<<

Review

Cite This: ACS Chem. Neurosci. 2018, 9, 2408−2427 pubs.acs.org/chemneuro

Dark Classics in Chemical Neuroscience: 3,4- Methylenedioxymethamphetamine Lee E. Dunlap,† Anne M. Andrews,‡ and David E. Olson*,†,§,∥

† Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, California 95616, United States ‡ Departments of Psychiatry and Chemistry & Biochemistry, Semel Institute for Neuroscience and Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, California 90095, United States § Department of Biochemistry & Molecular Medicine, School of Medicine, University of California, Davis, 2700 Stockton Blvd, Suite 2102, Sacramento, California 95817, United States ∥ Center for Neuroscience, University of California, Davis, 1544 Newton Ct, Davis, California 95616, United States

ABSTRACT: Better known as “ecstasy”, 3,4-methylenedioxy- (MDMA) is a small molecule that has played a prominent role in defining the ethos of today’s teenagers and young adults, much like lysergic acid diethyl- amide (LSD) did in the 1960s. Though MDMA possesses structural similarities to compounds like and , it produces subjective effects that are unlike any of the classical psychostimulants or hallucinogens and is one of the few compounds capable of reliably producing prosocial behavioral states. As a result, MDMA has captured the attention of recreational users, the media, artists, psychiatrists, and neuropharmacologists alike. Here, we detail the synthesis of MDMA as well as its pharmacology, metabolism, adverse effects, and potential use in medicine. Finally, we discuss its history and why it is perhaps the most important compound for the future of psychedelic sciencehaving the potential to either facilitate new psychedelic research initiatives, or to usher in a second Dark Age for the field. KEYWORDS: 3,4-methylenedioxymethamphetamine, MDMA, psychedelic, entactogen, empathogen

■ INTRODUCTION unable to substitute for the hallucinogen 2,5- 7 The psychoactive compound 3,4-methylenedioxymeth- dimethoxy-4-methylamphetamine (DOM). Furthermore, when rats are trained to discriminate racemic MDMA from amphetamine (MDMA, 1) is better known by one of its “ ” saline, incomplete generalization is observed using S- numerous street names, which include ecstasy , XTC, E, X, 5 1 (+)-amphetamine, LSD, or DOM. The discriminative MDM, Adam, and EA-1475. Additionally, the term “molly” is stimulus produced by MDMA seems to be modulated by 5- often used to refer to MDMA in the United States.2 HT1A,8 5-HT2A,8 and receptors,9 with less Downloaded via OPEN UNIV OF ISRAEL on November 22, 2020 at 12:56:56 (UTC). Structurally, MDMA possesses a single stereocenter, and due involvement from D1 receptors.10 The two enantiomers of to its small size (freebase MW = 193.24 g/mol) and 11 3 MDMA produce relatively similar discriminative stimuli. It See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles. hydrophobic nature (logP = 2.050), MDMA readily crosses should be noted that while MDMA only partially substitutes the blood−brain barrier (BBB).4 Chemically, MDMA is related for S-(+)-amphetamine in rats, it completely substitutes for S- 2 12 to amphetamine ( ), and contains the phenethylamine core (+)-amphetamine in rhesus monkeys. structure common to this class of psychostimulants, which also In humans, 75−150 mg of MDMA produces subjective − includes methamphetamine (3), and (4) effects that last for several hours.13 19 These include context- (Figure 1). The hallucinogens 2,5-dimethoxy-4-iodoamphet- dependent feelings of closeness with others, reduced social − amine (DOI, 5), 2,5-dimethoxy-4-bromophenethylamine (- inhibition, positive mood, and increased alertness.19 24 B, 6), and mescaline (7) are also structurally related to Regarding hallucinations, MDMA is considered to be weakly MDMA. As such, it is not surprising that MDMA produces hallucinogenic.25 Ingestion of MDMA does not cause auditory effects reminiscent of both psychostimulants and hallucino- ff hallucinations and only 20% of recreational users have reported gens. However, the interoceptive e ects of MDMA (i.e., sense experiencing visual hallucinations.15 The visual hallucinations of the body’s internal state) are distinct from those produced by either of these well-known classes of psychoactive compounds. Special Issue: DARK Classics in Chemical Neuroscience In rodent drug discrimination studies, MDMA only partially Received: April 4, 2018 substitutes for the S-(+)-amphetamine,5 or the Accepted: June 25, 2018 hallucinogen lysergic acid diethylamide (LSD),6 and is Published: July 12, 2018

© 2018 American Chemical Society 2408 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

Figure 1. Structural relationships between MDMA and other psychoactive compounds. The common phenethylamine core is highlighted in red. Compounds are classified as psychostimulants, hallucinogens, or entactogens based on the behavioral responses they produce in experimental animals and their subjective effects in humans. induced by MDMA do not tend to be well-formed, and defined can be subdivided into classical hallucinogens (e.g., instead, are often described as flashes of light in the peripheral , LSD, mescaline) and entactogens (e.g., MDMA) on visual field.15 This is in stark contrast to the profound visual the basis of their distinct subjective effects. disturbances experienced by most people following the The prosocial and stimulant effects of MDMA led to its administration of classical hallucinogenic agents such as widespread recreational use and cemented its place in rave LSD.26,27 In humans, the weak hallucinogenic effects of (dance party) culture.44 It is estimated that MDMA has been MDMA are blocked by , a selective 5-HT2A used by 7% of the population over the age of 12.45 This is in 25 antagonist. The role of 5-HT1A receptors in the subjective stark contrast to heroin, which is abused by only 2% of the 28−30 effects of MDMA appears to be negligible. In addition to population.45 The predominant users of MDMA are teenagers directly binding to 5-HT2A receptors, albeit with low affinity and young adults, with females being more likely to use ff 46 (vide infra), MDMA can produce subjective e ects by MDMA than males. In people 12−25 years of age, MDMA increasing the release of monoamines such as and 45 − accounts for more than 50% of all psychedelic drug use. In .17,31 35 The perceptual effects of MDMA are 36,37 recent years, the recreational use of MDMA by people with more intense in females than in males, and have been college degrees has been increasing.47 shown in recent placebo-controlled studies to be clearly 38,39 Despite its popularity, MDMA is a controlled substance in distinct from those produced by other psychostimulants. the United States and many other countries making its The unique subjective effects of MDMA and related α production and sale illegal. The U.S. Drug Enforcement molecules such as 3,4-methylenedioxy-N-methyl- -ethylphe- Administration (DEA) has classified MDMA as a Schedule I nylethylamine (MBDB, 8) and 5,6-methylenedioxy-2-amino-  fi compound the most restricted class of chemicals. Schedule I indane (MDAI, 9) led to their classi cation as a separate family drugs are those deemed to have high abuse potential, do not of psychoactive compounds, distinct from both and have an accepted medical use, and lack accepted safety for use hallucinogens (Figure 1). Due to their strong propensity to under medical supervision. Drugs such as heroin, LSD, γ- induce empathy and feelings of connectedness, these drugs hydroxybutyric acid (GHB), and tetrahydrocannabinol (THC) were originally dubbed “empathogens” in the 1980sa term are also classified in Schedule I. Unfortunately, the legal, favored by Ralph Metzner.40 David Nichols later highlighted financial, and political hurdles that accompany Schedule I the ambiguous nature of the term empathogen. To avoid any “ ” ff classification significantly hinder scientific research into the negative connotations associated with pathos (i.e., su ering), ff “pathogen” (i.e., a disease producing agent), or “pathogenesis” e ects of MDMA. As it is one of the few compounds known to (i.e., the development of a disease), Nichols coined the new reliably produce a prosocial state, MDMA may possess term “entactogen,” which roughly translates from the Greek to potential as a neurochemical tool for elucidating the “ ” mechanisms of social behaviors and the neural underpinnings mean that which produces a touching within (en = within, 48 tactus = touch, gen = to produce).41 These terms are often of empathy and social bonding. Furthermore, MDMA may used interchangeably. We will use the latter term as it more possess therapeutic potential for treating disorders associated adequately captures the unique ability of these drugs to with disruptions in social interactions such as autism spectrum  disorders, social anxiety disorder, , and post- promote introspective states a property that has been 48 proposed to be useful in the context of psychotherapy (vide traumatic stress disorder (PTSD). infra). Though the subjective effects of MDMA appear to be Despite its relatively simple structure, MDMA elicits robust unique compared to those of LSD, both compounds tend to behavioral responses by binding with high affinity to a number increase openness, promote trust, and enhance emotional of neuroreceptors and transporters. Below, we discuss the empathy.42,43 synthesis of MDMA and its pharmacology, metabolism, and A major point of contention among psychopharmacologists adverse effects. Additionally, we review the prosocial and iswhetherornotMDMAshouldbeclassified as a psychoplastogenic (plasticity-promoting) properties of “psychedelic.” Because that term can be translated as “mind- MDMA, the differences between its enantiomers, and its manifesting,” we propose that MDMA, as well as more potent potential use in medicine. Finally, we provide brief historical 5-HT2A like psilocybin, are appropriately placed in context for the development of MDMA and conclude by this category. Using this classification, psychedelics broadly emphasizing the important role that MDMA is expected to

2409 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review play in determining the trajectory of future psychedelic literature for the first time by Polish chemists Biniecki and research. Krajewski.51 Alternatively, MDMA can be synthesized from 10 by Wacker oxidation followed by reductive amination of 12 ■ SYNTHESIS with methylamine and cyanoborohydride.52,53 Com- Racemic MDMAthe form used recreationally and in clinical pound 12 can also be accessed from 10, following olefin trialsis typically synthesized from safrole (10) or piperonal isomerization to produce isosafrole, peracid oxidation to the (13). The German chemist Anton Köllisch was the first to epoxide, and acid-catalyzed epoxide isomerization to the 54 synthesize MDMA in 1912.49 His synthetic route began with ketone. the hydrobromination of 10 to produce 11 (Figure 2). The synthesis of MDMA from piperonal (13) is also common, and begins with a Henry reaction between 13 and nitroethane. The key nitrostyrene intermediate formed can then be partially reduced and hydrolyzed to produce ketone 12, or fully reduced using lithium aluminum hydride to afford 3,4-methylenedioxyamphetamine (MDA, 14).55 Conversion of MDA into the carbamate or formamide followed by lithium aluminum hydride reduction yields MDMA. Purification of MDMA is typically achieved following vacuum distillation of the freebase and/or crystallization of the hydrochloride salt.16 The hydrochloride salt can exist as one of several different hydrated forms.1 While the racemate is the most commonly administered form of MDMA, recent research suggests that there are distinct differences in the pharmacology of the two enantiomers. Hence, the development of efficient asymmetric strategies for producing enantiopure MDMA is incredibly important. Traditional resolution via selective crystallization of diastereo- meric salt forms has not proven the most effective route for synthesizing MDMA in high enantiomeric excess.56 Instead, a more successful strategy has relied on the use of removable chiral auxiliaries. The first asymmetric synthesis of MDMA was reported by Nichols and co-workers (Figure 3A).6 Reductive amination of ketone 12 with (S)-α-methylbenzylamine (15) produced the (S,S) intermediate 16 following crystallization. Figure 2. Common synthetic strategies used to produce racemic The use of Raney nickel at 50 psi appears to be crucial for the MDMA. selectivity of this reaction. In our hands, Raney nickel catalyzed hydrogenation did not proceed under atmospheric conditions, Displacement of the bromide with methylamine produces and the use of hydride reducing agents such as NaBH3CN MDMA.50 A similar route was described in the peer-reviewed yielded an inseparable 1:1 mixture of diastereomers (unpub-

Figure 3. Synthetic strategies used to synthesize enantiopure MDMA. (A) Reductive amination using (S)-α-methylbenzylamine. (B) Reductive amination using Ellman’s sulfinamide. (C) Ring opening of an (S)-alanine-derived aziridine.

2410 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review lished results). Palladium-catalyzed hydrogenolysis afforded membrane, MDMA reverses the flux of monoamines through MDA (14), which was converted to MDMA after reduction of their transporters, expelling intracellular serotonin, , the formamide. In 2014, Escubedo and co-workers reported a and norepinephrine into the extracellular space. Inhibitors of similar approach using Ellman’s sulfonamide as the chiral SERT, DAT, and NET completely prevent MDMA-induced auxiliary (Figure 3B).57 monoamine efflux in rat brain slices,67 and from monoamine The chiral pool has also been exploited to produce transporter-expressing HEK293 cells preloaded with radio- enantiopure MDMA. Usingamethoddevelopedby labeled monoamines.35 However, for monoamines to reach Nenajdenko and co-workers,58 (S)-alaninol (23) can be sufficiently high cytosolic levels to be reverse transported by protected and converted to the aziridine 24. Ring opening in membrane transporters, they must first be released from the presence of copper(I) iodide using Grignard reagent 25 synaptic vesicles into the intracellular space. By directly affords Ts-protected MDA (26). Methylation of 26 followed binding to vesicular amine transporters (VMAT), MDMA by deprotection yields (S)-MDMA.59 The enantiomeric excess reverses the transport of molecules like serotonin.68,69 of (S)-MDMA produced by Huot and co-workers was not Additionally, as a weak base, MDMA passively diffuses across reported, but based on the stereospecific nature of the vesicular membranes to collapse the pH gradient established reactions employed and the fact that the stereocenter is by VMAT, which is necessary for maintaining high unlikely to epimerize under these conditions, it is assumed that concentrations of monoamines in vesicles.70,71 Monoamines MDMA can be produced as a single enantiomer using this released from vesicles might be partially protected from strategy. Nenajdenko and co-workers reported that this is degradation due to the ability of MDMA to inhibit both 58 indeed the case for related β-arylalkylamines. isoforms of monoamine oxidase.72 Moreover, MDMA may cause SERT internalization,73 which presumably contributes to ■ PHARMACODYNAMICS increased extracellular serotonin levels. The releasing effects of Effects on Monoamines. The most well characterized MDMA are greater for serotonin and norepinephrine, and effect of MDMA is its ability to increase brain levels of slightly weaker for dopamine (Tables 3 and 4).65 monoamines such as serotonin, dopamine, and norepinephr- ine, which is accomplished via complex mechanisms. First, Table 3. Effects of MDMA on Monoamine Release Using MDMA binds to and inhibits the serotonin transporter Synaptosomes Prepared from Rat Brains: Values for EC a 50 (SERT), dopamine transporter (DAT), and norepinephrine (nM) ± Standard Deviations65, transporter (NET), inhibiting monoamine reuptake and − leading to increased extracellular levels of these amines.35,60 63 compd NE release DA release 5-HT release Electrophysiology experiments suggest that this inhibition S-(+)-Amph 7.07 ± 0.95 24.8 ± 3.5 1765 ± 94 results from MDMA serving as a substrate, rather than a (±)-MDMA 77.4 ± 3.4 376 ± 16 56.6 ± 2.1 blocker, of these transporters.64 In contrast to (S)-amphet- aNE = norepinephrine; DA = dopamine; 5-HT = serotonin. amine, racemic MDMA is a more potent inhibitor of SERT than either DAT or NET (Tables 1 and 2).65 In addition to Table 4. Effects of MDMA on Monoamine Release Using Monoamine-Preloaded HEK293 Cells Stably Expressing Table 1. Effects of MDMA on Monoamine Reuptake Using Human Monoamine Transporters: Values for EC (μM), a50 Synaptosomes Prepared from Rat Brains: Values for K with 95% Confidence Intervals in Parentheses62, a i (nM) ± Standard Deviations65, compd DA release 5-HT release compd NE uptake DA uptake 5-HT uptake S-(+)-Amph 1.76 (1.1−2.9) >33 S-(+)-Amph 38.9 ± 1.8 34 ± 6 3830 ± 170 (±)-MDMA 22 (8.9−53) 5.63 (3.5−9.2) (±)-MDMA 462 ± 18 1572 ± 59 238 ± 13 aNE = norepinephrine; DA = dopamine; 5-HT = serotonin. aNE = norepinephrine; DA = dopamine; 5-HT = serotonin. While much of the work elucidating the monoamine- Table 2. Effects of MDMA on Monoamine Transport releasing properties of MDMA have employed in vitro and ex Inhibition Using HEK293 Cells Stably Expressing Human vivo models, recently, the DA and 5-HT releasing effects of μ Monoamine Transporters: Values for IC50 ( M) with 95% MDMA have been observed in vivo using microdialysis in the 62 a Confidence Intervals in Parentheses , striatum and frontal cortex of rats.74 There is a general compd NE uptake DA uptake 5-HT uptake consensus that MDMA increases the release of monoamines; however, there is at least one study using fast-scan cyclic S-(+)-Amph 0.094 (0.06−0.14) 1.30 (0.83−2.0) >10 ± − − − voltammetry (FSCV) in brain slices that suggests that increases ( )-MDMA 0.447 (0.33 0.60) 17 (12 24) 1.36 (1.0 2.0) in monoamine concentrations following MDMA treatment a NE = norepinephrine; DA = dopamine; 5-HT = serotonin. might be due to inhibition of monoamine reuptake and not release per se.75 inhibiting the uptake of extracellular monoamines, MDMA also Direct Effects on Receptors. In addition to directly prevents transport of monoamines into vesicles. While, interacting with monoamine transporters, MDMA has been MDMA has been shown to inhibit the uptake of serotonin shown to bind with modest affinities to a variety of and dopamine into both synaptosomes and vesicles, it does not neuroreceptors including , , histaminer- affect the cellular uptake and/or vesicular packaging of either γ- gic, and muscarinic receptors.76,77,62 The binding profile of aminobutyric acid (GABA) or glutamate.66 MDMA across much of the recepterome is shown in Table 5. In addition to being a monoamine uptake inhibitor, MDMA The low micromolar affinities observed support the notion that is a potent releaser of these neurochemicals, and again, MDMA MDMA induces most of its effects indirectly by modulating accomplishes this via several mechanisms. At the cellular monoamine levels. The 5-HT2B is one of the few

