MOBILE VIEW  | 

HALLUCINOGENIC AMPHETAMINES

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Hallucinogenic amphetamines are primarily used as drugs of abuse. They are usually ingested, but occasionally may be taken intranasally or intravenously. Phenylethylamines stimulate catecholamine, dopamine and serotonin activity. MDMA is a synthetic member of this class. Other common synthetic amphetamine analogues that are hallucinogenic include 4-bromo-2,5-dimethoxyphenylethylamine and 2,5-dimethoxy-4-N-propylthipehenethylamine, among others. Naturally occurring phenylethylamines include mescaline and other nonnative cactus species (See PLANTS-PEYOTE/MESCALINE).
    B) The methoxylated and/or methylenedioxy derivatives of amphetamine have properties in common with both amphetamine and mescaline.
    C) BODY PACKERS/BODY STUFFERS: Please refer to the appropriate management if body packing or body stuffing is known/suspected.

Specific Substances

    A) ALEPH (synonym)
    1) 4-Methylthio-2,5-dimethoxy-amphetamine (synonym)
    ALEPH 2 (synonym)
    1) 4-Ethylthio-2,5-dimethoxy-amphetamine (synonym)
    ALEPH 4 (synonym)
    1) 4-Isopropylthio-2,5-dimethoxy-amphetamine (synonym)
    ALEPH 6 (synonym)
    1) 4-Phenylthio-2,5-dimethoxy-amphetamine (synonym)
    ALEPH 7 (synonym)
    1) 4-Propylthio-2,5-dimethoxy-amphetamine (synonym)
    BEATRICE (synonym)
    1) 2,5-dimethoxy-4,N-dimethyl-amphetamine (synonym)
    BDB (synonym)
    1) Benzodioxolylbutanamine (synonym)
    BOB (synonym)
    1) 4-Bromo-2,5,beta-trimethoxy-phenethylamine (synonym)
    BOD (synonym)
    1) 2,5,beta-trimethoxy-4-methyl-phenethylamine (synonym)
    BOH (synonym)
    1) beta-Methoxy-3,4-methylenedioxy-phenethylamine (synonym)
    BOHD (synonym)
    1) 2,5-Dimethoxy-beta-hydroxy-4-methyl-phenethylamine (synonym)
    BOM (synonym)
    1) 3,4,5,beta-tetramethoxy-phenethylamine (synonym)
    1-benzylpiperazine (synonym)
    1) BZP (synonym)
    2) "Pep pills" (synonym)
    1-(3-trifluoromethylphenyl) piperazine (synonym)
    1) TFMPP (synonym)
    1-(8-bromobenzo[1,2-b; 4,5-b']difuran-4-yl)-2-aminopropane (synonym)
    1) Bromo-Dragonfly (synonym)
    2,4-DMA (synonym)
    1) 2,4-dimethoxy-amphetamine (synonym)
    2,5-DMA (synonym)
    1) 2,5-dimethoxy-amphetamine (synonym)
    3C-B-Fly (synonym)
    1) 1-(8-bromo-2,3,6,7-tetrahydrobenzo[1,2-b;4,5-b']-difuran-4-yl)-2-aminopropane (synonym)
    2) 3CBFly (synonym)
    3) Bromofly (synonym)
    3,4-DMA (synonym)
    1) 3,4-dimethoxy-amphetamine (synonym)
    4-METHYL AMINOREX (synonym)
    1) 4,5-dihydro-4-methyl-5-phenyl-2-oxazolamine (synonym)
    4-D (synonym)
    1) 4-Trideuteromethyl-3,5-dimethoxy-phenyethylamine (synonym)
    Beta-D
    1) beta,beta-Dideutero-3,4,5-trimethoxy-phenethylamine (synonym)
    BENZO FURY (synonym)
    1) 6-(2-aminopropyl)benzofuran
    2) 6-APB
    3) Benzofuran
    BROMO-DMA (synonym)
    1) 4-bromo-2,5-dimethoxy amphetamine (synonym)
    2) Bromo-"STP" (synonym)
    3) DOB (synonym)
    DESOXY (synonym)
    1) 4-methyl-3,5-dimethoxy-phenethylamine (synonym)
    diphenyl-2-pyrrolidinemethanol
    1) diphenylprolinol (synonym)
    2) D2PM (synonym)
    3) Head Candy (synonym)
