vital NA, Malamud J, Taeymans J, Mueller-Pfeiffer C. Functional impairment in posttraumatic stress disorder: a systematic review and meta-analysis. J Psychiatr Res. (2021) 136:14–22. doi: 10.1016/j.jpsychires.2021.01.039

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Carvajal C. Posttraumatic stress disorder as a diagnostic entity - clinical perspectives. Dialogues Clin Neurosci. (2018) 20:161–8. doi: 10.31887/DCNS.2018.20.3/ccarvajal

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Kuester A, Köhler K, Ehring T, Knaevelsrud C, Kober L, Krüger-Gottschalk A, et al. Comparison of DSM-5 and proposed ICD-11 criteria for PTSD with DSM-IV and ICD-10: changes in PTSD prevalence in military personnel. Eur J Psychotraumatol. (2017) 8:1386988. doi: 10.1080/20008198.2017.1386988

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Pai A, Suris AM, North CS. Posttraumatic stress disorder in the DSM-5: controversy, change, conceptual considerations. Behav Sci. (2017) 7:7. doi: 10.3390/bs7010007

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Sareen J. Posttraumatic stress disorder in adults: impact, comorbidity, risk factors, and treatment. Can J Psychiatry. (2014) 59:460–7. doi: 10.1177/070674371405900902

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Schrader C, Ross A. A review of PTSD and current treatment strategies. Mo Med. (2021) 118:546–551.

PubMed Abstract | Google Scholar

29. Bryant RA. Post-traumatic stress disorder: a state-of-the-art review of evidence and challenges. World Psychiatry. (2019) 18:259–69. doi: 10.1002/wps.20656

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Giesinger I, Li J, Takemoto E, Cone JE, Farfel MR, Brackbill RM. Association between posttraumatic stress disorder and mortality among responders and civilians following the september 11, 2001, disaster. JAMA Netw Open. (2020) 3:e1920476. doi: 10.1001/jamanetworkopen.2019.20476

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Lohr JB, Palmer BW, Eidt CA, Aailaboyina S, Mausbach BT, Wolkowitz OM, et al. Is post-traumatic stress disorder associated with premature senescence? A review of the literature. Am J Geriatr Psychiatry. (2015) 23:709–25. doi: 10.1016/j.jagp.2015.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

32. McFarlane AC. The long-term costs of traumatic stress: intertwined physical and psychological consequences. World Psychiatry. (2010) 9:3–10. doi: 10.1002/j.2051-5545.2010.tb00254.x

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Nilaweera D, Freak-Poli R, Ritchie K, Chaudieu I, Ancelin M-L, Ryan J, et al. The long-term consequences of trauma and posttraumatic stress disorder symptoms on later life cognitive function and dementia risk. Psychiatry Res. (2020) 294:113506. doi: 10.1016/j.psychres.2020.113506

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Bisson JI, Cosgrove S, Lewis C, Robert NP. Post-traumatic stress disorder. BMJ. (2015) 351:h6161. doi: 10.1136/bmj.h6161

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Holliday R, Borges LM, Stearns-Yoder KA, Hoffberg AS, Brenner LA, Monteith LL. Posttraumatic stress disorder, suicidal ideation, and suicidal self-directed violence among U.S. military personnel and veterans: a systematic review of the literature from 2010 to 2018. Front Psychol. (2020) 11:1998. doi: 10.3389/fpsyg.2020.01998

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Fox V, Dalman C, Dal H, Hollander AC, Kirkbride JB, Pitman A. Suicide risk in people with post-traumatic stress disorder: a cohort study of 3.1 million people in Sweden. J Affect Disord. (2021) 279:609–16. doi: 10.1016/j.jad.2020.10.009

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Torrisi SA, Leggio GM, Drago F, Salomone S. Therapeutic challenges of post-traumatic stress disorder: focus on the dopaminergic system. Front Pharmacol. (2019) 10:404. doi: 10.3389/fphar.2019.00404

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Kataoka T, Fuchikami M, Nojima S, Nagashima N, Araki M, Omura J, et al. Combined brain-derived neurotrophic factor with extinction training alleviate impaired fear extinction in an animal model of post-traumatic stress disorder. Genes Brain Behav. (2019) 18:e12520. doi: 10.1111/gbb.12520