2411 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review a Table 5. Binding Affinity Profile for (±)-MDMA

receptor radiolabeled radioligand classification species Ki (nM) ref 5-HT1A 3H-8-OH-DPAT human >10,000 77 5-HT1A 3H-8-OH-DPAT agonist human 12,200 62 5-HT1B 3H-GR-125743 antagonist human >10,000 77 5-HT1D 3H-GR-125743 antagonist human >10,000 77 5-HT1E 3H-5-HT agonist human >10,000 77 5-HT2A 3H-ketanserin antagonist rat >10,000 77 5-HT2A 3H-ketanserin antagonist human 7,800 62 5-HT2B 3H-LSD agonist human 500 77 5-HT2C 3H- antagonist rat >10,000 77 5-HT2C 3H-mesulergine antagonist human >13,000 62 5-HT3 3H- antagonist human >10,000 77 5-HT5 3H-LSD agonist human >10,000 77 5-HT6 3H-LSD agonist human >10,000 77 5-HT7 3H-LSD human >10,000 77 Alpha1A 125I-HEAT antagonist human >10,000 77 Alpha1A 3H- inverse agonist human >6,000 62 Alpha1B 125I-HEAT antagonist human >10,000 77 Alpha2A 3H- agonist human 2,532 77 Alpha2A 3H- antagonist human 15,000 62 Alpha2B 3H-clonidine agonist human 1,785 77 Alpha2C 3H-clonidine agonist human 1,123 77 Beta1 125I- partial agonist rat >10,000 77 Beta2 125I-pindolol partial agonist rat >10,000 77 CB1 3H-CP-55940 agonist rat (brain) >10,000 77 M1 3H-QNB antagonist human >10,000 77 M2 3H-QNB antagonist human >10,000 77 M3 3H-QNB antagonist human 1,851 77 M4 3H-QNB antagonist human 8,245 77 M5 3H-QNB antagonist human 6,339 77 nicotinic Alpha1Beta2 3H- agonist human >10,000 77 nicotinic Alpha2Beta2 3H-epibatidine agonist human >10,000 77 nicotinic Alpha2Beta4 3H-epibatidine agonist human >10,000 77 nicotinic Alpha3Beta2 3H-epibatidine agonist human >10,000 77 nicotinic Alpha3Beta4 3H-epibatidine agonist human >10,000 77 nicotinic Alpha7 3H-epibatidine agonist human >10,000 77 D1 3H-SCH23390 antagonist human >10,000 77 D1 3H-SCH23390 antagonist human >13,600 62 D2 3H-NMSP antagonist human >10,000 77 D2 3H- antagonist human 25,200 62 D3 3H-NMSP antagonist human >10,000 77 D3 3H-spiperone antagonist human >17,700 62 D4 3H-NMSP antagonist rat >10,000 77 D5 3H-SCH23390 antagonist human >10,000 77 GABA A 3H-muscimol agonist rat (forebrain) >10,000 77 GABA B 3H-baclofen agonist rat (forebrain) >10,000 77 NMDA 3H-MK-801 antagonist rat (forebrain) >10,000 77 H1 3H-pyrilamine antagonist human 2,138 77 H1 3H-Pyrilamine antagonist human >14,400 62 H2 3H-Pyrilamine antagonist human >10,000 77 prostaglandin EP3 3H-PGE2 agonist human >10,000 77 prostaglandin EP4 3H-PGE2 agonist human >10,000 77 NET 3H- inhibitor human >10,000 77 NET 3H-nisoxetine inhibitor human 30,500 62 DAT 3H-GBR 12935 inhibitor human >10,000 77 DAT 3H-WIN35,428 inhibitor human 6,500 62 SERT 3H- inhibitor human >10,000 77 SERT 3H-citalopram inhibitor human 13,300 62

2412 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review Table 5. continued

receptor radiolabeled ligand radioligand classification species Ki (nM) ref TAAR1 3H-RO5166017 agonist rat 370 62 TAAR1 3H-RO5166017 agonist mouse 2,400 62 aRadioligand binding assays were performed using stably or transiently expressing cell lines (HEK, HEKT, or CHO) unless noted otherwise.

− receptors that MDMA binds to with submicromolar affinity (K cin.18,38,91 96 It is possible that some of these hormonal i − = 500 nM), though the role of this receptor in the effects of changes are the result of serotonergic activity,97 99 and it is MDMA is unclear. For example, MDMA failed to produce a likely that they modulate some of the effects of MDMA.100 For response in a 5-HT2B functional assay using HEK293 cells.78 example, the rise in plasma DHEA levels was significantly However, it is believed that 5-HT2B agonism is at least partly correlated with feelings of euphoria.18 Furthermore, the effects responsible for the 5-HT releasing effects of MDMA, as of MDMA on oxytocin levels are often invoked to explain the pharmacological inhibition or genetic deletion of 5-HT2B drug’s prosocial effects. Dumont and co-workers were the first 79 receptors block MDMA-induced release of 5-HT. Binding of to demonstrate in a controlled laboratory setting that MDMA MDMA to 5-HT2B receptors was studied using a radiolabeled increases oxytocin levels.93 They also found that increases in agonist, while many of the other receptor binding assays (e.g., blood oxytocin levels were correlated with the subjective 5-HT2A ad 5-HT2C) utilized radiolabeled antagonists. prosocial feelings induced by MDMA more so than blood ffi Therefore, it is possible that the binding a nity of MDMA levels of the drug itself. While numerous other studies have for many receptors has been underestimated. For example, it is replicated the finding that MDMA elevates oxytocin levels, now well established that MDMA binds directly to 5-HT2A they have all failed to reproduce a correlation between ffi receptors, albeit with micromolar a nity, though binding oxytocin levels and prosocial feelings, calling into question 3 assays performed with H-ketanserin do not always capture the relevance of this hormone for the prosocial effects of this interaction. Furthermore, MDMA is unable to displace MDMA.100,101 As such, the role of oxytocin in the effects of radiolabeled monoamine transporter inhibitors despite exhibit- − MDMA is currently controversial. ing nM potency in functional assays (Tables 1 4), which is Behavioral Effects in Rodents. Like other serotonergic consistent with its proposed role as a monoamine releaser psychedelics, MDMA produces behavioral effects consistent rather than a competitive uptake inhibitor. with serotonin syndrome such as flat body posture, hind limb -Associated Receptor 1 (TAAR1). The abduction, and forepaw treading.102 At lower doses, MDMA trace-amine associated receptor (TAAR1) has also been “ ” fi ff produces amphetamine-like hyperactivity in the open eld. suggested as a key target mediating the e ects of MDMA. Both of these effects are enhanced following repeated Bunzow and co-workers demonstrated that MDMA acts as an administration of MDMA, demonstrating that MDMA is agonist at rat TAAR1 receptors to increase cAMP production 103 capable of producing behavioral sensitization. Behavioral in a TAAR1-expressing HEK293 cell line.80 Like MDMA, sensitization is correlated with the enhanced ability of MDMA several other hallucinogens and psychostimulants have been to increase monoamine levels (measured via microdialysis) shown to bind to and activate TAAR1 to a greater extent than following repeated dosing.104 The locomotor effects of MDMA neurotransmitters such as serotonin, dopamine or norepi- 80−82 are perhaps the best-studied behavioral responses in rodents, nephrine. Due to the known modulatory influence of 83 and they are modulated by a variety of neuroreceptors TAAR1 on monoamine transporter function, it is likely that 105 106 107 107 TAAR1 activation contributes to the effects of MDMA on including 5-HT1B, 5-HT2A, D1, and D2 receptors. Unlike amphetamine, selective serotonin reuptake inhibitors extracellular monoamine levels. Interestingly, 4-hydroxyam- 108 phetamine proved to be a particularly potent agonist of block MDMA-induced increases in locomotion. Further- more, MDMA does not produce this behavioral effect in mice TAAR1 (EC50 = 51 nM). As MDMA is metabolized into 4- hydroxy-substituted compounds, there is the distinct possibility genetically lacking SERT, further implicating this monoamine ff 109 that metabolites of MDMA may potently activate TAAR1. transporter in the hyperlocomotive e ects of MDMA. However, to the best of our knowledge, this hypothesis has not In rodent models of anxiety, MDMA produces complex effects. At low acute and subchronic doses, MDMA tends to be been directly tested. Finally, it is unclear if TAAR1 plays any 110−112 role in the effects of MDMA in humans, as MDMA does not anxiogenic in the elevated plus maze (EPM). However, activate human TAAR1 in cellular assays like it does mouse at higher acute and subchronic doses, MDMA produces ff and rat TAAR1.84 e ects in the EPM. When tested in the light-dark Sigma-1 Receptor. Radioligand binding studies have box paradigm, MDMA does not alter preferences of mice for 113 shown that MDMA binds to both sigma-1 and sigma-2 the two compartments. receptors with Ki values in the low micromolar range, which Some of the rodent behaviors most relevant to potential are comparable to the affinities of MDMA for monoamine therapeutic uses of MDMA are related to social behaviors. A 5 transporters.85 Moreover, treatment with BD1063, a selective mg/kg dose of MDMA decreased aggressive behaviors in rats sigma-1 antagonist, blocked the effects of MDMA on rodent and increased the time spent engaging in social behaviors such locomotion.85 The sigma-1 receptor has been proposed to be a as sniffing, following, crawling under, crawling over, mutual − novel target for the treatment of depression and anxiety,86 90 grooming, and adjacent lying.114 Additionally, MDMA has and it is reasonable to hypothesize that this receptor plays been shown to induce a social conditioned place preference.115 some role in the behavioral and clinical effects of MDMA. Adjacent lyinga behavior in rats where two unfamiliar Hormonal Effects. Administration of MDMA to humans animals lie passively next to each otheris perhaps one of the leads to robust increases in plasma levels of , prolactin, more robust prosocial behaviors induced by MDMA in (DHEA), vasopressin, and oxyto- rodents.

2413 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

In terms of mechanism, systemic MDMA increases plasma that MDMA, and several other psychedelic compounds, levels of oxytocin in rats and activates oxytocinergic neurons in significantly increased the complexity of dendritic arbors in the hypothalamus, as measured by Fos immunohistochemis- cultured cortical neurons128 Moreover, this phenotype is not try.99 Increases in oxytocin levels and adjacent lying behavior produced by all psychostimulants and drugs of abuse, as S- induced by MDMA were abolished by treatment with a 5- (+)-amphetamine had no effect.128 Future studies should HT1A antagonist, while 8-OH-DPAT (a 5-HT1A agonist) assess the in vivo effects of a single moderate dose of MDMA produced effects similar to MDMA.99 This led McGregor and on dendritic branching and spine density. co-workers to propose that MDMA induces oxytocin release via stimulation of 5-HT1A receptors, and that increased ■ METABOLISM AND PHARMACOKINETICS adjacent lying resulted from activation of oxytocin receptors. The primary routes for metabolism of MDMA are N- This hypothesis was supported by the fact that intra- demethylation and loss of the methylene bridge connecting cerebroventricular administration of tocinoic acid, an oxytocin the catechol (Figure 4), both of which are mediated by various , blocked MDMA-induced adjacent lying.99 However, in a follow-up study, McGregor and co-workers could not prevent MDMA-induced adjacent lying using C25,116 a systemically administered non-peptidic antagonist of oxytocin receptors. In contrast, they were able to prevent this behavior using an antagonist of the vasopressin receptor 1A.117 There are two possibilities that might explain these contradictory results. First, tocinoic acid could have non- selective antagonistic effects at the vasopressin receptor 1A. Alternatively, C25 might not have been able to cross the blood- brain barrier. In addition to its prosocial effects, MDMA has been shown by Howell and co-workers to promote fear extinction learning in mice.118 This seminal study potentially provides a mechanistic explanation for the therapeutic efficacy of MDMA in patients suffering from treatment-resistant PTSD (vide infra). Similar findings have been described for other psychedelics such as psilocybin in mice119 and N,N- dimethyltryptamine (DMT) in rats.120 The facilitation of fear extinction memory by MDMA appears to be dependent on 121 Figure 4. Common metabolites of MDMA. CYP = cytochrome P450; SERT. COMT = catechol-O-methyltransferase. Plasticity-Promoting Effects. Like most psychostimu- lants, MDMA causes robust changes in gene expression and 122 protein levels associated with neural plasticity. Acute cytochrome P450s.129 The common metabolites of MDMA treatment with MDMA (10 mg/kg) causes differential gene (1)includeMDA(14), 3,4-dihydroxymethamphetamine expression of BDNF in the frontal cortex and hippocampus of (HHMA, 28), 3,4-dihydroxyamphetamine (HHA, 29), 4- rats, with BDNF levels increasing in the former brain region hydroxy-3-methoxy-methamphetamine (HMMA, 30), and 4- and decreasing in the latter.123 The administration of 4 doses hydroxy-3-methoxy-amphetamine (HMA, 31). The major over a period of 6 h to rats led to robust increases in BDNF metabolite of MDMA in humans is HMMA, which is mainly transcript levels in several cortical regions both 1 and 7 h excreted as the glucuronic acid conjugate.130 following dosing.124 The largest effects were seen in the Recent genetic findings suggests that a variety of cytochrome prefrontal cortex with increases in TrkB expression observed in P450s, including CYP2C19, CYP2B6, and CYP1A2, play a role that region 24 h after dosing.124 Here, MDMA produced in the demethylation of MDMA.131,132 Mutations in the weaker effects on NT3 and TrkC gene expression.124 Chronic CYP2C19 or CYP2B6 genes that reduce enzyme function have treatment with MDMA in mice125 and subchronic admin- been shown to increase the ratio of MDMA/MDA but do not istration of large doses (20 mg/kg) in rats126 led to increases in alter HMMA concentrations.131,132 Subjects with decreased BDNF transcription and translation in the hippocampus. The CYP2C19 function also showed greater cardiovascular latter study also observed a reduced number of dendritic spines responses with faster onset times. Mutations in the CYP2B6 in the hippocampus of rats. Finally, MDMA was observed to gene resulting in decreased enzyme function only influenced inhibit neurite outgrowth in PC12 cells,127 though the metabolism at later time points (i.e., 3−4 h) suggesting that it relevance of this cell line to studies on neural plasticity is is a secondary metabolizer of MDMA.131 debatable. When MDMA is administered to humans at a dose of 100 To date, most studies assessing the psychoplastogenic effects mg, it has a half-life of approximately 8−9 h and yields plasma 133 of MDMA have observed a reduction in dendritic branching Cmax and tmax values of 222.5 ng/mL and 2.3 h, respectively. and/or dendritic spine numbers. However, these studies are However, MDMA is known to exhibit nonlinear pharmacoki- often conducted using extremely high doses of MDMA netics in both humans131,134,135 and squirrel monkeys.136 This administered over extended periods of time, and probably means that increasing doses of MDMA prolong its half-life, more accurately reflect neurotoxicity resulting from over- potentially exacerbating the risk for adverse effects and stimulation of psychoplastogenic receptors. More modest neurotoxicity. The nonlinear pharmacokinetics observed doses would likely yield increases, as opposed to decreases, following administration of MDMA is likely the result of in dendritic branching and spine density. Recently, we reported cytochrome P450 inhibition by MDMA and its metabo-