    DMCPA (synonym)
    1) 2-(2,5-dimethoxy-4-methylphenyl)-cyclopropylamine (synonym)
    DME (synonym)
    1) 3,4-dimethoxy-beta-hydroxy-phenethylamine (synonym)
    DMMDA (synonym)
    1) 2,5-dimethoxy-3,4-methylenedioxy-amphetamine (synonym)
    DMMDA-2 (synonym)
    1) 2,3-dimethoxy-4,5-methylenedixoy-amphetamine (synonym)
    DMPEA (synonym)
    1) 3,4-dimethoxy-phenethylamine (synonym)
    DOAM (synonym)
    1) 4-amyl-2,5-dimethoxy-amphetamine (synonym)
    DOB (synonym)
    1) 4-Bromo-2,5-dimethoxy-amphetamine (synonym)
    DOBU (synonym)
    1) 4-Butyl-2,5-dmethoxy-amphetamine (synonym)
    DOC (synonym)
    1) 2,5-dimethoxy-4-chloramphetamine (synonym)
    DOEF (synonym)
    1) 4-(2-fluoroethyl)-2,5-dimethoxy-amphetamine (synonym)
    DOET (synonym)
    1) 4-ethyl-2,5-dimethoxy-amphetamine (synonym)
    DOI (synonym)
    1) 2,5-dimethoxy-4-iodoamphetamine (synonym)
    DOM (synonym)
    1) 2,5-dimethoxy-4-methylamphetamine (synonym)
    2) STP (synonym)
    3) Serenity, Tranquility, and Peace (synonym)
    DON (synonym)
    1) 4-Nitro-2,5-dimethoxy-amphetamine (synonym)
    DOPR (synonym)
    1) 4-Propyl-2,5-dimethoxy-amphetamine (synonym)
    G3 (synonym)
    1) 3,4-Trimethylene-2,5-dimethoxy-amphetamine (synonym)
    G4 (synonym)
    1) 3,4-Tetramethylene-2,5-dimethoxy-amphetamine (synonym)
    G5 (synonym)
    1) 3,4-Norbomyl-2,5-dimethoxy-amphetamine (synonym)
    MBDB (synonym)
    1) methylbenzodioxolylbutamine (synonym)
    MDA (synonym)
    1) 3,4-methylenedioxyamfetamine (synonym)
    2) "Eve" (synonym)
    3) Madness (slang term for MDA) (synonym)
    MDE (synonym)
    1) 3,4-methylenedioxy-N-ethylamfetamine (synonym)
    2) "Eve" (synonym)
    3) MDEA (synonym)
    4) Methylenedioxyethamphetamine (synonym)
    MDMA (synonym)
    1) Ecstasy (synonym)
    2) 3,4-methylenedioxymethamphetamine (synonym)
    3) MDM (synonym)
    4) Methylenedioxymethamphetamine (synonym)
    5) N-alpha-dimethyl-1,3-benzodioxole-t-ethanamine (synonym)
    6) Stacking (3 or more MDMA tablets in combination) (synonym)
    7) Madness (slang term for MDMA) (synonym)
    8) Lick 'em (MDMA powder suspended in jello) (synonym)
    9) Lickum (MDMA powder suspended in jello) (synonym)
    10) Lickim (MDMA powder suspended in jello) (synonym)
    11) Weisels Taub (German slang term for MDMA) (synonym)
    12) Weasel dust (German slang term for MDMA) (synonym)
    METHYL AMINOREX (synonym)
    1) "U4EuH" (synonym)
    2) CAS 29493-77-4 (synonym)
    3) Euphoria (slang term for methyl aminorex) (synonym)
    4) Mft (slang term for methyl aminorex) (synonym)
    PPP (synonym)
    1) R,S-alpha-pyrrolidinopropiophenone (synonym)
    TFMPP (synonym)
    1) 1-(3-trifluoromethylphenyl)-piperazine (synonym)
    TMA (synonym)
    1) 3,4,5-trimethoxyamphetamine (synonym)
    TMA-2 (synonym)
    1) 2,4,5-trimethoxyamphetamine (synonym)
    TMA-3 (synonym)
    1) 2,3,4-trimethoxyamphetamine (synonym)
    TMA-4 (synonym)
    1) 2,3,5-trimethoxyamphetamine (synonym)
    TMA-5 (synonym)
    1) 2,3,6-trimethoxyamphetamine (synonym)
    TMA-6 (synonym)
    1) 2,4,6-trimethoxyamphetamine (synonym)
    GENERAL TERMS
    1) Blue Dolphin ecstasy (synonym)