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Boyd JE, Lanius RA, McKinnon MC. Mindfulness-based treatments for posttraumatic stress disorder: a review of the treatment literature and neurobiological evidence. J Psychiatry Neurosci. (2018) 43:7–25. doi: 10.1503/jpn.170021

PubMed Abstract | CrossRef Full Text | Google Scholar

40. 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

PubMed Abstract | CrossRef Full Text | Google Scholar

41. The Management of Posttraumatic Stress Disorder Work Group (2017). VA/DOD Clinical Practice Guideline for the Management of Posttraumatic Stress Disorder and Acute Stress Disorder. Department of Veterans Affairs. Available online at: https://www.healthquality.va.gov/guidelines/mh/ptsd/index.asp

Google Scholar

42. Friedman MJ. Finalizing PTSD in DSM-5: getting here from there and where to go next. J Trauma Stress. (2013) 26:548–56. doi: 10.1002/jts.21840

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Ehret M. Treatment of posttraumatic stress disorder: focus on pharmacotherapy. Ment Health Clin. (2019) 9:373–82. doi: 10.9740/mhc.2019.11.373

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Akiki TJ, Abdallah CG. Are there effective psychopharmacologic treatments for PTSD? J Clin Psychiatry. (2018) 80:18ac12473. doi: 10.4088/JCP.18ac12473

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Hoskins MD, Bridges J, Sinnerton R, Nakamura A, Underwood J, Slater A, et al. Pharmacological therapy for post-traumatic stress disorder: a systematic review and meta-analysis of monotherapy, augmentation and head-to-head approaches. Eur J Psychotraumatol. (2021) 12:1802920. doi: 10.1080/20008198.2020.1802920

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Huang ZD, Zhao YF, Li S, Gu HY, Lin LL, Yang ZY, et al. Comparative efficacy and acceptability of pharmaceutical management for adults with post-traumatic stress disorder: a systematic review and meta-analysis. Front Pharmacol. (2020) 11:559. doi: 10.3389/fphar.2020.00559

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Sotille R, Singh H, Weisman A, Vida T. Unraveling the mysteries of mental illness with psilocybin. Cureus. (2022) 14:e25414. doi: 10.7759/cureus.25414

PubMed Abstract | CrossRef Full Text | Google Scholar

48. 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. J Psychoactive Drugs. (2021) 53:85–95. doi: 10.1080/02791072.2020.1817639

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Kline AC, Cooper AA, Rytwinksi NK, Feeny NC. Long-term efficacy of psychotherapy for posttraumatic stress disorder: a meta-analysis of randomized controlled trials. Clin Psychol Rev. (2018) 59:30–40. doi: 10.1016/j.cpr.2017.10.009

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Lewis C, Roberts NP, Andrew M, Starling E, Bisson JI. Psychological therapies for post-traumatic stress disorder in adults: systematic review and meta-analysis. Eur J Psychotraumatol. (2020) 11:1729633. doi: 10.1080/20008198.2020.1729633

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Simmons C, Meiser-Stedman R, Baily H, Beazley P. A meta-analysis of dropout from evidence-based psychological treatment for post-traumatic stress disorder (PTSD) in children and young people. Eur J Psychotraumatol. (2021) 12:1947570. doi: 10.1080/20008198.2021.1947570

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Chen JA, Fortney JC, Bergman HE, Browne KC, Grubbs KM, Hudson TJ, et al. Therapeutic alliance across trauma-focused and non-trauma-focused psychotherapies among veterans with PTSD. Psychol Serv. (2020) 17:452–60. doi: 10.1037/ser0000329

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Fuentes JJ, Fonseca F, Elices M, Farré M, Torrens M. Therapeutic use of LSD in psychiatry: a systematic review of randomized-controlled clinical trials. Front Psychiatry. (2020) 10:943. doi: 10.3389/fpsyt.2019.00943

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Henner RL, Keshavan MS, Hill KP. Review of potential psychedelic treatments for PTSD. J Neurol Sci. (2022) 439:120302. doi: 10.1016/j.jns.2022.120302

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Schenberg EE. Psychedelic-assisted psychotherapy: a paradigm shift in psychiatric research and development. Front Pharmacol. (2018) 9:733. doi: 10.3389/fphar.2018.00733