2414 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review lites.131,137,138 Additionally, the enantiomers of MDMA are before.163 Consuming MDMA as a part of a drug metabolized at different rates, with the R-enantiomer having a mixture can be extremely dangerous due to drug−drug − longer half-life than the S-enantiomer.139 141 interactions,164 and has important implications for evaluating the neurotoxic potential of MDMA in humans. ■ ADVERSE EFFECTS Certainly, the most controversial aspect of MDMA Similar to other , MDMA produces a number of pharmacology is its potential to induce neurotoxicity. The ff neurotoxic effects of MDMA have been extensively reviewed adverse e ects including trismus, tachycardia, bruxism, dry 165−171 mouth, palpitations, diaphoresis, and insomnia.15,19,36,142 by others, and thus, we will focus only on the key Rhabdomyolysis, cardiac arrhythmias, hyperthermia, hypona- studies. Additionally, we will attempt to highlight why this is tremia, and acute renal failure are the more severe side-effects such a contentious area and why the controversy is not likely and are common causes of death following MDMA to be resolved soon. intoxication.143,144 Themoresevereadverseeffects of People who consume MDMA (particularly those who do so MDMA are potentially exacerbated by the intense exercise regularly, and in high doses) perform poorly on various tests and hot environment characteristic of raves. In Long-Evans related to attention, learning, and memory (e.g., working and ı̈ rats, slight increases in ambient temperature resulted in declarative memory) when compared to MDMA-na ve 172−175 excessive brain hyperthermia leading to death at an MDMA controls. Those with a history of only moderate fi MDMA use do not seem to exhibit memory impairments.176 dose that is signi cantly lower than the LD50 in rats at room temperature.145 Similarly, Fantegrossi and co-workers found However, acute MDMA intoxication produces memory 176 that MDMA lethality was increased when NIH Swiss mice deficits. Heavy MDMA users tend to have lower cerebral were housed at high densities (>6 mice per cage), which spinal fluid levels of 5-hydroxyindoleacetic acid (5-HIAA) reduces the ability to dissipate body heat.146 Risk for serotonin the principal metabolite of serotoninand thus, serotonergic 177 syndromea collection of symptoms that include high body toxicity has been presumed. In general, neuroimaging temperature, sweating, and tremor (among others)increases studies used to assess the effects of MDMA in humans have with higher doses of MDMA.147 produced mixed findings, with no clear evidence that MDMA 178,179 The effects of MDMA on heart function are also significant, is safe or neurotoxic. Finally, when compared with with norepinephrine mediating a significant portion of the MDMA-naıvë controls, MDMA users are more likely to be cardiostimulant effects observed following MDMA adminis- afflicted with mental illnesses including depression, psychotic 180 tration.34,35,148,149 In addition to increasing systolic blood disorders, eating disorders, and anxiety disorders. pressure,19,36,92 the drug can induce cardiac arrhythmias and While retrospective studies on MDMA-using populations myocarditis.150 Myocardial infarction can also occur following are certainly important, there are several confounding factors MDMA use, though this tends to happen less frequently than that limit the interpretability of these data. First, MDMA after or amphetamine administration.151,152 In the long produced by clandestine laboratories is often contaminated term, MDMA use can result in valvular heart disease,153 which with other drugs of abuse and neurotoxic compounds such as could be due to oxidative stress154 or the activation of 5-HT2B methamphetamine. Second, recreational MDMA users are 181 receptors by MDMA.77 typically polydrug users. Third, recreational MDMA is often In terms of the addictive potential of MDMA, the data are consumed at crowded dance parties (i.e., raves), where mixed. Several people have argued that MDMA has lower excessive activity, high temperatures, and dehydration could abuse potential because recreational users have reported that exacerbate any inherently neurotoxic effects of the drug. its pleasurable effects diminish with repeated use, but its side Together, these facts make it difficult, if not impossible, to effects increase.15 However, in animal models, MDMA does distinguish the neurotoxic effects induced by MDMA itself produce some of the same behavioral effects characteristic of versus those caused by impurities, drug-drug interactions, or addictive drugs like cocaine and , albeit to a lesser drug-environment interactions. Furthermore, due to the extent. For instance, MDMA is known to produce conditioned retrospective nature of many human studies regarding the place preference in rats155 and mice,156,157 and MDMA is self- effects of MDMA, it is unclear if the cognitive impairments and administered by a variety of species (e.g., rats, mice, non- neuropsychiatric disorders observed in groups who have used human primates).158 Interpretation of self-administration MDMA reflect a cause or consequence of MDMA use. studies using MDMA are complicated by a variety of factors Prospective studies are incredibly important for answering such as dose, timing, and prior exposure of the test animals to these questions. One prospective study from the Netherlands other drugs of abuse. For an overview of these issues, we refer found that sensation-seeking, impulsivity, and depression did the reader to an excellent review by Susan Schenk.158 Taken not predict future MDMA use.182 However, a much larger together, MDMA does seem to have reinforcing properties, but study from Germany concluded that MDMA users had these appear to be significantly weaker than those of cocaine. significantly higher risk for nearly all DSM-IV mental disorders, Determining the adverse effects of MDMA in people who and moreover, that the onset of these disorders typically consume it recreationally is complicated by the fact that some preceded the first use of MDMA.183 “MDMA” sold on the street does not contain any MDMA at Because of the many factors that can confound human all,159 while other batches of illegally produced “MDMA” are studies, researchers have turned to well controlled model adulterated.160 Contaminating drugs include, but are not systems in the laboratory to investigate MDMA neurotoxicity. limited to, amphetamine, methamphetamine, MDA, pseudoe- However, the relevance of these models to human neuro- phedrine, , and caffeine.161,162 Many recreational toxicity is often questioned. Capela and co-workers found that MDMA users prefer “molly” as it is believed to be of high MDMA can induce apoptotic cell death in embryonic rat purity, however, a recent study employing hair follicle testing cortical neurons via a 5-HT2A-dependent mechanism.184 revealed that 48% of molly users tested positive for synthetic Furthermore, they discovered that the metabolites of MDMA cathinones despite having reported that they had never used are more potent neurotoxins.185 Similarly, Stumm and co-

2415 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review workers reported that MDMA and related amphetamines kill using model systems that more closely recapitulate the cultured rat cortical neurons at comparable concentrations.186 pharmacokinetics of MDMA in humans may be more useful. It is important to note that the concentration of MDMA Like rats, non-human primates experience serotonergic required to produce substantial neurotoxic effects in these neurotoxicity following administration of large doses of studies is >200 μM, while the maximal brain concentration of MDMA.203,204 Unlike rats, these changes seem to be relatively MDMA in rats following a 20 mg/kg subcutaneous dose (10x long-lasting in most primate brain regions.205,206 Abnormal the behaviorally relevant dose) is only ca. 100−200 μM.187 At serotonergic innervation patterns were observed 7 years a more modest concentration (10 μM), our group determined following MDMA exposure in squirrel monkeys,207 and these that MDMA produced robust psychoplastogenic effects in patterns seemed to result from axotomy as raphe cell bodies embryonic rat cortical cultures without cell death.128 For remained intact.207 In rhesus monkeys, persistent decreases in comparison, we have observed that several SSRIs and cerebrospinal fluid levels of 5-HIAA were accompanied by commonly prescribed are cytotoxic functional changes as measured by electrophysiology.208 Most to cultured rat cortical neurons in the range of 10−100 μM of the studies assessing the neurotoxic effects of MDMA in (unpublished results). primates administered multiple subcutaneous doses. However, In addition to studies using cultured neurons, in vivo animal humans typically consume a single oral dose of MDMA either models are frequently used to test the neurotoxic potential of recreationally or during MDMA-assisted psychotherapy (vide MDMA. While findings dating back to 1987 suggest that infra). To address this discrepancy, Ricaurte and co-workers MDMA has neurotoxic effects in animals,188 the relevance of compared both dose frequency and route of administration in these models to human neurotoxicity is highly debated. Some squirrel monkeys. They found that repeated dosing and of the contentious questions the field has to grapple with subcutaneous administration produces greater neurotoxic include (1) what dosing paradigm most effectively models effects than oral dosing.209 Importantly, they found that a human use, (2) what species is most relevant, (3) is allometric single, modest (5 mg/kg), oral dose of MDMA still produced scaling appropriate, (4) how should the nonlinear human serotonin depletion in the thalamus and hypothalamus 2 weeks pharmacokinetics of MDMA be factored in, (5) what route of after administration.209 administration should be utilized, and (6) how should The mechanism of MDMA-induced neurotoxicity probably “neurotoxicity” be defined/measured (e.g., monoamine levels, involves a combination of mechanisms including glutamate- neurite degeneration, cell body loss). induced excitotoxicity,210 increased oxidative stress,211 hyper- In mice, MDMA tends to produce dopaminergic, but not thermia,212 mitochondrial damage, and increased inflamma- − serotonergic, neurotoxicity.189 191 This is in sharp contrast to tion.169,171 While the results of the numerous studies rats, for which the opposite seems to be true. Two weeks investigating MDMA-induced neurotoxicity still leave ques- following systemic administration of MDMA to rats (20 mg/ tions unanswered about the safety of MDMA administered to kg, subcutaneous, twice daily for 4 d) loss of 5-HT axons (but humans, it is reasonable to conclude that use of MDMA under not axons) projecting to the forebrain was common recreational conditions (e.g., high doses, multiple observed.192 Interestingly, axonal degeneration was not doses, polydrug use, high temperatures, prolonged physical accompanied by loss of raphe cell bodies.192 As a result, activity, dehydration, etc.) is likely to cause adverse effects. serotonergic axons regenerate in rats administered MDMA, However, in controlled studies in the clinic using low doses to however, it is unknown how well these newly sprouted axons assist psychotherapy, MDMA may be safe and well tolerated, function.193 Additionally, large doses of MDMA produce as discussed below. When a variety of factors were considered, − reductions in levels of 5-HT, 5-HIAA, and SERT.194 196 Levels including physical, social, and economic factors, MDMA of 5-HT reuptake sites in rats partially recovered 6 months consistently ranked as being less harmful than illegal drugs following MDMA exposure and were fully recovered after 1 such as heroin, cocaine, and methamphetamine, as well as legal 213,214 year.194,197 Similarly, 8 doses of MDMA given to rats over 4 drugs such as and . days decreased brain 5-HT2 receptor levels by 80% when measured 6 h after the last dose.198 Receptor levels recovered ■ POTENTIAL USE IN MEDICINE to 62% after 24 h and were completely normalized after 21 In recent years there has been renewed interest in using days.198 The MDMA-induced serotonergic neurotoxicity in psychedelic compounds like psilocybin and MDMA to treat − rats is exacerbated by increased ambient temperature199 and neuropsychiatric disorders.215 217 This should not be surpris- can be prevented by blocking SERT with fluoxetine.200 It ing because before MDMA was placed on the Schedule I list, it should be noted that the doses of MDMA used in rats and was widely used by some psychiatrists to assist in treating a mice to induce neurotoxicity are much higher than those often variety of disorders including anxiety disorders and depression. used by humans. Some researchers have justified these large The benefits of MDMA were believed to result from increased rodent doses on the basis of allometric scaling201 and the fact introspection, a decrease in fear response upon accessing that experienced recreational users of MDMA often develop painful memories, and the promotion of trust between patients tolerance, leading them to ingest multiple doses in a short and their therapists.218,219 However, most of the work period of time to achieve the desired subjective effects of the conducted during this period yielded only anecdotal reports, drug.202 Others have argued that MDMA doses used in and there were no placebo-controlled clinical trials conducted animals are too high, as MDMA produces behavioral effects at that adhered to current rigorous standards. approximately the same dose (1−2 mg/kg) in humans and In contrast, recent clinical studies assessing the therapeutic rats.167 Finally, it has been posited that species differences in potential of MDMA for treating PTSD are carefully controlled − metabolism and neurotoxicity (e.g., dopaminergic toxicity in and well documented.220 222 First, patients are screened for mice vs serotonergic toxicity in rats) suggest that the medical conditions, including various neuropsychiatric dis- metabolites of MDMA, and not necessarily MDMA itself, are orders, that might exclude them from the study. Next, they are responsible for the neurotoxic effects of MDMA.168 Therefore, assessed at baseline using the Clinician-Administered PTSD

2416 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

Scale (CAPS). Patients then receive training sessions to face had a negative emotional valence, MDMA-treated establish rapport with an experienced clinician. The environ- individuals performed poorly.229 Moreover, Carhart-Harris ment is carefully controlled so that it is aesthetically pleasing and co-workers found that while under the influence of and resembles a living space rather than a medical facility. MDMA, participants rated their best and worst memories as Music is often used to facilitate relaxation and/or evoke being significantly more positive and less negative, respec- emotions. Both a male and a female therapist are present for tively.230 Related to its subjective effects, MDMA increased the duration of the treatment session. After the drug is bilateral blood flow in the ventromedial prefrontal cortex and administered, there is limited verbal communication between reduced blood flow in the left amygdala231two brain regions the therapists and the patient. Instead, the patient is that play important roles in the processing of emotional stimuli encouraged to explore any feelings that the experience might and memories. Due to its general tendencies to reduce evoke. The therapists provide nurturing physical contact responses to threatening stimuli while enhancing responses to whenever necessary to help ease tension or distress. After the positive social cues, MDMA is being investigated for treating MDMA session, the patient receives additional non-drug social anxiety in autistic adults,232 and it has been suggested psychotherapy sessions. that MDMA may prove useful in other conditions with a An effective dosing paradigm was established by Oehen and significant social component.48 co-workers utilizing low dose MDMA as an active placebo.221 Finally, MDMA may hold some promise for treating The use of an active placebo is an important part of the substance use disorders (SUDs).233,234 Initial reports suggest experimental design implemented by Oehen and co-workers. that MDMA might decrease substance use,235 and a pilot study Inactive placebos, such as lactose, fail to produce physiological conducted by Howell and co-workers demonstrated that R- and psychological responses noticeable to trained clinicians or (−)-MDMA decreased response rates during a cocaine self- experienced MDMA users. This raises the question as to administration paradigm in squirrel monkeys.236 Though very whether or not studies utilizing inactive placebos can truly be few animals were used in the latter study, the results are considered double-blind experiments. Patients in the exper- encouraging. While other psychedelic compounds such as imental treatment group received an initial dose of 125 mg of LSD, psilocybin, and have been more extensively MDMA followed by an additional 62.5 mg after 2.5 h. The studied than MDMA with respect to their abilities to treat active placebo group received an initial dose of 25 mg of SUDs, the minimal perceptual disturbances caused by MDMA MDMA followed by an additional 12.5 mg 2.5 h later. The may offer a distinct advantage over the classical hallucinogens. dose of MDMA used for the active placebo group was chosen S-(+)-MDMA vs R-(−)-MDMA. While racemic MDMA is to stimulant mild but detectable psychological effects. the form used both recreationally and in clinical trials, The most common use for MDMA in medicine is as an preclinical work and some human data suggest that there are adjunct to psychotherapy for treating anxiety disorders.223 Of distinct differences between the R- and S-enantiomers of particular note is recent clinical work demonstrating that MDMAthe non-superposable mirror images of each other.13 MDMA can produce beneficial effects in treatment-resistant The R- and S-enantiomers are sometimes referred to as the l- PTSD patients when it is paired with psychotherapy.220,221,224 and d-enantiomers, respectively. An excellent review on this The beneficial effects of this treatment paradigm seemed to be subject was published recently by Howell and co-workers,236 relatively long-lasting, as demonstrated by follow-up studies so we will only cover the highlights here. conducted several years later.225 A recent meta-analysis Regarding the monoamine releasing and reuptake inhibiting determined that MDMA-assisted psychotherapy produced properties of MDMA, there is a general consensus that the S- − larger effect sizes in both clinician-observed outcomes and enantiomer is the more potent compound.59,77,237 245 This is patient self-reports when compared to prolonged exposure consistent with what is known about the effects of S- therapy.226 Furthermore, fewer patients in the MDMA-assisted (+)-amphetamine on monoamine levels. However, R- psychotherapy group dropped out of the study.226 These (−)-MDMA appears to be a more potent direct binder of 5- studies and others have indicated that MDMA was well HT2A receptors (Table 6),246,247 which perhaps explains why tolerated when administered in a clinical setting as a single it has a greater propensity for causing perceptual disturbances. dose in the range of 75−125 mg.19 Recently, MDMA was Neither enantiomer is particularly effective at stimulating granted “breakthrough therapy” status by the FDA for the phosphatidyl inositol turnover in either 5-HT2A or 5-HT2C treatment of PTSD. The phase III clinical trials are estimated expressing cells.248 When rats were trained to discriminate S- to be completed within the next five years, and if the results are (+)-amphetamine, LSD, and saline from each other in a 3-lever positive, it is anticipated that a New Drug Application for drug discrimination paradigm, R-(−)-MDMA and S- MDMA will be submitted to the FDA around 2021.227 (+)-MDMA produced more hallucinogen-like and amphet- Recent clinical work to understand the mechanism of amine-like discriminative stimuli, respectively.249 Furthermore, MDMA’s therapeutic effects has revealed that this drug experiments using mice trained to discriminate either S- impacts the processing of emotionally salient information. (+)-MDMA or R-(−)-MDMA from vehicle demonstrated that Using functional magnetic resonance imaging (fMRI), de Wit the S-enantiomer produced more psychostimulant-like effects and co-workers found that MDMA attenuated the blood- while the R-enantiomer was more hallucinogen-like.250 oxygen-level dependent (BOLD) response to angry faces in the In terms of their influences on hormone levels, the amygdala, while also enhancing the activation of the ventral enantiomers of MDMA also have differential effects. Ex vivo striatum in response to happy faces.228 In this study, MDMA studies utilizing rat hypothalamus tissue demonstrated that S- also impacted the performance of people during the Reading (+)-MDMA is a more potent inducer of oxytocin release than the Mind in the Eyes Testa test that has participants attempt the racemate, while R-(−)-MDMA has no effect.251 However, to predict what a person is thinking/feeling based on a picture R-(−)-MDMA was more effective at increasing the activation of their face. Specifically, MDMA improved scores when the of hypothalamic oxytocinergic neurons, as measured by the stimulus had a positive emotional valence. However, when the number of c-fos positive neurons.236 Both enantiomers appear