Available Forms Sources

    A) FORMS
    1) More than 50 hallucinogenic amphetamines have been synthesized. Some agents (eg, MDA, MDMA) have been classified by the FDA as schedule I substances (no accepted medical use and high abuse potential).
    2) Since most hallucinogenic amphetamines have not yet been classified, they can be synthesized and sold without restriction or legal penalties. The purity and identity of street samples should always be questioned until confirmed by laboratory analysis.
    3) Hallucinogenic amphetamines are usually ingested but occasionally may be taken intranasally or intravenously.
    4) 2,5-Dimethoxy-4-n-propylthiophenethylamine (2C-T-7): This agent is structurally and pharmacodynamically similar to methylenedioxymethamphetamine (MDMA). A 20-year-old man died after insufflating 35 mg of 2C-T-7 (Curtis et al, 2003).
    5) 2-[2,5-dimethoxy-4-isopropylsulfanylphenyl) ethanamine (2C-T-4): This agent is a phenethylethanamine analog and structurally similar to 2C-T-7. At the time of this review, 2C-T-4 is not legally controlled in the US or Japan (Miyajima et al, 2008).
    6) Street names of the most popular agents are given below.
    AgentStreet Names
    DOBBromo DMA, Golden Eagle, PBR, LSD-25, Psychodrine, Tile, 100X, Bromo-STP
    DOMPink Wedge, STP (Serenity, Tranquility, and Peace)
    MDAHarmony, Love, Love Drug, Speed for Lovers
    MDEAEve
    MDMAAdam, Bean, Buffalo, California Sunrise, Dove, Double Diamond, Ecstasy, Love Heart, MDM, X, XTC, Essence, Roll, Rhubarb & Custard (red and yellow capsule), Snowball, White Burger, Whizz, Whizz Bomb
    PMADeath, Dr. Death
    2CBEve, Spectrum
    2C-T-7Blue Mystic, Beautiful, Belladonna, Tweetybird Mescaline