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Hoskins MD, Sinnerton R, Nakamura A, Underwood J, Slater A, Lewis C, et al. Pharmacological-assisted psychotherapy for post-traumatic stress disorder: a systematic review and meta-analysis. Eur J Psychotraumatol. (2021) 12:1853379. doi: 10.1080/20008198.2020.1853379

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Wagner MT, Mithoefer MC, Mithoefer AT, MacAulay RK, Jerome L, Yazar-Klosinski B, et al. Therapeutic effect of increased openness: investigating mechanism of action in MDMA-assisted psychotherapy. J Psychopharmacol. (2017) 31:967–74. doi: 10.1177/0269881117711712

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Nutt D. Psychedelic drugs-a new era in psychiatry? Dialogues Clin Neurosci. (2019) 21:139–47. doi: 10.31887/DCNS.2019.21.2/dnutt

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Penn A, Dorsen CG, Hope S, Rosa WE. Psychedelic-assisted therapy: emerging treatments in mental health disorders. Am J Nurs. (2021) 121:34–40. doi: 10.1097/01.NAJ.0000753464.35523.29

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Mithoefer MC, Feduccia AA, Jerome L, Mithoefer A, Wagner M, Walsh Z, 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) 236:2735–45. doi: 10.1007/s00213-019-05249-5

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Tupper KW, Wood E, Yensen R, Johnson MW. Psychedelic medicine: a re-emerging therapeutic paradigm. CMAJ. (2015) 187:1054–9. doi: 10.1503/cmaj.141124

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Ponte L, Jerome L, Hamilton S, Mithoefer MC, Yazar-Klosinski BB, Vermetten E, et al. Sleep quality improvements after MDMA-assisted psychotherapy for the treatment of posttraumatic stress disorder. J Trauma Stress. (2021) 34:851–63. doi: 10.1002/jts.22696

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Dunlap LE, Andrews AM, Olson DE. Dark classics in chemical neuroscience: 3,4-Methylenedioxymethamphetamine. ACS Chem Neurosci. (2018) 9:2408–27. doi: 10.1021/acschemneuro.8b00155

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Puerta E, Aguirre N. Methylenedioxymethamphetamine (MDMA, ‘Ecstasy'): neurodegeneration versus neuromodulation. Pharmaceuticals. (2011) 4:992–1018. doi: 10.3390/ph4070992

CrossRef Full Text | Google Scholar

65. National Center for Biotechnology Information (2022). PubChem Compound Summary for CID 1615, 3,4-Methylenedioxymethamphetamine. Available online at: https://pubchem.ncbi.nlm.nih.gov/compound/3_4-Methylenedioxymethamphetamine (accessed June 29, 2022).

Google Scholar

66. Benzenhofer U, Passie T. Rediscovering MDMA (ecstasy): the role of the American chemist Alexander T. Shulgin. Addiction. (2010) 105:1355–61. doi: 10.1111/j.1360-0443.2010.02948.x

PubMed Abstract | CrossRef Full Text | Google Scholar

67. de la Torre R, Farré M, Roset PN, Pizarro N, Abanades S, Segura M, et al. Human pharmacology of MDMA. Ther Drug Monit. (2004) 26:137–44. doi: 10.1097/00007691-200404000-00009

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Lyon RA, Glennon RA, Titeler M. 3,4-Methylenedioxymethamphetamine (MDMA): stereoselective interactions at brain 5-HT1 and 5-HT2 receptors. Psychopharmacology. (1986) 88:525–6. doi: 10.1007/BF00178519

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Mustafa NS, Bakar N, Mohamad N, Adnan L, Fauzi N, Thoarlim A, et al. MDMA and the brain: a short review on the role of neurotransmitters in neurotoxicity. Basic Clin Neurosci. (2020) 11:381–8. doi: 10.32598/bcn.9.10.485

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Kamilar-Britt P, Bedi G. The prosocial effects of 3,4-methylenedioxymethamphetamine (MDMA): controlled studies in humans and laboratory animals. Neurosci Biobehav Rev. (2015) 57:433–46. doi: 10.1016/j.neubiorev.2015.08.016

PubMed Abstract | CrossRef Full Text | Google Scholar

71. 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

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Wardle MC, Kirkpatrick MG, de Wit H. 'Ecstasy' as a social drug: MDMA preferentially affects responses to emotional stimuli with social content. Soc Cogn Affect Neurosci. (2014) 9:1076–81. doi: 10.1093/scan/nsu035