2417 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

Table 6. Binding Affinity Profile for (±)-MDMA, R-(−)-MDMA, and S-(+)-MDMA246 ± Ki(nM) SEM receptor radiolabeled ligand radioligand classification (±)-MDMA R-(−)-MDMA S-(+)-MDMA 5-HT1 3H-5-HT agonist 6,850 ± 1,300 4,200 ± 500 >10,000 5-HT2 3H-ketanserin antagonist 8,300 ± 1,100 3,310 ± 140 >10,000 D2 3H-NMSP antagonist >10,000 >10,000 >10,000 to increase vasopressin secretion comparably from the “” and “”, which refer to the R- and S- hypothalamus ex vivo.251 R-(−)-MDMA more potently enantiomers of the fast-acting , increased plasma prolactin levels in rhesus macaques.244 respectively. If armdma proves to be an effective and safe Pretreatment with fluoxetine attenuated this effect, but did therapeutic in humans, we hope this new terminology will not block it completely. The selective 5-HT2A antagonist eliminate any potential stigma associated with using a M100907 was required to completely inhibit R-(−)-MDMA- perceived “party-drug” as a medicine. induced increases in prolactin, suggesting that indirect effects on 5-HT levels, as well as direct binding to 5-HT2A receptors ■ HISTORY AND IMPORTANCE IN NEUROSCIENCE contribute to the ability of R-(−)-MDMA to increase prolactin levels.245 Urban legend, rumor, and myth have clouded the true history Behaviorally, both enantiomers increase affiliative social of MDMA. Several excellent historical accounts of the ff discovery and development of MDMA have been reported behaviors in squirrel monkeys, and this e ect seems to be 44,49,255,256 dependent on activation of 5-HT2A receptors.252 In mice, R- previously, and thus, we only discuss the highlights (−)-MDMA and the racemate (but not S-(+)-MDMA) here (Figure 5). First, it is a common misconception that increased social interaction and facilitated fear extinction MDMA was originally designed to be an appetite suppressor or learning, effects that could be relevant to using MDMA as a a weight loss drug. Instead, MDMA originated from a therapeutic.253 Furthermore, the R-enantiomer did not campaign by Merck to sidestep a patent on the hemostatic ’ increase locomotor activity, a behavioral effect commonly drug hydrastinine held by Bayer, one of Merck s top rivals. In 253 fi produced by psychostimulants. fact, MDMA was rst synthesized in 1912 and subsequently As discussed, the primary concern for using MDMA in the patented, but as it was only intended to be an intermediate en clinic is its potential neurotoxicity. Most neurotoxicity studies route to the desired compound, its biological activity was not were performed using the racemate, however, there is some assessed. It was not until 15 years after its initial synthesis that evidence to suggest that the neurotoxic effects of MDMA stem MDMA was actually tested in animal models. Merck was from the S-enantiomer, with the R-enantiomer being relatively interested in identifying compounds that mimicked the effects 49 benign. Unlike R-(−)-MDMA, S-(+)-MDMA increased body of epinephrine, and MDMA was one candidate tested owing temperature and promoted the activation of microglia and to its structural similarities. Unfortunately, the results of these 255 astroglia.254 However, this study employed a relatively low tests could not be found in the Merck archive. dose of R-(−)-MDMA. To more definitely establish a lack of Research on MDMA appeared to stagnate until the 1950s. neurotoxicity following R-(−)-MDMA administration, Howell At that time the US military began using mescaline-like and co-workers administered high doses of R-(−)-MDMA compounds, including MDMA, as part of pharmacologically 257 (four injections of 50 mg/kg given over 2 days) to mice and assisted interrogation programs. In essence, they were trying compared effects to those produced by the racemic mixture to identify so-called “truth drugs”compounds capable of (four injections of 20 mg/kg given over 2 days).253 These lowering inhibitions making people more likely to reveal secret authors assessed body temperature, mortality, and markers of information. The chemical warfare code of MDMA was EA- 44 neurotoxicity. Unlike the racemate, high dose R-(−)-MDMA 1475. The methylenedioxy-containing entactogens, such as did not influence body temperature or survival. Furthermore, MDA and MDMA, were of particular interest to the military the R-enantiomer had no effect on glial fibrillary acidic protein because these compounds tended to encourage people to (GFAP) immunoreactivity, DA content, or DAT expression. speak more openly without causing overwhelming perceptual Theracematesignificantly increased astrogliosis while disturbances. The characteristic hallucinations produced by decreasing both DA content and DAT expression. This study compounds like LSD and mescaline typically disrupted provides compelling evidence that at least in mice, the R- interrogation sessions. In the early 1950s, the military began enantiomer of MDMA lacks many of the negative effects testing several of these compounds on patients at the New associated with the racemate, while still maintaining the ability York State Psychiatric Institute. In 1952, a patient named to promote social interaction and to facilitate fear extinction Harold Blauer was administered several compounds over the learning. course of a month before succumbing to a fatal dose of MDA Thus, R-(−)-MDMA may be an effective pharmaceutical (450 mg).257 Realizing that safety data on these compounds with an acceptable therapeutic index. However, neurotoxicity were woefully lacking, the military contracted a group at the and other negative effects associated with S-(+)-MDMA and University of Michigan to conduct pharmacokinetic and safety racemic MDMA will always be associated with the acryonym studies in mice, rats, guinea pigs, dogs, and monkeys.258 After “MDMA,” having the potential to bias regulatory bodies, declassification, these data were published in 1973 and doctors, and patients. Therefore, to identify a term suitable for revealed that the methylenedioxy compounds were more common parlance, but devoid of negative connotations, we toxic than their methoxy counterparts.258 suggest the use of the alternate terms “armdma”, “esmdma”, The first report of the synthesis of MDMA in the peer- and “racmdma” to refer to R-(−)-MDMA, S-(+)-MDMA, and reviewed literature was in 1960.259 Afterward, MDMA (±)-MDMA, respectively. These terms are analogous to remained relatively unexplored until Alexander Shulgin learned

2418 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

Figure 5. Timeline of important events related to research on MDMA and classical psychedelics. Note the different peaks for research on MDMA and LSD. The data were obtained from a PubMed search for papers having titles that contained the search terms “3,4- methylenedioxymethamphetamine or MDMA” and “lysergic acid diethylamide or LSD” conducted on March 20, 2018. of the unique effects of the compound and tested MDMA on Science paper a year later.263 Further analysis revealed that himself in 1976.260 Thereafter, Shulgin distributed it to friends animals used in the original study were likely dosed with and psychotherapists in northern California, who began using methamphetamine, a known dopamine neurotoxin, instead of MDMA to facilitate psychotherapy. Shulgin and Nichols were MDMA, due to a mix-up in the labeling of sample vials. the first to publish on the effects of MDMA in humans in Despite its retraction, the Ricaurte study had dealt a serious 1978.13,14 Though Shulgin is often credited with the blow to the credibility of MDMA as a safe therapeutic. Heated rediscovery of MDMA,260 the role of David Nichols should public debate ensued about the potential dangers of the drug also be emphasized as Nichols was a co-author on these first and its government regulation. In 2009, David Nutt published reports of the effects of MDMA in humans. Furthermore, he an editorial where he compared the dangers of using ecstasy (1 was largely responsible for reclassifying MDMA and related serious adverse event in 10,000) to those of horseback riding compounds as entactogens, due to their unique qualities or “equasy” (1 serious adverse event in 350).264 This editorial relative to hallucinogens and psychostimulants.41 highlighted the fact that people in the scientific community felt During the period from 1978 to 1985, it is estimated that that government agencies were not using objective criteria for thousands of patients were treated with MDMA.216 However, assessing risk when establishing regulations for psychoactive these initial studies did not adhere to the same rigorous compounds like MDMA. Since the retraction of the Ricaurte standards that we demand of clinical trials today. As a result, study, there have been multiple clinical trials investigating the the true therapeutic potential of MDMA was not captured in effects of MDMA, and thus far, all data suggest that MDMA the scientific literature. Furthermore, the properties of MDMA can be administered safely under these conditions. that made it an effective aid to psychotherapy also led to its In 2011, the first completed clinical trial evaluating the widespread use in social situations. During this period of time, potential of MDMA-assisted psychotherapy for alleviating recreational use of MDMA increased dramatically, and treatment-resistant PTSD was published.220 The results were mounting evidence suggested that MDA, a structurally related positive, and in 2017, MDMA was granted “breakthrough compound, was neurotoxic. At the time, there was little data on therapy” status by the FDA. This designation helps to expedite the safety of MDMA, and thus, the DEA decided to place it on the review and potential approval process for promising the Schedule I list in 1985 largely based on its structural therapeutics. Phase III clinical trials are currently being similarity to MDA.261 This decision was protested by a large planned, and if the results of those trials warrant approval by number of scientists and therapists, and challenged in court, the FDA, a bona fide accepted medical use for MDMA will but ultimately, MDMA was permanently placed on the have been established. This would necessitate the removal of Schedule I list in 1988. MDMA from the Schedule I list, a regulatory change that could In a 2002 paper published in Science, Ricaurte and co- have profound implications for the field of psychedelic workers described experiments performed in nonhuman medicine. Schedule I status has severely hampered access to primates demonstrating severe dopaminergic (and to a lesser psychedelics for research purposes. In sum, this trajectory is extent serotonergic) neurotoxicity of MDMA.262 These perhaps why MDMA is the most influential compound for the authors suggested that MDMA might put users at risk for future of psychedelic research. However, MDMA is also a developing Parkinson’s disease. The results of the study were highly divisive compound having the potential to swing public rapidly disseminated by the popular media, leading to the opinion against general use of psychedelics in medicine. widespread public belief that administration of “recreational Since 2012, there has been an upswing in the numbers of doses” of MDMA (3 doses of 2 mg/kg spaced over 6 h) could songs and pop culture references to “molly,” a trend that have major health consequences. When Ricaurte and co- parallels that seen for LSD in the 1960s and 1970s. Extensive workers could not reproduce their results, they retracted their proselytizing about the nonmedical uses of LSD contributed to

2419 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review the creation of the Controlled Substance Act of 1970. This (11) Baker, L. E., Broadbent, J., Michael, E. K., Matthews, P. K., legislation has been a huge barrier to legitimate scientific Metosh, C. A., Saunders, R. B., West, W. B., and Appel, J. B. (1995) research on the effects of these drugs and led to the first “Dark Assessment of the discriminative stimulus effects of the optical ” fi  isomers of ecstasy (3,4-methylenedioxymethamphetamine; MDMA). Age for the eld the period of time from roughly 1970 to − 1994 when relatively little psychedelic research was conducted. Behav. Pharmacol. 6, 263 275. (12) Kamien, J. B., Johanson, C. E., Schuster, C. R., and Woolverton, If public discourse on MDMA takes a similar course to that of W. L. (1986) The effects of (±)-methylenedioxymethamphetamine LSD, we may be doomed to repeat the mistakes of the past. and (±)-methylenedioxyamphetamine in monkeys trained to This would be unfortunate as MDMA is an important discriminate (+)-amphetamine from saline. Drug Alcohol Depend. 18, neurochemical tool for elucidating the neural mechanisms of 139−147. social behaviors and empathy, and it has the potential to offer (13) Anderson, G. M., III, Braun, G., Braun, U., Nichols, D. E., and real relief to people suffering from PTSD and other anxiety Shulgin, A. T. (1978) Absolute configuration and psychotomimetic disorders. However, because of its history and neurotoxic activity. NIDA Res. Monogr. Ser. 22,8−15. potential, MDMA may never achieve clinical and/or societal (14) Shulgin, A. T., and Nichols, D. E. (1978) Characterization of acceptance. Perhaps the true potential of MDMA lies in its use three new psychotomimetics, in The Psychopharmacology of − as a lead structure for the development of safer and more Hallucinogens, Pergamon Press, New York), pp 74 83. efficacious alternatives. (15) Peroutka, S. J., Newman, H., and Harris, H. (1988) Subjective effects of 3,4-methylenedioxymethamphetamine in recreational users. − ■ AUTHOR INFORMATION Neuropsychopharmacology 1, 273 277. (16) Shulgin, A., and Shulgin, A. T. (1991) PiHKAL: A Chemical ORCID Love Story, 1st ed., Transform Press, Berkeley, CA. Anne M. Andrews: 0000-0002-1961-4833 (17) Liechti, M. E., and Vollenweider, F. X. (2001) Which David E. Olson: 0000-0002-4517-0543 neuroreceptors mediate the subjective effects of MDMA in humans? A summary of mechanistic studies. Hum. Psychopharmacol. 16, 589− Author Contributions 598. D.E.O. conceived the structure of the Review. L.E.D. and (18) Harris, D. S., Baggott, M., Mendelson, J. H., Mendelson, J. E., D.E.O. wrote the manuscript with input from A.M.A. All and Jones, R. T. (2002) Subjective and hormonal effects of 3,4- authors edited the manuscript. methylenedioxymethamphetamine (MDMA) in humans. Psychophar- Funding macology (Berlin) 162, 396−405. This work was funded in part by a UC Davis Science (19) Vizeli, P., and Liechti, M. E. (2017) Safety pharmacology of acute MDMA administration in healthy subjects. J. Psychopharmacol. Translation and Innovative Research (STAIR) Grant and the − National Institute on Drug Abuse (DA045550). 31, 576 588. (20) Bershad, A. K., Miller, M. A., Baggott, M. J., and de Wit, H. Notes (2016) The effects of MDMA on socioemotional processing: does The authors declare no competing financial interest. MDMA differ from other stimulants? J. Psychopharmacol. 30, 1248− 1258. ■ REFERENCES (21) Kamilar-Britt, P., and Bedi, G. (2015) The prosocial effects of (1) Shulgin, A. T. (1986) The background and chemistry of MDMA. 3,4-methylenedioxymethamphetamine (MDMA): controlled studies − J. Psychoact. Drugs 18, 291−304. in humans and laboratory animals. Neurosci. Biobehav. Rev. 57, 433 (2) Palamar, J. J. (2018) What’s in a name? Correlates of ecstasy 446. users knowing or agreeing that molly is ecstasy/MDMA. J. Psychoact. (22) Cami, J., Farre, M., Mas, M., Roset, P. N., Poudevida, S., Mas, Drugs 50,88−93. A., San, L., and de la Torre, R. (2000) Human pharmacology of 3,4 “ ” (3) Calculated using advanced chemistry development (ACD/ methylenedioxymethamphetamine ( ecstasy ): psychomotor perform- − Laboratories) Software V11.02 via SciFinder Scholar. ance and subjective effects. J. Clin. Psychopharmacol. 20, 455 466. (4) Pajouhesh, H., and Lenz, G. R. (2005) Medicinal chemical (23) Kolbrich, E. A., Goodwin, R. S., Gorelick, D. A., Hayes, R. J., properties of successful central nervous system drugs. NeuroRx 2, Stein, E. A., and Huestis, M. A. (2008) Physiological and subjective 541−553. responses to controlled oral 3,4-methylenedioxymethamphetamine (5) Oberlender, R., and Nichols, D. E. (1988) Drug discrimination administration. J. Clin. Psychopharmacol. 28, 432−440. studies with MDMA and amphetamine. Psychopharmacology (Berlin) (24) Vollenweider, F. X., Gamma, A., Liechti, M. E., and Huber, T. 95,71−76. (1998) Psychological and cardiovascular effects and short-term (6) Nichols, D. E., Hoffman, A. J., Oberlender, R. A., Jacob, P., and sequelae of MDMA (“ecstasy”) in MDMA-naive healthy volunteers. Shulgin, A. T. (1986) Derivatives of 1-(1,3-benzodioxol-5-yl)-2- Neuropsychopharmacology 19, 241−251. butanamine: representatives of a novel therapeutic class. J. Med. Chem. (25) Liechti, M. E., Saur, M. R., Gamma, A., Hell, D., and 29, 2009−2015. Vollenweider, F. X. (2000) Psychological and physiological effects of (7) Glennon, R. A., Young, R., Rosecrans, J. A., and Anderson, G. M. MDMA (“Ecstasy”) after pretreatment with the 5HT(2) antagonist (1982) Discriminative stimulus properties of MDA analogs. Biol. ketanserin in healthy humans. Neuropsychopharmacology 23, 396−404. Psychiatry 17, 807−814. (26) Nichols, D. E. (2004) Hallucinogens. Pharmacol. Ther. 101, (8) Mori, T., Uzawa, N., Kazawa, H., Watanabe, H., Mochizuki, A., 131−181. Shibasaki, M., Yoshizawa, K., Higashiyama, K., and Suzuki, T. (2014) (27) Nichols, D. E. (2016) Psychedelics. Pharmacol. Rev. 68, 264− Differential substitution for the discriminative stimulus effects of 3,4- 355. methylenedioxymethamphetamine and methylphenidate in rats. J. (28) Hasler, F., Studerus, E., Lindner, K., Ludewig, S., and Pharmacol. Exp. Ther. 350, 403−411. Vollenweider, F. (2009) Investigation of serotonin1A receptor (9) Broadbear, J. H., Tunstall, B., and Beringer, K. (2011) Examining function in the human psychopharmacology of MDMA. J. the role of oxytocin in the interoceptive effects of 3,4-methylenediox- Psychopharmacol. 23, 923−935. ymethamphetamine (MDMA,‘ecstasy’) using a drug discrimination (29) Kuypers, K. P., de la Torre, R., Farre, M., YuberoLahoz, S., paradigm in the rat. Addiction Biology 16, 202−214. Dziobek, I., Van den Bos, W., and Ramaekers, J. G. (2014) No (10) Harper, D. N., Crowther, A., and Schenk, S. (2011) A evidence that MDMA induced enhancement of emotional empathy is Comparison of MDMA and amphetamine in the drug discrimination related to peripheral oxytocin levels or 5HT1a receptor activation. paradigm. Open Addict. J. 4,22−23. PLoS One 9, e100719.