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Vegting Y, Reneman L, Booij J. The effects of ecstasy on neurotransmitter systems: a review on the findings of molecular imaging studies. Psychopharmacology. (2016) 233:3473–501. doi: 10.1007/s00213-016-4396-5

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Levy KB, O'Grady KE, Wish ED, Arria AM. An in-depth qualitative examination of the ecstasy experience: results of a focus group with ecstasy-using college students. Subst Use Misuse. (2005) 40:1427–41. doi: 10.1081/JA-200066810

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Costa G, Golembiowska K. Neurotoxicity of MDMA: main effects and mechanisms. Exp Psychol. (2022) 347:113894. doi: 10.1016/j.expneurol.2021.113894

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Steinkellner T, Freissmuth M, Sitte HH, Montgomery T. The ugly side of amphetamines: short- and long-term toxicity of 3,4-methylenedioxymethamphetamine (MDMA, 'Ecstasy'), methamphetamine and D-amphetamine. Biol Chem. (2011) 392:103–15. doi: 10.1515/bc.2011.016

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Mitchell JM, Bogenschutz M, Lilienstein A, Harrison C, Kleiman S, Parker-Guilbert K, et al. MDMA-assisted therapy for severe PTSD: a randomized, double-blind, placebo-controlled phase 3 study. Nat Med. (2021) 27:1025–33. doi: 10.1038/s41591-021-01336-3

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Pantoni MM, Kim JL, Van Alstyne KR, Anagnostaras SG. MDMA and memory, addiction, and depression: dose-effect analysis. Psychopharmacology. (2022) 239:935–49. doi: 10.1007/s00213-022-06086-9

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Verrico CD, Miller GM, Madras BK. MDMA (Ecstasy) and human dopamine, norepinephrine, and serotonin transporters: implications for MDMA-induced neurotoxicity and treatment. Psychopharmacology. (2007) 189:489–503. doi: 10.1007/s00213-005-0174-5

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Docherty JR, Alsufyani HA. Pharmacology of drugs used as stimulants. J Clin Pharmacol. (2021) 61:S53–69. doi: 10.1002/jcph.1918

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Heifets BD, Salgado JS, Taylor MD, Hoerbelt P, Cardozo Pinto DF, Steinberg EE, et al. Distinct neural mechanisms for the prosocial and rewarding properties of MDMA. Sci Transl Med. (2019) 11:eaaw6435. doi: 10.1126/scitranslmed.aaw6435

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Sandtner W, Stockner T, Hasenhuetl PS, Partilla JS, Seddik A, Zhang YW, et al. Binding mode selection determines the action of ecstasy homologs at monoamine transporters. Mol Pharmacol. (2016) 89:165–75. doi: 10.1124/mol.115.101394

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Oeri HE. Beyond ecstasy: alternative entactogens to 3,4-methylenedioxymethamphetamine with potential applications in psychotherapy. J Psychopharmacol. (2021) 35:512–36. doi: 10.1177/0269881120920420

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Dipasquale O, Selvaggi P, Veronese M, Gabay AS, Turkheimer F, Mehta MA. Receptor-Enriched Analysis of functional connectivity by targets (REACT): a novel, multimodal analytical approach informed by PET to study the pharmacodynamic response of the brain under MDMA. Neuroimage. (2019) 195:252–60. doi: 10.1016/j.neuroimage.2019.04.007

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Vidal-Infer A, Roger-Sánchez C, Daza-Losada M, Aguilar MA, Miñarro J, Rodríguez-Arias M. Role of the dopaminergic system in the acquisition, expression and reinstatement of MDMA-induced conditioned place preference in adolescent mice. PLoS ONE. (2012) 7:e43107. doi: 10.1371/journal.pone.0043107

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Mujawar S, Patil J, Chaudhari B, Saldanha D. (2021). Memory: Neurobiological mechanisms and assessment. Ind. Psychiatry J. 30:S311–S314. doi: 10.4103/0972-6748.328839

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Tyng CM, Amin HU, Saad M, Malik AS. The influences of emotion on learning and memory. Front Psychol. (2017) 8:1454. doi: 10.3389/fpsyg.2017.01454

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Zarrindast MR, Khakpai F. State-dependent memory and its modulation by different brain areas and neurotransmitters. EXCLI J. (2020) 19:1081–99. doi: 10.17179/excli2020-2612