2420 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

(30) Van Wel, J. H., Kuypers, K. P., Theunissen, E. L., Bosker, W. (47) Palamar, J. J., Mauro, P. M., Han, B. H., and Martins, S. S. M., Bakker, K., and Ramaekers, J. G. (2012) Effects of acute MDMA (2017) Shifting characteristics of ecstasy users ages 12−34 in the intoxication on mood and impulsivity: role of the 5HT2 and 5HT1 United States, 2007−2014. Drug Alcohol Depend. 181,20−24. receptors. PLoS One 7, e40187. (48) Heifets, B. D., and Malenka, R. C. (2016) MDMA as a probe (31) Farre, M., Abanades, S., Roset, P. N., Peiro, A. M., Torrens, M., and treatment for social behaviors. Cell 166, 269−272. O’Mathuna, B., Segura, M., and de la Torre, R. (2007) (49) Karch, S. B. (2011) A historical review of MDMA. Open − Pharmacological interaction between 3,4-methylenedioxymetham- Forensic Sci. J. 4,20 24. phetamine (ecstasy) and paroxetine: pharmacological effects and (50) Renton, R. J., Cowie, J. S., and Oon, M. C. (1993) A study of pharmacokinetics. J. Pharmacol. Exp. Ther. 323, 954−962. the precursors, intermediates and reaction by-products in the (32) Liechti, M. E., Baumann, C., Gamma, A., and Vollenweider, F. synthesis of 3,4-methylenedioxymethylamphetamine and its applica- − X. (2000) Acute psychological effects of 3,4-methylenedioxymetham- tion to forensic drug analysis. Forensic Sci. Int. 60, 189 202. phetamine (MDMA, “Ecstasy”) are attenuated by the serotonin (51) Biniecki, S., and Krajewski, E. (1960) Preparation of dl-(3,4- − methylenedioxyphenyl)-2-(methylamino)propane and dl-(3,4- dime- uptake inhibitor citalopram. Neuropsychopharmacology 22, 513 521. − (33) Tancer, M., and Johanson, C. E. (2007) The effects of thoxyphenyl)-2-(methylamino)propane. Acta Polym. Pharm. 17, 421 425. on the subjective and physiological effects of 3,4- (52) Cox, M., and Klass, G. (2006) Synthesis by-products from the methylenedioxymethamphetamine (MDMA) in humans. Psychophar- Wacker oxidation of safrole in methanol using rho-benzoquinone and macology 189, 565−573. palladium chloride. Forensic Sci. Int. 164, 138−147. (34) Hysek, C. M., Simmler, L. D., Ineichen, M., Grouzmann, E., (53) Braun, U., Shulgin, A. T., and Braun, G. (1980) Centrally active Hoener, M. C., Brenneisen, R., Huwyler, J., and Liechti, M. E. (2011) N-substituted analogs of 3,4-methylenedioxyphenylisopropylamine Thenorepinephrinetransporterinhibitor reduces − “ ” (3,4-methylenedioxyamphetamine). J. Pharm. Sci. 69, 192 195. stimulant effects of MDMA ( ecstasy ) in humans. Clin. Pharmacol. (54) Heather, E., Shimmon, R., and McDonagh, A. M. (2015) − Ther. 90, 246 255. Organic impurity profiling of 3,4-methylenedioxymethamphetamine (35) Hysek, C. M., Simmler, L. D., Nicola, V., Vischer, N., Donzelli, (MDMA) synthesised from catechol. Forensic Sci. Int. 248, 140−147. ̈ ̈ M., Krahenbuhl, S., Grouzmann, E., Huwyler, J., Hoener, M. C., and (55) Parker, M., Marona-Lewicka, D., Kurrasch, D., Shulgin, A. T., Liechti, M. E. (2012) Duloxetine inhibits effects of MDMA and Nichols, D. E. (1998) Synthesis and pharmacological evaluation (“ecstasy”) in vitro and in humans in a randomized placebo- of ring-methylated derivatives of 3,4-(methylenedioxy)amphetamine controlled laboratory study. PLoS One 7, e36476. (MDA). J. J. Med. Chem. 41, 1001−1005. (36) Liechti, M. E., Gamma, A., and Vollenweider, F. X. (2001) (56) Pizarro, N., de la Torre, R., Farre,́ M., Segura, J., Llebaria, A., Gender differences in the subjective effects of MDMA. Psychophar- and Joglar, J. (2002) Synthesis and capillary electrophoretic analysis macology (Berlin) 154, 161−168. of enantiomerically enriched reference standards of MDMA and its (37) Vizeli, P., and Liechti, M. E. (2017) Safety pharmacology of main metabolites. Bioorg. Med. Chem. 10, 1085−1092. acute MDMA administration in healthy subjects. J. J. Psychopharma- (57) Llabres,́ S., García-Rates,́ S., Cristobal-Lecina,́ E., Riera, A., col. 31, 576−588. Borrell, J. I., Camarasa, J., Pubill, D., Luque, F. J., and Escubedo, E. (38) Dolder, P. C., Muller, F., Schmid, Y., Borgwardt, S. J., and (2014) Molecular basis of the selective binding of MDMA Liechti, M. E. (2018) Direct comparison of the acute subjective, enantiomers to the alpha4beta2 nicotinic receptor subtype: synthesis, emotional, autonomic, and endocrine effects of MDMA, methyl- pharmacological evaluation and mechanistic studies. Eur. J. Med. phenidate, and in healthy subjects. Psychopharmacology Chem. 81,35−46. (Berlin) 235, 467−479. (58) Nenajdenko, V. G., Karpov, A. S., and Balenkova, E. S. (2001) (39) Schmid, Y., Hysek, C. M., Simmler, L. D., Crockett, M. J., A new convenient approach to chiral -aryl(heteroaryl)alkylamines. − Quednow, B. B., and Liechti, M. E. (2014) Differential effects of Tetrahedron: Asymmetry 12, 2517 2527. MDMA and methylphenidate on social cognition. J. Psychopharmacol. (59) Huot, P., Johnston, T. H., Lewis, K. D., Koprich, J. B., Reyes, 28, 847−856. M. G., Fox, S. H., Piggott, M. J., and Brotchie, J. M. (2011) (40) Yensen, R., Metzner, R., Shakespeare, W., and Nichols, D. E. Characterization of 3,4-methylenedioxymethamphetamine (MDMA) (1993) The great entactogen - empathogen debate. Newsl. Multidiscip. enantiomers in vitro and in the MPTP-lesioned primate: R-MDMA − reduces severity of dyskinesia, whereas S-MDMA extends duration of Assoc. Psychedelic Studies MAPS 4,47 49. − (41) Nichols, D. E. (1986) Differences between the mechanism of ON-time. J. Neurosci. 31, 7190 7198. action of MDMA, MBDB, and the classic hallucinogens. Identification (60) Iversen, L., Gibbons, S., Treble, R., Setola, V., Huang, X. P., and Roth, B. L. (2013) Neurochemical profiles of some novel psycho- of a new therapeutic class: entactogens. J. Psychoact. Drugs 18, 305− active substances. Eur. J. Pharmacol. 700, 147−151. 313. (61) Jones, D. C., Lau, S. S., and Monks, T. J. (2004) Thioether (42) Dolder, P. C., Schmid, Y., Mueller, F., Borgwardt, S., and metabolites of 3,4-methylenedioxyamphetamine and 3,4-methylene- Liechti, M. E. (2016) LSD acutely impairs fear recognition and dioxymethamphetamine inhibit human serotonin transporter enhances emotional empathy and sociality. Neuropsychopharmacology − (hSERT) function and simultaneously stimulate dopamine uptake 41, 2638 2646. into hSERT-expressing SK-N-MC cells. J. Pharmacol. Exp. Ther. 311, (43) Schmid, Y., Enzler, F., Gasser, P., Grouzmann, E., Preller, K. H., − ̈ 298 306. Vollenweider, F. X., Brenneisen, R., Muller, F., Borgwardt, S., and (62) Simmler, L., Buser, T., Donzelli, M., Schramm, Y., Dieu, L. H., Liechti, M. E. (2015) Acute effects of lysergic acid diethylamide in Huwyler, J., et al. (2013) Pharmacological characterization of designer − healthy subjects. Biol. Psychiatry 78, 544 553. cathinones in vitro. Br. J. Pharmacol. 168, 458−470. (44) Pentney, A. R. (2001) An exploration of the history and (63) Han, D. D., and Gu, H. H. (2006) Comparison of the controversies surrounding MDMA and MDA. J. Psychoact. Drugs 33, monoamine transporters from human and mouse in their sensitivities 213−221. to psychostimulant drugs. BMC Pharmacol. 6,6. (45) SAMHSA (2015) National Survey on Drug Use and Health, (64) Sandtner, W., Stockner, T., Hasenhuetl, P. S., Partilla, J. S., 2002−2014, Substance Abuse and Mental Health Services Admin- Seddik, A., Zhang, Y. W., Cao, J., Holy, M., Steinkellner, T., Rudnick, istration, Rockville, MD, https://www.samhsa.gov/data/sites/ G., Baumann, M. H., Ecker, G. F., Newman, A. H., and Sitte, H. H. default/files/NSDUH-DetTabs2014/NSDUH-DetTabs2014.pdf. (2016) Binding mode selection determines the action of ecstasy (46) Wu, P., Liu, X., Pham, T. H., Jin, J., Fan, B., and Jin, Z. (2010) homologs at monoamine transporters. Mol. Pharmacol. 89, 165−175. Ecstasy use among US adolescents from 1999 to 2008. Drug Alcohol (65) Rothman, R. B., Baumann, M. H., Dersch, C. M., Romero, D. Depend. 112,33−38. V., Rice, K. C., Carroll, F. I., and Partilla, J. S. (2001) Amphetamine-

2421 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review type central nervous system stimulants release norepinephrine more Bonini, J. A., Pathirana, S., Boyle, N., Pu, X., Kouranova, E., Lichtblau, potently than they release dopamine and serotonin. Synapse 39,32− H., Ochoa, F. Y., Branchek, T. A., and Gerald, C. (2001) Trace 41. amines: Identification of a family of mammalian G protein-coupled (66) Bogen, I. L., Haug, K. H., Myhre, O., and Fonnum, F. (2003) receptors. Proc. Natl. Acad. Sci. U. S. A. 98, 8966−8971. Short- and long-term effects of MDMA (“ecstasy”) on synaptosomal (82) Accorroni, A., and Zucchi, R. (2016) Trace Amine-Associated and vesicular uptake of neurotransmitters in vitro and ex vivo. Receptors: Ligands and putative role in the central nervous system Neurochem. Int. 43, 393−400. (Farooqui, A., Ed.) Academic Press, San Diego, CA, pp 151−164. (67) Fitzgerald, J. L., and Reid, J. J. (1990) Effects of (83) Miller, G. M. (2011) The emerging role of trace amine- methylenedioxymethamphetamine on the release of monoamines associated receptor 1 in the functional regulation of monoamine from rat brain slices. Eur. J. Pharmacol. 191, 217−220. transporters and dopaminergic activity. J. Neurochem. 116, 164−176. (68) Rudnick, G., and Wall, S. C. (1992) The molecular mechanism (84) Simmler, L. D., Buchy, D., Chaboz, S., Hoener, M. C., and of “ecstasy” [3,4-methylenedioxy-methamphetamine (MDMA)]: Liechti, M. E. (2016) In vitro characterization of psychoactive serotonin transporters are targets for MDMA-induced serotonin substances at rat, mouse, and human trace amineassociated receptor release. Proc. Natl. Acad. Sci. U. S. A. 89, 1817−1821. 1. J. Pharmacol. Exp. Ther. 357, 134−144. (69) Mlinar, B., and Corradetti, R. (2003) Endogenous 5-HT, (85) Brammer, M. K., Gilmore, D. L., and Matsumoto, R. R. (2006) released by MDMA through serotonin transporter- and secretory Interactions between 3,4-methylenedioxymethamphetamine and σ1 vesicle-dependent mechanisms, reduces hippocampal excitatory Receptors. Eur. J. Pharmacol. 553, 141−145. synaptic transmission by preferential activation of 5-HT1B receptors (86) Cobos, E. J., Entrena, J. M., Nieto, F. R., Cendan, C. M., and located on CA1 pyramidal neurons. Eur. J. Neurosci. 18, 1559−1571. Del Pozo, E. (2008) Pharmacology and therapeutic potential of (70) Schuldiner, S., Steiner-Mordoch, S., Yelin, R., Wall, S.C., and sigma(1) receptor ligands. Curr. Neuropharmacol. 6, 344−366. Rudnick, G. (1993) Amphetamine derivatives interact with both (87) Hayashi, T. (2015) Sigma-1 receptor: the novel intracellular plasma membrane and secretory vesicle transporters. target of neuropsychotherapeutic drugs. J. Pharmacol. Sci. 127,2−5. Mol. Pharmacol. 44, 1227−1231. (88) Maurice, T., and Su, T.-P. (2009) The pharmacology of sigma- (71) Sulzer, D., and Rayport, S. (1990) Amphetamine and other 1 receptors. Pharmacol. Ther. 124, 195−206. psychostimulants reduce pH gradients in midbrain dopaminergic (89) Villard, V., Meunier, J., Chevallier, N., and Maurice, T. (2011) neurons and chromaffin granules: a mechanism of action. Neuron 5, Pharmacological interaction with the sigma1 (σ1)-receptor in the 797−808. acute behavioral effects of . J. Pharmacol. Sci. 115, (72) Leonardi, E. T., and Azmitia, E. C. (1994) MDMA (ecstasy) 279−292. inhibition of MAO type A and type B, comparisons with (90) Stahl, S. M. (2008) The sigma enigma: can sigma receptors and fluoxetine (Prozac). Neuropsychopharmacology 10, 231−238. provide a novel target for disorders of mood and cognition? J. Clin. (73) Kittler, K., Lau, T., and Schloss, P. (2010) Antagonists and Psychiatry 69, 1673−1674. substrates differentially regulate serotonin transporter cell surface (91) Grob, C. S., Poland, R. E., Chang, L., and Ernst, T. (1995) expression in serotonergic neurons. Eur. J. Pharmacol. 629,63−67. Psychobiologic effects of 3,4-methylenedioxymethamphetamine in (74) Gołembiowska, K., Jurczak, A., Kaminska,́ K., Noworyta- humans: methodological considerations and preliminary observations. Sokołowska, K., and Gorska,́ A. (2016) Effect of some psychoactive Behav. Brain Res. 73, 103−107. drugs used as ‘legal highs’ on brain neurotransmitters. Neurotoxic. Res. (92) Mas, M., Farre, M., de la Torre, R., Roset, P. N., Ortuno, J., 29, 394−407. Segura, J., and Cami, J. (1999) Cardiovascular and neuroendocrine (75) Iravani, M. M., Asari, D., Patel, J., Wieczorek, W. J., and Kruk, effects and pharmacokinetics of 3,4-methylenedioxymethamphet- Z. L. (2000) Direct effects of 3,4-methylenedioxymethamphetamine amine in humans. J. Pharmacol. Exp. Ther. 290,1−10. (MDMA) on serotonin or dopamine release and uptake in the (93) Dumont, G. J., Sweep, F. C., van der Steen, R., Hermsen, R., caudate putamen, nucleus accumbens, substantia nigra pars reticulata, Donders, A. R., Touw, D. J., van Gerven, J. M., Buitelaar, J. K., and and the dorsal raphénucleus slices. Synapse 36, 275−285. Verkes, R. J. (2009) Increased oxytocin concentrations and prosocial (76) Battaglia, G., Brooks, B. P., Kulsakdinun, C., and De Souza, E. feelings in humans after ecstasy (3,4-methylenedioxymethamphet- B. (1988) Pharmacologic profile of MDMA (3,4-methylenediox- amine) administration. Soc. Neurosci. 4, 359−366. ymethamphetamine) at various brain recognition sites. Eur. J. (94) Seibert, J., Hysek, C. M., Penno, C. A., Schmid, Y., Kratschmar, Pharmacol. 149, 159−163. D. V., Liechti, M. E., and Odermatt, A. (2014) Acute effects of 3,4- (77) Setola, V., Hufeisen, S. J., Grande-Allen, K. J., Vesely, I., methylenedioxymethamphetamine and methylphenidate on circulat- Glennon, R. A., Blough, B., Rothman, R. B., and Roth, B. L. (2003) ing steroid levels in healthy subjects. Neuroendocrinology 100,17−25. 3,4-methylenedioxymethamphetamine (MDMA, “Ecstasy”) induces (95) Henry, J. A., Fallon, J. K., Kicman, A. T., Hutt, A. J., Cowan, D. fenfluramine-like proliferative actions on human cardiac valvular A., and Forsling, M. (1998) Lowdose MDMA (“ecstasy”) induces interstitial cells in vitro. Mol. Pharmacol. 63, 1223−1229. vasopressin secretion. Lancet 351, 1784. (78) Rickli, A., Hoener, M. C., and Liechti, M. E. (2015) (96) Wolff, K., Tsapakis, E. M., Winstock, A. R., Hartley, D., Holt, Monoamine transporter and receptor interaction profiles of novel D., Forsling, M. L., and Aitchison, K. J. (2006) Vasopressin and psychoactive substances: parahalogenated amphetamines and pyrova- oxytocin secretion in response to the consumption of ecstasy in a lerone cathinones. Eur. Neuropsychopharmacol. 25, 365−376. clubbing population. J. Psychopharmacol. 20, 400−410. (79) Doly, S., Valjent, E., Setola, V., Callebert, J., Herve,́ D., Launay, (97) Lefebvre, H., Contesse, V., Delarue, C., Feuilloley, M., Hery, F., J. M., and Maroteaux, L. (2008) Serotonin 5-HT2B receptors are Grise, P., Raynaud, G., Verhofstad, A. A., Wolf, L. M., and Vaudry, H. required for 3,4-methylenedioxymethamphetamine-induced hyper- (1992) Serotonin-induced stimulation of cortisol secretion from locomotion and 5-HT release in vivo and in vitro. J. Neurosci. 28, human adrenocortical tissue is mediated through activation of a 2933−2940. serotonin-4 receptor subtype. Neuroscience 47, 999−1007. (80) Bunzow, J. R., Sonders, M. S., Arttamangkul, S., Harrison, L. (98) Prescott, R. W., Kendall-Taylor, P., Weightman, D. R., Watson, M., Zhang, G., Quigley, D. I., Darland, T., Suchland, K. L., M. J., and Ratcliffe, W. A. (1984) The effect of ketanserin, a specific Pasumamula, S., Kennedy, J. L., Olson, S. B., Magenis, R. E., serotonin antagonist on the PRL, GH, ACTH, and cortisol responses Amara, S. G., and Grandy, D. K. (2001) Amphetamine, 3,4- to hypoglycaemia in normal subjects. Clin. Endocrinol. (Oxford, U. K.) methylenedioxymethamphetamine, lysergic acid diethylamide, and 20, 137−142. metabolites of the catecholamine neurotransmitters are agonists of a (99) Thompson, M. R., Callaghan, P. D., Hunt, G. E., Cornish, J. L., rat trace amine receptor. Mol. Pharmacol. 60, 1181−1188. and McGregor, I. S. (2007) A role for oxytocin and 5-HT(1A) (81)Borowsky,B.,Adham,N.,Jones,K.A.,Raddatz,R., receptors in the prosocial effects of 3,4-methylenedioxymethamphet- Artymyshyn, R., Ogozalek, K. L., Durkin, M. M., Lakhlani, P. P., amine (“ecstasy”). Neuroscience 146, 509−514.

2422 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

(100) Parrott, A. C. (2016) Oxytocin, cortisol and 3,4- 3,4-methylenedioxymethamphetamine in rats: involvement of the methylenedioxymethamphetamine: neurohormonal aspects of recrea- V1A receptor. Neuropsychopharmacology 38, 2249−2259. tional ‘ecstasy’. Behav. Pharmacol. 27, 649−658. (118) Young, M. B., Andero, R., Ressler, K. J., and Howell, L. L. (101) Kuypers, K. P. C., Dolder, P. C., Ramaekers, J. G., and Liechti, (2015) 3,4-Methylenedioxymethamphetamine facilitates fear extinc- M. E. (2017) Multifaceted empathy of healthy volunteers after single tion learning. Transl. Psychiatry 5, e634. doses of MDMA: a pooled sample of placebo-controlled studies. J. (119) Catlow, B. J., Song, S., Paredes, D. A., Kirstein, C. L., and Psychopharmacol. 31, 589−598. Sanchez-Ramos, J. (2013) Effects of psilocybin on hippocampal (102) Fox, M. A., Andrews, A. M., Wendland, J. R., Lesch, K. P., neurogenesis and extinction of trace fear conditioning. Exp. Brain Res. Holmes, A., and Murphy, D. L. (2007) A pharmacological analysis of 228, 481−491. mice with a targeted disruption of the serotonin transporter. (120) Cameron, L. P., Benson, C. J., Dunlap, L. E., and Olson, D. E. Psychopharmacology (Berlin) 195, 147−166. (2018) Effects of N,N-dimethyltryptamine (DMT) on rat behaviors (103) Spanos, L. J., and Yamamoto, B. K. (1989) Acute and relevant to anxiety and depression. ACS Chem. Neurosci., subchronic effects of methylenedioxymethamphetamine DOI: 10.1021/acschemneuro.8b00134. [(±)MDMA] on locomotion and serotonin syndrome behavior in (121) Young, M. B., Norrholm, S. D., Khoury, L. M., Jovanovic, T., the rat. Pharmacol., Biochem. Behav. 32, 835−840. Rauch, S. A. M., Reiff, C. M., Dunlop, B. W., Rothbaum, B. O., and (104) Kalivas, P. W., Duffy, P., and White, S. R. (1998) MDMA Howell, L. L. (2017) Inhibition of serotonin transporters disrupts the elicits behavioral and neurochemical sensitization in rats. Neuro- enhancement of fear memory extinction by 3,4-methylenedioxyme- − psychopharmacology 18, 469 479. thamphetamine (MDMA). Psychopharmacology 234, 2883−2895. (105) Scearce-Levie, K., Viswanathan, S. S., and Hen, R. (1999) (122) Nyberg, F. (2014) Structural plasticity of the brain to Locomotor response to MDMA is attenuated in knockout mice psychostimulant use. Neuropharmacology 87, 115−124. − lacking the 5-HT1B receptor. Psychopharmacology (Berlin) 141, 154 (123) Martinez-Turrillas, R., Moyano, S., Del Rio, J., and Frechilla, 161. D. (2006) Differential effects of 3,4-methylenedioxymethamphet- (106) Kehne, J. H., Ketteler, H. J., McCloskey, T. C., Sullivan, C. K., amine (MDMA, “ecstasy”) on BDNF mRNA expression in rat frontal Dudley, M. W., and Schmidt, C. J. (1996) Effects of the selective 5- cortex and hippocampus. Neurosci. Lett. 402, 126−130. HT2A receptor antagonist MDL 100,907 on MDMA-induced (124) Hemmerle, A. M., Dickerson, J. W., Herring, N. R., Schaefer, − locomotor stimulation in rats. Neuropsychopharmacology 15, 116 124. T. L., Vorhees, C. V., Williams, M. T., and Seroogy, K. B. (2012) (107) Risbrough, V. B., Masten, V. L., Caldwell, S., Paulus, M. P., ±)3,4-Methylenedioxymethamphetamine (“Ecstasy”) treatment mod- Low, M. J., and Geyer, M. A. (2006) Differential contributions of ulates expression of neurotrophins and their receptors in multiple dopamine D1, D2, and D3 receptors to MDMA-induced effects on regions of adult rat brain. J. Comp. Neurol. 520, 2459−2474. locomotor behavior patterns in mice. Neuropsychopharmacology 31, − (125) Abad, S., Camarasa, J., Pubill, D., Camins, A., and Escubedo, 2349 2358. E. (2016) Adaptive plasticity in the hippocampus of young mice (108) Callaway, C. W., Wing, L. L., and Geyer, M. A. (1990) intermittently exposed to MDMA could be the origin of memory Serotonin release contributes to the locomotor stimulant effects of deficits. Mol. Neurobiol. 53, 7271−7283. 3,4-methylenedioxymethamphetamine in rats. J. Pharmacol. Exp. Ther. (126) Abad, S., Fole, A., del Olmo, N., Pubill, D., Pallas,̀ M., Junyent, 254, 456−464. F., Camarasa, J., Camins, A., and Escubedo, E. (2014) MDMA (109) Bengel, D., Murphy, D. L., Andrews, A. M., Wichems, C. H., enhances hippocampal-dependent learning and memory under Feltner, D., Heils, A., Mossner, R., Westphal, H., and Lesch, K. P. restrictive conditions, and modifies hippocampal spine density. (1998) Altered brain serotonin homeostasis and locomotor Psychopharmacology (Berlin) 231, 863−874. insensitivity to 3, 4-methylenedioxymethamphetamine (“ecstasy”)in (127) Kaizaki, A., Tanaka, S., Tsujikawa, K., Numazawa, S., and serotonin transporter-deficient mice. Mol. Pharmacol. 53, 649−655. Yoshida, T. (2010) Recreational drugs, 3,4-Methylenedioxymeth- (110) Lin, H. Q., Burden, P. M., Christie, M. J., and Johnston, G. A. (1999) The anxiogenic-like and anxiolytic-like effects of MDMA on amphetamine(MDMA), 3,4-methylenedioxyamphetamine (MDA) and , inhibit neurite outgrowth in PC12 cells. J. mice in the elevated plus-maze: a comparison with amphetamine. − Pharmacol., Biochem. Behav. 62, 403−408. Toxicol. Sci. 35, 375 381. (111) Navarro, J. F., and Maldonado, E. (2002) Acute and (128) Ly, C., Greb, A. C., Cameron, L. P., Wong, J. M., Barragan, E. subchronic effects of MDMA (“ecstasy”) on anxiety in male mice V., Wilson, P. C., Burbach, K. F., Soltanzadeh Zarandi, S., Sood, A., tested in the elevated plus-maze. Prog. Neuro-Psychopharmacol. Biol. Paddy, M. R., Duim, W. C., Dennis, M. Y., McAllister, A. K., Ori- Psychiatry 26, 1151−1154. McKenney, K. M., Gray, J. A., and Olson, D. E. (2018) Psychedelics (112) Ho, Y. J., Pawlak, C. R., Guo, L., and Schwarting, R. K. (2004) promote structural and functional neural plasticity. Cell Rep. 23, − Acute and long-term consequences of single MDMA administration 3170 3182. in relation to individual anxiety levels in the rat. Behav. Brain Res. 149, (129) Kreth, K., Kovar, K., Schwab, M., and Zanger, U. M. (2000) 135−144. Identification of the human cytochromes P450 involved in the “ ” (113) Maldonado, E., and Navarro, J. F. (2000) Effects of 3,4- oxidative metabolism of Ecstasy -related designer drugs. Biochem. − methylenedioxy-methamphetamine (MDMA) on anxiety in mice Pharmacol. 59, 1563 1571. tested in the light-dark box. Prog. Neuro-Psychopharmacol. Biol. (130) Shima, N., Kamata, H., Katagi, M., Tsuchihashi, H., Sakuma, Psychiatry 24, 463−472. T., and Nemoto, N. (2007) Direct determination of glucuronide and (114) Morley, K. C., and McGregor, I. S. (2000) (±)-3,4- sulfate of 4-hydroxy-3-methoxymethamphetamine, the main metabo- Methylenedioxymethamphetamine (MDMA, ‘Ecstasy’)increases lite of MDMA, in human urine. J. Chromatogr. B: Anal. Technol. social interaction in rats. Eur. J. Pharmacol. 408,41−49. Biomed. Life Sci. 857, 123−129. (115) Ramos, L., Hicks, C., Caminer, A., Goodwin, J., and (131) Vizeli, P., Schmid, Y., Prestin, K., Meyer zu Schwabedissen, H. McGregor, I. S. (2015) Oxytocin and MDMA (’Ecstasy’) enhance E., and Liechti, M. E. (2017) Pharmacogenetics of ecstasy: CYP1A2, social reward in rats. Psychopharmacology (Berlin) 232, 2631−2641. CYP2C19, and CYP2B6 polymorphisms moderate pharmacokinetics (116) Brown, A., Brown, T. B., Calabrese, A., Ellis, D., Puhalo, N., of MDMA in healthy subjects. Eur. Neuropsychopharmacol. 27, 232− Ralph, M., and Watson, L. (2010) Triazole oxytocin antagonists: 238. Identification of an aryloxyazetidine replacement for a biaryl (132) Schmid, Y., Vizeli, P., Hysek, C. M., Prestin, K., Meyer zu substituent. Bioorg. Med. Chem. Lett. 20, 516−520. Schwabedissen, H. E., and Liechti, M. E. (2016) CYP2D6 function (117) Ramos, L., Hicks, C., Kevin, R., Caminer, A., Narlawar, R., moderates the pharmacokinetics and pharmacodynamics of MDMA Kassiou, M., and McGregor, I. S. (2013) Acute prosocial effects of in a controlled study in healthy subjects. Pharmacogenet. Genomics 26, oxytocin and vasopressin when given alone or in combination with 397−401.