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Engen HG, Anderson MC. Memory control: a fundamental mechanism of emotion regulation. Trends Cogn Sci. (2018) 22:982–95. doi: 10.1016/j.tics.2018.07.015

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Luchkina NV, Bolshakov VY. Mechanisms of fear learning and extinction: synaptic plasticity-fear memory connection. Psychopharmacology. (2019) 236:163–82. doi: 10.1007/s00213-018-5104-4

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Sun Y, Gooch H, Sah P. Fear conditioning and the basolateral amygdala. F1000Res. (2020) 9:F1000 Faculty Rev−53. doi: 10.12688/f1000research.21201.1

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Goode TD, Holloway-Erickson CM, Maren S. Extinction after fear memory reactivation fails to eliminate renewal in rats. Neurobiol Learn Mem. (2017) 142:41–7. doi: 10.1016/j.nlm.2017.03.001

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Harnett NG, Goodman AM, Knight DC. PTSD-related neuroimaging abnormalities in brain function, structure, and biochemistry. Exp Neurol. (2020) 330:113331. doi: 10.1016/j.expneurol.2020.113331

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Marek R, Sah P. Neural circuits mediating fear learning and extinction. Syst Neurosci. (2018) 21:35–48. doi: 10.1007/978-3-319-94593-4_2

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Nikolenko VN, Oganesyan MV, Rizaeva NA, Kudryashova VA, Nikitina AT, Pavliv MP, et al. Amygdala: neuroanatomical and morphophysiological features in terms of neurological and neurodegenerative diseases. Brain Sci. (2020) 10:502. doi: 10.3390/brainsci10080502

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Carlen M. What constitutes the prefrontal cortex? Science. (2017) 358:478–82. doi: 10.1126/science.aan8868

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Wible CG. Hippocampal physiology, structure and function and the neuroscience of schizophrenia: a unified account of declarative memory deficits, working memory deficits and schizophrenic symptoms. Behav Sci. (2013) 3:298–315. doi: 10.3390/bs3020298

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Liao Z, Tao Y, Guo X, Cheng D, Wang F, Liu X, et al. Fear conditioning downregulates Rac1 activity in the basolateral amygdala astrocytes to facilitate the formation of fear memory. Front Mol Neurosci. (2017) 10:396. doi: 10.3389/fnmol.2017.00396

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Maren S. Out with the old and in with the new: synaptic mechanisms of extinction in the amygdala. Brain Res. (2015) 1621:231–8. doi: 10.1016/j.brainres.2014.10.010

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Meis S, Endres T, Lessmann V. Neurotrophin signalling in amygdala-dependent cued fear learning. Cell Tissue Res. (2020) 382:161–72. doi: 10.1007/s00441-020-03260-3

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Stansley BJ, Fisher NM, Gogliotti RG, Lindsley CW, Conn PJ, Niswender CM. Contextual fear extinction induces hippocampal metaplasticity mediated by metabotropic glutamate receptor 5. Cereb Cortex. (2018) 28:4291–304. doi: 10.1093/cercor/bhx282

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Porter JT, Sepulveda-Orengo MT. Learning-induced intrinsic and synaptic plasticity in the rodent medial prefrontal cortex. Neurobiol Learn Mem. (2020) 169:107117. doi: 10.1016/j.nlm.2019.107117

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Do Monte FH, Quirk GJ, Li B, Penzo MA. Retrieving fear memories, as time goes by…. Mol Psychiatry. (2016) 21:1027–36. doi: 10.1038/mp.2016.78

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Cahill EN, Milton AL. Neurochemical and molecular mechanisms underlying the retrieval-extinction effect. Psychopharmacology. (2019) 236:111–32. doi: 10.1007/s00213-018-5121-3

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Salinas-Hernández XI, Duvarci S. Dopamine in fear extinction. Front Synaptic Neurosci. (2021) 13:635879. doi: 10.3389/fnsyn.2021.635879

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Auchter AM, Shumake J, Gonzalez-Lima F, Monfils MH. Preventing the return of fear using reconsolidation updating and methylene blue is differentially dependent on extinction learning. Sci Rep. (2017) 7:46071. doi: 10.1038/srep46071