2423 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

(133) de la Torre, R., Farre,́ M., Roset, P. N., Lopez, C. H., Mas, M., contribute to the acute effects of MDMA in humans. J. Clin. Ortuño, J., Menoyo, E., Pizarro, N., Segura, J., and Cami, J. (2000) Psychopharmacol. 33, 658−666. Pharmacology of MDMA in humans. Ann. N. Y. Acad. Sci. 914, 225− (150) Badon, L. A., Hicks, A., Lord, K., Ogden, B. A., Meleg-Smith, 237. S., and Varner, K. J. (2002) Changes in cardiovascular responsiveness (134) de la Torre, R., Farre,́ M., Ortuño, J., Mas, M., Brenneisen, R., and cardiotoxicity elicited during binge administration of Ecstasy. J. Roset,P.N.,Segura,J.,andCamí,J.(2000)Non-linear Pharmacol. Exp. Ther. 302, 898−907. pharmacokinetics of MDMA (‘ecstasy’) in humans. Br. J. Clin. (151) Furst, S. R., Fallon, S. P., Reznik, G. N., and Shah, P. K. Pharmacol. 49, 104−109. (1990) Myocardial infarction after inhalation of methamphetamine. (135) Kolbrich, E. A., Goodwin, R. S., Gorelick, D. A., Hayes, R. J., N. Engl. J. Med. 323, 1147−1148. Stein, E. A., and Huestis, M. A. (2008) Plasma pharmacokinetics of (152) Qasim, A., Townend, J., and Davies, M. (2001) Ecstasy 3,4-methylenedioxymethamphetamine after controlled oral admin- induced acute myocardial infarction. Br. Heart J. 85, e10. istration to young adults. Ther. Drug Monit. 30, 320−332. (153) Bhattacharyya, S., Schapira, A. H., Mikhailidis, D. P., and (136) Mueller, M., Peters, F. T., Maurer, H. H., McCann, U. D., and Davar, J. (2009) Drug-induced fibrotic valvular heart disease. Lancet Ricaurte, G. A. (2008) Nonlinear pharmacokinetics of (±)3,4- 374, 577−585. methylenedioxymethamphetamine (MDMA, “Ecstasy”) and its major (154) Shenouda, S. K., Varner, K. J., Carvalho, F., and Lucchesi, P. metabolites in squirrel monkeys at plasma concentrations of MDMA A. (2009) Metabolites of MDMA induce oxidative stress and that develop after typical psychoactive doses. J. Pharmacol. Exp. Ther. contractile dysfunction in adult rat left ventricular myocytes. 327,38−44. Cardiovasc. Toxicol. 9,30−38. (137) Delaforge, M., Jaouen, M., and Bouille, G. (1999) Inhibitory (155) Bilsky, E. J., Hubbell, C. L., Delconte, J. D., and Reid, L. D. metabolite complex formation of methylenedioxymethamphetamine (1991) MDMA produces a conditioned place preference and elicits with rat and human cytochrome P450. Particular involvement of ejaculation in male rats: a modulatory role for the endogenous CYP2D. Environ. Toxicol. Pharmacol. 7, 153−158. opioids. Pharmacol., Biochem. Behav. 40, 443−447. (138) Yang, J., Jamei, M., Heydari, A., Yeo, K. R., de la Torre, R., (156) Daza-Losada, M., Rodríguez-Arias, M., Aguilar, M. A., and Farre,M.,Tucker,G.T.,andRostami-Hodjegan,A.(2006)́ Miñarro, J. (2009) Acquisition and reinstatement of MDMA-induced Implications of mechanism-based inhibition of CYP2D6 for the conditioned place preference in mice pre-treated with MDMA or pharmacokinetics and toxicity of MDMA. J. Psychopharmacol. 20, cocaine during adolescence. Addict. Biol. 14, 447−456. 842−849. (157) Vidal-Infer, A., Roger-Sanchez,́ C., Daza-Losada, M., Aguilar, (139) Pizarro, N., Farre,́ M., Pujadas, M., Peiro,́ A. M., Roset, P. N., M. A., Miñarro, J., and Rodríguez-Arias, M. (2012) Role of the Joglar, J., and de la Torre, R. (2004) Stereochemical analysis of 3,4- dopaminergic system in the acquisition, expression and reinstatement methylenedioxymethamphetamine and its main metabolites in human of MDMA-induced conditioned place preference in adolescent mice. samples including the catechol-type metabolite (3,4-dihydroxyme- PLoS One 7, e43107. thamphetamine). Drug Metab. Dispos. 2, 1001−1007. (158) Schenk, S. (2009) MDMA self-administration in laboratory (140) Fallon, J. K., Kicman, A. T., Henry, J. A., Milligan, P. J., animals: a summary of the literature and proposal for future research. Cowan, D. A., and Hutt, A. J. (1999) Stereospecific analysis and Neuropsychobiology 60, 130−136. enantiomeric disposition of 3, 4-methylenedioxymethamphetamine (159) Coelho Neto, J., Faraco, R. F. P., Alves, C. F., Castro, S. M. (Ecstasy) in humans. Clin. Chem. 45, 1058−1069. M., and Machado, Y. (2018) Genuine tablets sold in Brazil (141) Steuer, A. E., Schmidhauser, C., Schmid, Y., Rickli, A., Liechti, disguised as MDMA. Forensic Sci. Int. 283,e8−12. M. E., and Kraemer, T. (2015) Chiral plasma pharmacokinetics of 3,4- (160) Saleemi, S., Pennybaker, S. J., Wooldridge, M., and Johnson, methylenedioxymethamphetamine and its phase I and II metabolites M. W. (2017) Who is ’Molly’? MDMA adulterants by product name following controlled administration to humans. Drug Metab. Dispos. and the impact of harm-reduction services at raves. J. Psychopharma- 43, 1864−1871. col. 31, 1056−1060. (142) Liechti, M. E., and Vollenweider, F. X. (2000) The serotonin (161) Milroy, C. M., Clark, J. C., and Forrest, A. R. (1996) uptake inhibitor citalopram reduces acute cardiovascular and Pathology of deaths associated with “ecstasy” and “eve” misuse. J. Clin. vegetative effects of 3,4methylenedioxymethamphetamine (’Ecstasy’) Pathol. 49, 149−153. in healthy volunteers. J. Psychopharmacol. 14, 269−274. (162) Giraudon, I., and Bello, P. Y. (2007) Monitoring ecstasy (143) Henry, J. A., Jeffreys, K. J., and Dawling, S. (1992) Toxicity content in France: results from the National Surveillance System and deaths from 3,4-methylenedioxymethamphetamine (“ecstasy”). 1999−2004. Subst. Use Misuse 42, 1567−1578. Lancet 340, 384−387. (163) Palamar, J. J., Salomone, A., Vincenti, M., and Cleland, C. M. (144) Henry, J. A. (1992) Ecstasy and the dance of death. BMJ. 305, (2016) Detection of ″″ and other novel psychoactive 5−6. substances in hair samples of ecstasy/MDMA/″Molly″ users. Drug (145) Brown, P. L., and Kiyatkin, E. A. (2004) Brain hyperthermia Alcohol Depend. 161, 200−205. induced by MDMA (ecstasy): modulation by environmental (164) Yuki, F., Rie, I., Miki, K., Mitsuhiro, W., Naotaka, K., and conditions. Eur. J. Neurosci. 20,51−58. Kenichiro, N. (2013) Warning against co-administration of 3,4- (146) Fantegrossi, W. E., Godlewski, T., Karabenick, R. L., Stephens, methylenedioxymethamphetamine (MDMA) with methamphetamine J.M.,Ullrich,T.,Rice,K.C.,andWoods,J.H.(2003) from the perspective of pharmacokinetic and pharmacodynamic Pharmacological characterization of the effects of 3,4-methylenediox- evaluations in rat brain. Eur. J. Pharm. Sci. 49,57−64. ymethamphetamine (“ecstasy”) and its enantiomers on lethality, core (165) Moratalla, R., Khairnar, A., Simola, N., Granado, N., García- temperature, and locomotor activity in singly housed and crowded Montes, J. R., Porceddu, P. F., Tizabi, Y., Costa, G., and Morelli, M. mice. Psychopharmacology (Berlin) 166, 202−211. (2017) Amphetamine-related drugs neurotoxicity in humans and in (147) Spanos, L. J., and Yamamoto, B. K. (1989) Acute and experimental animals: Main mechanisms. Prog. Neurobiol. 155, 149− subchronic effects of methylenedioxymethamphetamine 170. [(±)MDMA] on locomotion and serotonin syndrome behavior in (166) Gouzoulis-Mayfrank, E., and Daumann, J. (2009) Neuro- the rat. Pharmacol., Biochem. Behav. 32, 835−840. toxicity of drugs of abuse - the case of methylenedioxy amphetamines (148) Schmid, Y., Rickli, A., Schaffner, A., Duthaler, U., Grouzmann, (MDMA, ecstasy), and amphetamines. Dialogues Clin. Neurosci. 11, E., Hysek, C. M., and Liechti, M. E. (2015) Interactions between 305−317. and 3,4-methylenedioxymethamphetamine in healthy (167) Baumann, M. H., Wang, W., and Rothman, R. B. (2007) subjects. J. Pharmacol. Exp. Ther. 353, 102−111. Methylenedioxymethamphetamine (MDMA) neurotoxicity in rats: a (149) Hysek, C. M., Fink, A. E., Simmler, L. D., Donzelli, M., reappraisal of past and present findings. Psychopharmacology (Berlin) Grouzmann, E., and Liechti, M. E. (2013) Alpha-adrenergic receptors 189, 407−424.

2424 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

(168) Green, A. R., King, M. V., Shortall, S. E., and Fone, K. C. Neurotoxicity of Ecstasy metabolites in rat cortical neurons, and (2012) Lost in translation: preclinical studies on 3,4-methylenediox- influence of hyperthermia. J. Pharmacol. Exp. Ther. 316,53−61. ymethamphetamine provide information on mechanisms of action, (186) Stumm, G., Schlegel, J., Schafer,̈ T., Würz, C., Mennel, H. D., but do not allow accurate prediction of adverse events in humans. Br. Krieg, J. C., and Vedder, H. (1999) Amphetamines induce apoptosis J. Pharmacol. 166, 1523−1536. and regulation of bcl-x splice variants in neocortical neurons. FASEB J. (169) Halpin, L. E., Collins, S. A., and Yamamoto, B. K. (2014) 13, 1065−1072. Neurotoxicity of methamphetamine and 3,4-methylenedioxymetham- (187) Chu, T., Kumagai, Y., DiStefano, E. W., and Cho, A. K. (1996) phetamine. Life Sci. 97,37−44. Disposition of methylenedioxy methamphetamine and three metab- (170)Steinkellner,T.,Freissmuth,M.,Sitte,H.H.,and olites in the brains of different rat strains and their possible roles in Montgomery, T. (2011) The ugly side of amphetamines: short- and acute serotonin depletion. Biochem. Pharmacol. 51, 789−796. long-term toxicity of 3,4-methylenedioxymethamphetamine (MDMA, (188) Commins, D. L., Vosmer, G., Virus, R. M., Woolverton, W. L., ‘Ecstasy’), methamphetamine and D-amphetamine. Biol. Chem. 392, Schuster, C. R., and Seiden, L. S. (1987) Biochemical and histological 103−115. evidence that methylenedioxymethylamphetamine (MDMA) is toxic (171) Lyles, J., and Cadet, J. L. (2003) Methylenedioxymeth- to neurons in the rat brain. J. Pharmacol. Exp. Ther. 241, 338−345. amphetamine (MDMA, Ecstasy) neurotoxicity: cellular and molecular (189) Logan, B. J., Laverty, R., Sanderson, W. D., and Yee, Y. B. mechanisms. Brain Res. Rev. 42, 155−168. (1988) Differences between rats and mice in MDMA (methylene- (172) Schilt, T., de Win, M. M., Koeter, M., Jager, G., Korf, D. J., van dioxymethylamphetamine) neurotoxicity. Eur. J. Pharmacol. 152, den Brink, W., and Schmand, B. (2007) Cognition in novice ecstasy 227−234. users with minimal exposure to other drugs: a prospective cohort (190) Granado, N., O’Shea, E., Bove, J., Vila, M., Colado, M. I., and study. Arch. Gen. Psychiatry 64, 728−736. Moratalla, R. (2008) Persistent MDMA-induced dopaminergic (173) Wunderli, M. D., Vonmoos, M., Fürst, M., Schadelin,̈ K., neurotoxicity in the striatum and substantia nigra of mice. J. Kraemer, T., Baumgartner, M. R., Seifritz, E., and Quednow, B. B. Neurochem. 107, 1102−1112. (2017) Discrete memory impairments in largely pure chronic users of (191) O’Callaghan, J. P., and Miller, D. B. (1994) Neurotoxicity MDMA. Eur. Neuropsychopharmacol. 27, 987−999. profiles of substituted amphetamines in the C57BL/6J mouse. J. (174) McCann, U. D., Mertl, M., Eligulashvili, V., and Ricaurte, G. Pharmacol. Exp. Ther. 270, 741−751. A. (1999) Cognitive performance in (±) 3,4-methylenedioxyme- (192) O’Hearn, E., Battaglia, G., De Souza, E. B., Kuhar, M. J., and thamphetamine (MDMA, “ecstasy”) users: a controlled study. Molliver, M. E. (1988) Methylenedioxyamphetamine (MDA) and Psychopharmacology (Berlin) 143, 417−425. methylenedioxymethamphetamine (MDMA) cause selective ablation (175) Gouzoulis-Mayfrank, E., Daumann, J., Tuchtenhagen, F., Pelz, of serotonergic axon terminals in forebrain: immunocytochemical S., Becker, S., Kunert, H., Fimm, B., and Sass, H. (2000) Impaired evidence for neurotoxicity. J. Neurosci. 8, 2788−2803. cognitive performance in drug free users of recreational ecstasy (193) Molliver, M. E., Berger, U. V., Mamounas, L. A., Molliver, D. (MDMA). J. Neurol., Neurosurg. Psychiatry 68, 719−725. C., O’Hearn, E., and Wilson, M. A. (1990) Neurotoxicity of MDMA (176) Kuypers, K. P., Theunissen, E. L., van Wel, J. H., de Sousa and related compounds: anatomic studies. Ann. N. Y. Acad. Sci. 600, Fernandes Perna, E. B., Linssen, A., Sambeth, A., Schultz, B. G., and 640−661. Ramaekers, J. G. (2016) Verbal memory impairment in polydrug (194) Battaglia, G., Yeh, S. Y., and De Souza, E. B. (1988) MDMA- ecstasy users: A clinical perspective. PLoS One 11, e0149438. induced neurotoxicity: parameters of degeneration and recovery of (177) McCann, U. D., Ridenour, A., Shaham, Y., and Ricaurte, G. A. brain serotonin neurons. Pharmacol., Biochem. Behav. 29, 269−274. (1994) Serotonin neurotoxicity after (±)3,4-methylenedioxymetham- (195) Xie, T., Tong, L., McLane, M. W., Hatzidimitriou, G., Yuan, J., phetamine (MDMA; “Ecstasy”): a controlled study in humans. McCann, U., and Ricaurte, G. (2006) Loss of serotonin transporter Neuropsychopharmacology 10, 129−138. protein after MDMA and other ring-substituted amphetamines. (178) Mueller, F., Lenz, C., Steiner, M., Dolder, P. C., Walter, M., Neuropsychopharmacology 31, 2639−2651. Lang, U. E., Liechti, M. E., and Borgwardt, S. (2016) Neuroimaging in (196)Schmidt,C.J.,Wu,L.,andLovenberg,W.(1986) moderate MDMA use: A systematic review. Neurosci. Biobehav. Rev. Methylenedioxymethamphetamine: a potentially neurotoxic amphet- 62,21−34. amine analogue. Eur. J. Pharmacol. 124, 175−178. (179) Garg, A., Kapoor, S., Goel, M., Chopra, S., Chopra, M., (197) Scanzello, C. R., Hatzidimitriou, G., Martello, A. L., Katz, J. L., Kapoor, A., McCann, U. D., and Behera, C. (2015) Functional and Ricaurte, G. A. (1993) Serotonergic recovery after (±)3,4- magnetic resonance imaging in abstinent MDMA users: A Review. (methylenedioxy) methamphetamine injury: observations in rats. J. Curr. Drug Abuse Rev. 8,15−25. Pharmacol. Exp. Ther. 264, 1484−1491. (180) Schifano, F., Di Furia, L., Forza, G., Minicuci, N., and Bricolo, (198) Scheffel, U., Lever, J. R., Stathis, M., and Ricaurte, G. A. R. (1998) MDMA (’ecstasy’) consumption in the context of polydrug (1992) Repeated administration of MDMA causes transient down- abuse: a report on 150 patients. Drug Alcohol Depend. 52,85−90. regulation of serotonin 5-HT2 receptors. Neuropharmacology 31, (181) Gouzoulis-Mayfrank, E., and Daumann, J. J. (2006) The 881−893. confounding problem of polydrug use in recreational ecstasy/MDMA (199) Malberg, J. E., and Seiden, L. S. (1998) Small changes in users: a brief overview. J. Psychopharmacol. 20, 188−193. ambient temperature cause large changes in 3,4-methylenedioxyme- (182) de Win, M. M., Schilt, T., Reneman, L., Vervaeke, H., Jager, thamphetamine (MDMA)-induced serotonin neurotoxicity and core G., Dijkink, S., Booij, J., and van den Brink, W. (2006) Ecstasy use body temperature in the rat. J. Neurosci. 18, 5086−5094. and self-reported depression, impulsivity, and sensation seeking: a (200) Sanchez, V., Camarero, J., Esteban, B., Peter, M. J., Green, A. prospective cohort study. J. Psychopharmacol. 20, 226−235. R., and Colado, M. I. (2001) The mechanisms involved in the long- (183) Lieb, R., Schuetz, C. G., Pfister, H., von Sydow, K., and lasting neuroprotective effect of fluoxetine against MDMA (’ecstasy’)- Wittchen, H. (2002) Mental disorders in ecstasy users: a prospective- induced degeneration of 5-HT nerve endings in rat brain. Br. J. longitudinal investigation. Drug Alcohol Depend. 68, 195−207. Pharmacol. 134,46−57. (184) Capela, J. P., Ruscher, K., Lautenschlager, M., Freyer, D., (201) McCann, U. D., and Ricaurte, G. A. (2001) Caveat emptor: Dirnagl, U., Gaio, A. R., Bastos, M. L., Meisel, A., and Carvalho, F. editors beware. Neuropsychopharmacology 24, 333−334. (2006) Ecstasy-induced cell death in cortical neuronal cultures is (202) Parrott, A. C. (2005) Chronic tolerance to recreational serotonin 2A-receptor-dependent and potentiated under hyper- MDMA (3,4-methylenedioxymethamphetamine) or Ecstasy. J. thermia. Neuroscience 139, 1069−1081. Psychopharmacol. 19,71−83. (185) Capela, J. P., Meisel, A., Abreu, A. R., Branco, P. S., Ferreira, L. (203) Ricaurte, G. A., Forno, L. S., Wilson, M. A., DeLanney, L. E., M., Lobo, A. M., Remiao,̃ F., Bastos, M. L., and Carvalho, F. (2006) Irwin, I., Molliver, M. E., and Langston, J. W. (1988) (±)3,4-

2425 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

Methylenedioxymethamphetamine selectively damages central sero- ymethamphetamine)-assisted psychotherapy for treatment of resist- tonergic neurons in nonhuman primates. JAMA 260,51−55. ant, chronic post- traumatic stress disorder (PTSD). J. Psychopharma- (204) Insel, T. R., Battaglia, G., Johannessen, J. N., Marra, S., and De col. (London, U. K.) 27,40−52. Souza, E. B. (1989) 3,4-Methylenedioxymethamphetamine (“ec- (222) Mithoefer, M. C. (2011) MDMA-Assisted Psychotherapy for stasy”) selectively destroys brain serotonin terminals in rhesus the Treatment of Posttraumatic Stress Disorder: A Revised Teaching monkeys. J. Pharmacol. Exp. Ther. 249, 713−720. Manual, http://www.maps.org/research/mdma/Manual_ (205) Ricaurte, G. A., Martello, A. L., Katz, J. L., and Martello, M. B. MDMAPTSD_30Nov11.pdf (accessed Jun 12, 2018). (1992) Lasting effects of (±)-3,4-methylenedioxymethamphetamine (223) Johansen, P. O., and Krebs, T. S. (2009) How could MDMA (MDMA) on central serotonergic neurons in nonhuman primates: (ecstasy) help anxiety disorders? A neurobiological rationale. J. neurochemical observations. Primates: Neurochemical Observations. Psychopharmacol. (London, U. K.) 23, 389−391. J. Pharmacol. Exp. Ther. 261, 616−622. (224) Bouso, J. C., Doblin, R., Farre,́ M., Alcazar,́ M. A., and Gomez-́ (206) Scheffel, U., Szabo, Z., Mathews, W. B., Finley, P. A., Dannals, Jarabo, G. (2008) MDMA-assisted psychotherapy using low doses in R. F., Ravert, H. T., Szabo, K., Yuan, J., and Ricaurte, G. A. (1998) In a small sample of women with chronic post-traumatic stress disorder. vivo detection of short- and long-term MDMA neurotoxicity–a J. Psychoact. Drugs 40, 225−236. positron emission tomography study in the living baboon brain. (225) Mithoefer, M. C., Wagner, M. T., Mithoefer, A. T., Jerome, L., Synapse 29, 183−192. Martin, S. F., Yazar-Klosinski, B., Michel, Y., Brewerton, T. D., and (207) Hatzidimitriou, G., McCann, U. D., and Ricaurte, G. A. Doblin, R. (2013) Durability of improvement in post-traumatic stress (1999) Altered serotonin innervation patterns in the forebrain of disorder symptoms and absence of harmful effects or drug monkeys treated with (±)3,4-methylenedioxymethamphetamine dependency after 3,4-methylenedioxymethamphetamine-assisted psy- seven years previously: factors influencing abnormal recovery. J. chotherapy: a pro- spective long-term follow-up study. J. Psychophar- Neurosci. 19, 5096−5107. macol. (London, U. K.) 27,28−39. (208) Taffe, M. A., Weed, M. R., Davis, S., Huitron-Resendiz,́ S., (226) Amoroso, T., and Workman, M. (2016) Treating post- Schroeder, R., Parsons, L. H., Henriksen, S. J., and Gold, L. H. (2001) traumatic stress disorder with MDMA-assisted psychotherapy: A Functional consequences of repeated (±)3,4-methylenedioxymetham- preliminary meta-analysis and comparison to prolonged exposure phetamine (MDMA) treatment in rhesus monkeys. Neuropsychophar- therapy. J. Psychopharmacol. 30, 595−600. macology 24, 230−239. (227) Feduccia, A. A., Holland, J., and Mithoefer, M. C. (2018) (209) Ricaurte, G. A., DeLanney, L. E., Irwin, I., and Langston, J. W. Progress and promise for the MDMA drug development program. (1988) Toxic effects of MDMA on central serotonergic neurons in Psychopharmacology (Berlin) 235, 561−571. the primate: importance of route and frequency of drug (228) Bedi, G., Phan, K. L., Angstadt, M., and de Wit, H. (2009) administration. Brain Res. 446, 165−168. Effects of MDMA on sociability and neural response to social threat (210) Miller, D. B., and O’Callaghan, J. P. (1995) The role of and social reward. Psychopharmacology (Berlin) 207,73−83. temperature, stress, and other factors in the neurotoxicity of the (229) Hysek, C. M., Domes, G., and Liechti, M. E. (2012) MDMA substituted amphetamines 3,4-methylenedioxymethamphetamine and enhances “mind reading” of positive emotions and impairs “mind fenfluramine. Mol. Neurobiol. 11, 177−192. reading” of negative emotions. Psychopharmacology (Berlin) 222, 293− (211) Jayanthi, S., Ladenheim, B., Andrews, A. M., and Cadet, J. L. 302. (1999) Overexpression of human copper/zinc superoxide dismutase (230) Carhart-Harris, R. L., Wall, M. B., Erritzoe, D., Kaelen, M., in transgenic mice attenuates oxidative stress caused by methyl- Ferguson, B., De Meer, I., Tanner, M., Bloomfield, M., Williams, T. enedioxymethamphetamine (Ecstasy). Neuroscience 91, 1379−1387. M., Bolstridge, M., Stewart, L., Morgan, C. J., Newbould, R. D., (212) Johnson, E. A., Sharp, D. S., and Miller, D. B. (2000) Restraint Feilding, A., Curran, H. V., and Nutt, D. J. (2014) The effect of as a stressor in mice: against the dopaminergic neurotoxicity of D- acutely administered MDMA on subjective and BOLD-fMRI MDMA, low body weight mitigates restraint-induced hypothermia responses to favourite and worst autobiographical memories. Int. J. and consequent neuroprotection. Brain Res. 875, 107−118. Neuropsychopharmacol. 17, 527−540. (213) Nutt, D., King, L. A., Saulsbury, W., and Blakemore, C. (2007) (231) Gamma, A., Buck, A., Berthold, T., Hell, D., and Vollenweider, Development of a rational scale to assess the harm of drugs of F. X. (2000) 3,4- Methylenedioxymethamphetamine (MDMA) potential misuse. Lancet 369, 1047−1053. modulates cortical and limbic brain activity as measured by (214) Nutt, D. J., King, L. A., and Phillips, L. D. (2010) Drug harms [H215O]-PET in healthy humans. Neuropsychopharmacology 23, in the UK: a multicriteria decision analysis. Lancet 376, 1558−1565. 388−395. (215) Kyzar, E. J., Nichols, C. D., Gainetdinov, R. R., Nichols, D. E., (232) Danforth, A. L., Struble, C. M., Yazar-Klosinski, B., and Grob, and Kalueff, A. V. (2017) Psychedelic drugs in biomedicine. Trends C. S. (2016) MDMA- assisted therapy: a new treatment model for Pharmacol. Sci. 38, 992−1005. social anxiety in autistic adults. Prog. Neuro-Psychopharmacol. Biol. (216) Mithoefer, M. C., Grob, C. S., and Brewerton, T. D. (2016) Psychiatry 64, 237−249. Novel psychopharmacological therapies for psychiatric disorders: (233) Jerome, L., Schuster, S., and Yazar-Klosinski, B. B. (2013) Can psilocybin and MDMA. Lancet Psychiatry 3, 481−488. MDMA play a role in the treatment of substance abuse? Curr. Drug (217) Sessa, B., and Nutt, D. (2015) Making a medicine out of Abuse Rev. 6,54−62. MDMA. Br. J. Psychiatry 206,4−6. (234) Sessa, B. (2017) Why MDMA therapy for alcohol use (218) Grinspoon, L., and Bakalar, J. B. (1986) Can drugs be used to disorder? And why now? Neuropharmacology, DOI: 10.1016/j.neuro- enhance the psychotherapeutic process? Am. J. Psychother. 40, 393− pharm.2017.11.004. 404. (235) Greer, G., and Tolbert, R. (1986) Subjective reports of the (219) Riedlinger, T. J., and Riedlinger, J. E. (1994) Psychedelic and effects of MDMA in a clinical setting. J. Psychoact. Drugs 18, 319−327. entactogenic drugs in the treatment of depression. J. Psychoact. Drugs (236) Pitts, E. G., Curry, D. W., Hampshire, K. N., Young, M. B., 26,41−55. and Howell, L. L. (2018) (±)-MDMA and its enantiomers: potential (220) Mithoefer, M. C., Wagner, M. T., Mithoefer, A. T., Jerome, L., therapeutic advantages of R(−)-MDMA. Psychopharmacology (Berlin) and Doblin, R. (2011) The safety and efficacy of {±}3,4- 235, 377−392. methylenedioxymethamphetamine-assisted psychotherapy in subjects (237) Nichols, D. E., Lloyd, D. H., Hoffman, A. J., Nichols, M. B., with chronic, treatment-resistant posttraumatic stress disorder: the and Yim, G. K. (1982) Effects of certain hallucinogenic amphetamine first randomized controlled pilot study. J. Psychopharmacol. (London, analogues on the release of [3H]serotonin from rat brain U. K.) 25, 439−452. synaptosomes. J. Med. Chem. 25, 530−535. (221) Oehen, P., Traber, R., Widmer, V., and Schnyder, U. (2013) A (238) Johnson, M. P., Hoffman, A. J., and Nichols, D. E. (1986) randomized, controlled pilot study of MDMA (±3,4-methylenediox- Effects of the enantiomers of MDA, MDMA and related analogues on

2426 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427 ACS Chemical Neuroscience Review

[3H]serotonin and [3H]dopamine release from superfused rat brain methylenedioxymethamphetamine has prosocial and therapeutic-like slices. Eur. J. Pharmacol. 132, 269−276. effects without signs of neurotoxicity in mice. Neuropharmacology 128, (239) Schmidt, C. J., Levin, J. A., and Lovenberg, W. (1987) In vitro 196−206. and in vivo neuro-chemical effects of methylenedioxymethamphet- (254) Frau, L., Simola, N., Plumitallo, A., and Morelli, M. (2013) amine on striatal systems in the rat brain. Biochem. Microglial and astroglial activation by 3,4-methylenedioxymetham- Pharmacol. 36, 747−755. phetamine (MDMA) in mice depends on S(+) enantiomer and is (240) Steele, T. D., Nichols, D. E., and Yim, G. K. W. (1987) associated with an increase in body temperature and motility. J. Stereochemical effects of 3,4-methylenedioxymethamphetamine Neurochem. 124,69−78. (MDMA) and related amphetamine derivatives on inhibition of (255) Freudenmann, R. W., Oxler, F., and Bernschneider-Reif, S. uptake of [3H] monoamines synaptosomes from different regions (2006) The origin of MDMA (ecstasy) revisited: the true story into of rat brain. Biochem. Pharmacol. 36, 2297−2303. reconstructed from the original documents. Addiction 101, 1241− (241) Verrico, C. D., Miller, G. M., and Madras, B. K. (2007) 1245. MDMA (ecstasy) and human dopamine, norepinephrine, and (256) Adam, D. (2006) Origins of ecstasy an urban myth. Nat. Rev. serotonin transporters: implications for MDMA-induced neuro- Drug Discovery 5, 806−807. toxicity and treatment. Psychopharmacology 189, 489−503. (257) Passie, T., and Benzenhöfer, U. (2018) MDA, MDMA, and (242) Hiramatsu, M., and Cho, A. K. (1990) Enantiomeric other “mescaline-like” substances in the US military’s search for a differences in the effects of 3,4-methylenedioxymethamphetamine truth drug (1940s to 1960s). Drug Test. Anal. 10,72−80. on extracellular mono- amines and metabolites in the striatum of (258) Hardman, H. F., Haavik, C. O., and Seevers, M. H. (1973) freely-moving rats: an in vivo microdialysis study. Neuropharmacology Relationship of the structure of mescaline and seven analogs to 29, 269−275. toxicity and behavior in five species of laboratory animals. Toxicol. (243) Acquas, E., Pisanu, A., Spiga, S., Plumitallo, A., Zernig, G., and Appl. Pharmacol. 25, 299−309. Di Chiara, G. (2007) Differential effects of intravenous R,S-(±)-3,4- (259) Biniecki, S., and Krajewski, E. (1960) Production of d,1-N- methylenedioxymethamphetamine (MDMA, ecstasy) and its S(+)- methyl-beta- (3,4-methylenedioxyphenyl)-isopropylamine and d,1-N- and R(−)-enantiomers on dopamine transmission and extracellular methyl-beta-(3,4-dimthoxyphenyl)-isopropylamine. Acta Polon. − signal regulated kinase phosphorylation (pERK) in the rat nucleus Pharm. 17, 421 425. ̈ accumbens shell. J. Neurochem. 102, 121−132. (260) Benzenhofer, U., and Passie, T. (2010) Rediscovering MDMA (244) Murnane, K. S., Fantegrossi, W. E., Godfrey, J. R., Banks, M. (ecstasy): the role of the American chemist Alexander T. Shulgin. − L., and Howell, L. L. (2010) Endocrine and neurochemical effects of Addiction 105, 1355 1361. 3,4- methylenedioxymethamphetamine and its stereoisomers in rhesus (261) Climko, R. P., Roehrich, H., Sweeney, D. R., and Al-Razi, J. monkeys. J. Pharmacol. Exp. Ther. 334, 642−650. (1987) Ecstacy: a review of MDMA and MDA. Int. J. Psychiatry Med. − (245) Murnane, K. S., Kimmel, H. L., Rice, K. C., and Howell, L. L. 16, 359 372. (2012) The neuropharmacology of prolactin secretion elicited by 3,4- (262) Ricaurte, G. A., Yuan, J., Hatzidimitriou, G., Cord, B. J., and methylenedioxymethamphetamine (“ecstasy”): a concurrent micro- McCann, U. D. (2002) Severe dopaminergic neurotoxicity in primates “ ” dialysis and plasma analysis study. Horm. Behav. 61, 181−190. after a common recreational dose regimen of MDMA ( ecstasy ). − (246) Lyon, R. A., Glennon, R. A., and Titeler, M. (1986) 3,4- Science 297, 2260 2263. Methylenedioxymethamphetamine (MDMA): stereoselective inter- (263) Ricaurte, G. A., Yuan, J., Hatzidimitriou, G., Cord, B. J., and actions at brain 5-HT1 and 5-HT2 receptors. Psychopharmacology McCann, U. D. (2003) Retraction: Severe dopaminergic neuro- − toxicity in primates after a common recreational dose regimen of (Berlin) 88, 525 526. “ ” (247) Toll, L., Berzetei-Gurske, I. P., Polgar, W. E., Brandt, S. R., MDMA ( ecstasy ). Science 301, 1479. Adapa, I. D., Rodriguez, L., Schwartz, R. W., Haggart, D., O’Brien, A., (264) Nutt, D. J. (2009) Equasy- an overlooked addiction with implications for the current debate on drug harms. J. Psychopharmacol. White, A., Kennedy, J. M., Craymer, K., Farrington, L., and Auh, J. S. − (1998) Standard binding and functional assays related to medications 23,3 5. development division testing for potential cocaine and opiate narcotic treatment medications. NIDA Res. Monogr. 178, 440−466. (248) Nash, J. F., Roth, B. L., Brodkin, J. D., Nichols, D. E., and Gudelsky, G. A. (1994) Effect of the R(−) and S(+) isomers of MDA and MDMA on phosphatidyl inositol turnover in cultured cells expressing 5-HT2A or 5-HT2C receptors. Neurosci. Lett. 177, 111− 115. (249) Baker, L. E., and Taylor, M. M. (1997) Assessment of the MDA and MDMA optical isomers in a stimulant-hallucinogen discrimination. Pharmacol., Biochem. Behav. 57, 737−748. (250) Murnane, K. S., Murai, N., Howell, L. L., and Fantegrossi, W. E. (2009) Discriminative stimulus effects of psychostimulants and hallucinogens in S(+)-3,4-methylenedioxymethamphetamine (MDMA) and R(−)-MDMA trained mice. J. Pharmacol. Exp. Ther. 331, 717−723. (251) Forsling, M. L., Fallon, J. K., Shah, D., Tilbrook, G. S., Cowan, D. A., Kicman, A. T., and Hutt, A. J. (2002) The effect of 3,4- methylenedioxymethamphetamine (MDMA, ‘ecstasy’) and its metab- olites on neurohypophysial hormone release from the isolated rat hypothalamus. Br. J. Pharmacol. 135, 649−656. (252) Pitts, E. G., Minerva, A. R., Chandler, E. B., Kohn, J. N., Logun, M. T., Sulima, A., Rice, K. C., and Howell, L. L. (2017) 3,4- Methylenedioxymethamphetamine increases affiliative behaviors in squirrel monkeys in a serotonin 2A receptor-dependent manner. Neuropsychopharmacology 42, 1962−1971. (253) Curry, D. W., Young, M. B., Tran, A. N., Daoud, G. E., and Howell, L. L. (2018) Separating the agony from ecstasy: R(−)-3,4-

2427 DOI: 10.1021/acschemneuro.8b00155 ACS Chem. Neurosci. 2018, 9, 2408−2427