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Young MB, Norrholm SD, Khoury LM, Jovanovic T, Rauch SA, Reiff CM, et al. Inhibition of serotonin transporters disrupts the enhancement of fear memory extinction by 3,4-methylenedioxymethamphetamine (MDMA). Psychopharmacology. (2017) 234:2883–95. doi: 10.1007/s00213-017-4684-8

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Kida S. Function and mechanisms of memory destabilization and reconsolidation after retrieval. Proc Jpn Acad Ser B Phys Biol Sci. (2020) 96:95–106. doi: 10.2183/pjab.96.008

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Kida S. Reconsolidation/destabilization, extinction and forgetting of fear memory as therapeutic targets for PTSD. Psychopharmacology. (2019) 236:49–57. doi: 10.1007/s00213-018-5086-2

PubMed Abstract | CrossRef Full Text | Google Scholar

110. An B, Kim J, Park K, Lee S, Song S, Choi S. Amount of fear extinction changes its underlying mechanisms. Elife. (2017) 6:e25224. doi: 10.7554/eLife.25224.018

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Do-Monte FH, Manzano-Nieves G, Quiñones-Laracuente K, Ramos-Medina L, Quirk GJ. Revisiting the role of infralimbic cortex in fear extinction with optogenetics. J Neurosci. (2015) 35:3607–15. doi: 10.1523/JNEUROSCI.3137-14.2015

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Aliev G, Beeraka NM, Nikolenko VN, Svistunov AA, Rozhnova T, Kostyuk S, et al. Neurophysiology and psychopathology underlying PTSD and recent insights into the PTSD therapies—A comprehensive review. J Clin Med. (2020) 9:2951. doi: 10.3390/jcm9092951

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Krystal JH, Abdallah CG, Averill LA, Kelmendi B, Harpaz-Rotem I, Sanacora G, et al. Synaptic loss and the pathophysiology of PTSD: implications for ketamine as a prototype novel therapeutic. Curr Psychiatry Rep. (2017) 19:74. doi: 10.1007/s11920-017-0829-z

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Garfinkel SN, Abelson JL, King AP, Sripada RK, Wang X, Gaines LM, et al. Impaired contextual modulation of memories in PTSD: an fMRI and psychophysiological study of extinction retention and fear renewal. J Neurosci. (2014) 34:13435–43. doi: 10.1523/JNEUROSCI.4287-13.2014

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Milad MR, Pitman RK, Ellis CB, Gold AL, Shin LM, Lasko NB, et al. Neurobiological basis of failure to recall extinction memory in posttraumatic stress disorder. Biol Psychiatry. (2009) 66:1075–82. doi: 10.1016/j.biopsych.2009.06.026

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Maren S, Holmes A. Stress and fear extinction. Neuropsychopharmacology. (2016) 41:58–79. doi: 10.1038/npp.2015.180

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Rousseau PF, El Khoury-Malhame M, Reynaud E, Boukezzi S, Cancel A, Zendjidjian X, et al. Fear extinction learning improvement in PTSD after EMDR therapy: an fMRI study. Eur J Psychotraumatol. (2019) 10:1568132. doi: 10.1080/20008198.2019.1568132

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Zuj DV, Palmer MA, Lommen MJJ, Felmingham KL. The centrality of fear extinction in linking risk factors to PTSD: a narrative review. Neurosci Biobehav Rev. (2016) 69:15–35. doi: 10.1016/j.neubiorev.2016.07.014

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Zhang HH, Meng SQ, Guo XY, Zhang JL, Zhang W, Chen YY, et al. Traumatic stress produces delayed alterations of synaptic plasticity in basolateral amygdala. Front Psychol. (2019) 10:2394. doi: 10.3389/fpsyg.2019.02394

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Stevens JS, Kim YJ, Galatzer-Levy IR, Reddy R, Ely TD, Nemeroff CB, et al. Amygdala reactivity and anterior cingulate habituation predict posttraumatic stress disorder symptom maintenance after acute civilian trauma. Biol Psychiatry. (2017) 81:1023–9. doi: 10.1016/j.biopsych.2016.11.015

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Morey RA, Gold AL, LaBar KS, Beall SK, Brown VM, Haswell CC, et al. Amygdala volume changes in posttraumatic stress disorder in a large case-controlled veterans group. Arch Gen Psychiatry. (2012) 69:1169–78. doi: 10.1001/archgenpsychiatry.2012.50

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Belleau EL, Ehret LE, Hanson JL, Brasel KJ, Larson CL, deRoon-Cassini TA. Amygdala functional connectivity in the acute aftermath of trauma prospectively predicts severity of posttraumatic stress symptoms. Neurobiol Stress. (2020) 12:100217. doi: 10.1016/j.ynstr.2020.100217

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Koenigs M, Grafman J. Posttraumatic stress disorder: the role of medial prefrontal cortex and amygdala. Neuroscientist. (2009) 15:540–8. doi: 10.1177/1073858409333072

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Bisby JA, Burgess N, Brewin CR. Reduced memory coherence for negative events and its relationship to posttraumatic stress disorder. Curr Dir Psychol Sci. (2020) 29:267–72. doi: 10.1177/0963721420917691

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Ousdal OT, Milde AM, Hafstad GS, Hodneland E, Dyb G, Craven AR, et al. The association of PTSD symptom severity with amygdala nuclei volumes in traumatized youths. Transl Psychiatry. (2020) 10:288. doi: 10.1038/s41398-020-00974-4

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Abdallah CG, Averill LA, Akiki TJ, Raza M, Averill CL, Gomaa H, et al. The neurobiology and pharmacotherapy of posttraumatic stress disorder. Annu Rev Pharmacol Toxicol. (2019) 59:171–89. doi: 10.1146/annurev-pharmtox-010818-021701

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Erjavec GN, Nikolac Perkovic M, Tudor L, Uzun S, Kovacic Petrovic Z, Konjevod M, et al. Moderating effects of BDNF genetic variants and smoking on cognition in PTSD veterans. Biomolecules. (2021) 11:641. doi: 10.3390/biom11050641

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Green CR, Corsi-Travali S, Neumeister A. The role of BDNF-TrkB signaling in the pathogenesis of PTSD. J Depress Anxiety. (2013) 2013:006. doi: 10.4172/2167-1044.S4-006

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Miller JK, McDougall S, Thomas S, Wiener J. The impact of the brain-derived neurotrophic factor gene on trauma and spatial processing. J Clin Med. (2017) 6:108. doi: 10.3390/jcm6120108

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Mojtabavi H, Saghazadeh A, van den Heuvel L, Bucker J, Rezaei N. Peripheral blood levels of brain-derived neurotrophic factor in patients with post-traumatic stress disorder (PTSD): a systematic review and meta-analysis. PLoS ONE. (2020) 15:e0241928. doi: 10.1371/journal.pone.0241928

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Zhang L, Li XX, Hu XZ. Post-traumatic stress disorder risk and brain-derived neurotrophic factor Val66Met. World J Psychiatry. (2016) 6:1–6. doi: 10.5498/wjp.v6.i1.1

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Miranda M, Morici JF, Zanoni MB, Bekinschtein P. Brain-derived neurotrophic factor: a key molecule for memory in the healthy and the pathological brain. Front Cell Neurosci. (2019) 13:363. doi: 10.3389/fncel.2019.00363

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Yang T, Nie Z, Shu H, Kuang Y, Chen X, Cheng J, et al. The role of BDNF on neural plasticity in depression. Front Cell Neurosci. (2020) 14:82. doi: 10.3389/fncel.2020.00082

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Ji LL, Peng JB, Fu CH, Cao D, Li D, Tong L, et al. Activation of Sigma-1 receptor ameliorates anxiety-like behavior and cognitive impairments in a rat model of post-traumatic stress disorder. Behav Brain Res. (2016) 311:408–15. doi: 10.1016/j.bbr.2016.05.056

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Zhao M, Wang W, Jiang Z, Zhu Z, Liu D, Pan F. Long-term effect of post-traumatic stress in adolescence on dendrite development and H3K9me2/BDNF expression in male rat hippocampus and prefrontal cortex. Front Cell Dev Biol. (2020) 8:682. doi: 10.3389/fcell.2020.00682

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Smith KW, Sicignano DJ, Hernandez AV, White CM. MDMA-assisted psychotherapy for treatment of posttraumatic stress disorder: a systematic review with meta-analysis. J Clin Pharmacol. (2022) 62:463–71. doi: 10.1002/jcph.1995

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Vermetten E, Yehuda R. MDMA-assisted psychotherapy for posttraumatic stress disorder: a promising novel approach to treatment. Neuropsychopharmacology. (2020) 45:231–2. doi: 10.1038/s41386-019-0482-9

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Hysek CM, Schmid Y, Simmler LD, Domes G, Heinrichs M, Eisenegger C, et al. MDMA enhances emotional empathy and prosocial behavior. Soc Cogn Affect Neurosci. (2014) 9:1645–52. doi: 10.1093/scan/nst161

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Thal SB, Lommen M. Current perspective on MDMA-assisted psychotherapy for posttraumatic stress disorder. J Contemp Psychother. (2018) 48:99–108. doi: 10.1007/s10879-017-9379-2

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Inserra A, De Gregorio D, Gobbi G. Psychedelics in psychiatry: neuroplastic, immunomodulatory, neurotransmitter mechanisms. Pharmacol Rev. (2021) 73:202–77. doi: 10.1124/pharmrev.120.000056

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Lukasiewicz K, Baker JJ, Zuo Y, Lu J. Serotonergic psychedelics in neural plasticity. Front Mol Neurosci. (2021) 14:748359. doi: 10.3389/fnmol.2021.748359

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Tedesco S, Gajaram G, Chida S, Ahmad A, Pentak M, Kelada M, et al. The efficacy of MDMA (3,4-Methylenedioxymethamphetamine) for post-traumatic stress disorder in humans: a systematic review and meta-analysis. Cureus. (2021) 13:e15070. doi: 10.7759/cureus.15070

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Abad S, Fole A, del Olmo N, Pubill D, Pallàs M, Junyent F, et al. MDMA enhances hippocampal-dependent learning and memory under restrictive conditions, and modifies hippocampal spine density. Psychopharmacology. (2013) 231:863–74. doi: 10.1007/s00213-013-3304-5

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Young MB, Andero R, Ressler KJ, Howell LL. 3, 4-Methylenedioxymethamphetamine facilitates fear extinction learning. Transl Psychiatry. (2015) 5:e634. doi: 10.1038/tp.2015.138

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Ly C, Greb AC, Cameron LP, Wong JM, Barragan EV, Wilson PC, et al. Psychedelics promote structural and functional neural plasticity. Cell Rep. (2018) 23:3170–82. doi: 10.1016/j.celrep.2018.05.022

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Olson DE. Psychoplastogens: A Promising Class of Plasticity-Promoting Neurotherapeutics. J. Exp. Neurosci. (2018) 12:1179069518800508. doi: 10.1177/1179069518800508

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Hake HS, Davis J, Wood RR, Tanner MK, Loetz EC, Sanchez A, 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

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Yazar-Klosinski BB, Mithoefer MC. Potential psychiatric uses for MDMA. Clin Pharmacol Ther. (2017) 101:194–6. doi: 10.1002/cpt.565

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Martínez-Turrillas R, Moyano S, Del Río J, Frechilla D. Differential effects of 3,4-methylenedioxymethamphetamine (MDMA, “ecstasy”) on BDNF mRNA expression in rat frontal cortex and hippocampus. Neurosci Lett. (2006) 402:126–30. doi: 10.1016/j.neulet.2006.03.055

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Inserra A. Current status of psychedelic therapy in Australia and New Zealand: Are we falling behind? Aust N J Psychiatry. (2019) 53:190–2. doi: 10.1177/0004867418824018

PubMed Abstract | CrossRef Full Text | Google Scholar

151. Sessa. Could MDMA be useful in the treatment of post-traumatic stress disorder? Prog Neurol Psychiatr. (2011) 15:4–7. doi: 10.1002/pnp.216

CrossRef Full Text | Google Scholar

Keywords: MDMA, PTSD, psychedelic-assisted psychotherapy, fear extinction, fear anxiety and relief

Citation: Sottile RJ and Vida T (2022) A proposed mechanism for the MDMA-mediated extinction of traumatic memories in PTSD patients treated with MDMA-assisted therapy. Front. Psychiatry 13:991753. doi: 10.3389/fpsyt.2022.991753

Received: 11 July 2022; Accepted: 29 August 2022;
Published: 12 October 2022.

Edited by:

Felix Mayer, Florida Atlantic University, United States

Reviewed by:

Thomas Steinkellner, Medical University of Vienna, Austria
Boris Heifets, Stanford University, United States

Copyright © 2022 Sottile and Vida. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Robert J. Sottile, sottile@unlv.nevada.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.