    7) MDMA
    a) Is found in tablet, capsule and powder form in doses of 50 to 110 mg. Of samples alleged to contain MDMA, 58% actually contained this compound. Samples were contaminated with MDA in 24 of 101 samples. Other adulterants were not found (Renfroe, 1986).
    b) Street samples alleged to contain 120 mg of MDMA actually contained an average of 101 mg; samples alleged to contain 130 to 135 mg actually contained 108 mg (Siegel, 1986).
    c) Although MDMA is classified as a schedule I drug, it is estimated by the US media that annually hundreds of thousands of doses are used illegally. Most commonly used by young people as they "roll" at underground rave parties that can last for many hours (Rella et al, 2000).
    d) NOTE: The term "Ecstasy" which is most often associated with MDMA has also been linked to gamma hydroxybutyrate (GHB), 'liquid ecstasy'; and 'herbal ecstasy', an ephedrine-containing preparation. Because these agents are pharmacologically different, it is necessary to correctly identify the suspected agent(s) (Doyon, 2001).
    8) PARAMETHOXYAMPHETAMINE
    a) Paramethoxyamphetamine (PMA, "death") is a ring substituted amphetamine (a methoxylated phenethylamine derivative) (White et al, 1997). See PARAMETHOXYAMPHETAMINE management for further information.
    9) ADULTERANTS
    a) Paramethoxyamphetamine (PMA, "death") a ring substituted amphetamine (a methoxylated phenethylamine derivative) has been deceptively marketed as MDMA (White et al, 1997; Felgate et al, 1998) and has resulted in several deaths (Byard et al, 1999). It has purportedly been used infrequently in Australia with minimal cases reported in the United States or United Kingdom (Byard et al, 1999).
    b) Two cases of lead poisoning associated with methamphetamine use have been reported; lead acetate is a component of one methamphetamine synthesis procedure (Alcott et al, 1987).
    c) In some studies, adulteration of MDMA tablets has included oriental herbal ephedrine (Ma Huang) and ketamine (Schwartz & Miller, 1997).
    d) In a study conducted in the United Kingdom, the composition of illicitly manufactured Ecstasy tablets sold on the streets was analyzed by gas chromatography. The results indicated frequently that other stimulants (caffeine, amphetamine, methamphetamine, paracetamol, ketamine) may be found in tablets that are sold as Ecstasy. In this particular study, Ecstasy tablets were not found to contain cocaine, heroin, or other opiates as might be reported by users. The authors also noted that "brand" names did not provide any guarantee of either the ingredient or concentration (Sherlock et al, 1999).
    e) The US nonprofit organization, DanceSafe, promotes harm reduction in the rave and nightclub community and tests voluntarily submitted Ecstasy tablets for purity. Between 1999 and July 2005, 1214 tablets were tested, and results found that 39% (n=473) of tablets contained MDMA only, 46% (n=558) of tablets contained substances other than MDMA, and 15.1% (n=183) of tablets contained MDMA and other non-MDMA substances (Tanner-Smith, 2006).
    f) BZP with TFMPP
    1) A combination product of BZP with TFMPP (1-(3-trifluoromethylphenyl) piperazine) has been sold as Ecstasy, and in animal models has been associated with MDMA-like effects. Three friends who each ingested 4 tablets over the course of one evening presented with sympathomimetic toxicity and dissociative-like symptoms. All made a full recovery within hours following supportive care (Wood et al, 2008).
    2) FORMS: BZP is most commonly ingested in tablet or capsule form as "party pills" or "herbal highs". The powder can also be snorted or mixed in a drink; rarely by the intravenous route. Users may ingest 2 to 3 tablets in one occasion, but this number is highly variable. Onset: 2 to 3 hours after ingestion. Of note, BZP is often made up of a blend of (1-(3-trifluoromethylphenyl) piperazine (TFMPP) in a ratio of 2:1 to 10:1 and both are considered amphetamine-like compounds; however, it has been suggested that TFMPP can produce psychedelic effects (Cohen & Butler, 2011).
    B) USES
    1) RING SUBSTITUTED METHYLENEDIOXYPHENETHYLAMINES
    a) SUBSTITUTED METHYLENEDIOXYPHENETHYLAMINES
    1) MDMA, 3,4-methylenedioxyamfetamine (MDA), methylbenzodioxolylbutaanamine (MBDB), benzodioxolylbutanamine (BDB) and 3,4-methylenedioxy-N-ethylamfetamine (MDE) are the more well known designer hallucinogenic amphetamines. MDMA is considered the most widely used designer drug (Hill & Thomas, 2011).
    2) MDMA is considered an entheogen (a neologism derived from Greek roots) which means to become divine from within. Entheogen is used to describe shamanic or ecstatic possession induced by ingestion of mind-altering drugs. The drug is popular because of the desire for euphoria, intense closeness, and a need to feel serenity or peace (Rella et al, 2000). Also known as the "hug drug" because it gives the user feelings that "all is right with the world" (Shannon, 2000).
    2) PHENETHYLAMINES
    a) Most phenethylamines have stimulant properties that are frequently altered to include psychoactive properties. Tolerance can develop. MDMA is one of the more familiar agents of this group. Its basic structure is based on the amino acid phenylalanine and consists of an aromatic ring with a 2 carbon side chain leading to a terminal amine group. When 'designer' substitutions are made to the aromatic ring this can lead to compounds that are psychoactive via either serotonin release (eg, MDMA, paramethoxyoxyamfetamine) or serotonin receptor agonism (2C and D series) that produce less stimulant effects. Hallucinogenic effects appear to be conferred by methoxy groups at the 2nd and 5th positions with a hydrophobic substitution at the 4th position. Iodine and bromine substituted phenethylamines produce more hallucinogenic effects than the hydrogen and nitrogen equivalents (Hill & Thomas, 2011).
    3) RING SUBSTITUTED PHENETHYLAMINES
    a) 2C SERIES
    1) The 2C series are a large group of chemicals characterized by methoxy groups at positions 2 and 5 of the benzene ring. The differ from D series of substituted amfetamines by not having a methyl group on the alpha carbon of the side chain. Bromine and iodine can be substituted at the fourth position on the benzene ring (eg, 2C-B, 2C-I). Onset of effects occurs within a few minutes after insufflation and approximately one hour after oral administration. Peak effects occur at 2 hours and last about 5 hours. Lower doses (eg, less than 10 mg for 2C-B) primarily produce stimulant effects, while doses greater than 10 mg produce psychoactive with hallucinogenic and entactogenic effects and doses of 30 mg or more produce severe hallucinations or psychosis. Deaths have been reported with these agents (Hill & Thomas, 2011). They were initially produced in the 1970s but became popular as a drug of abuse during the 1990s. 2C-B was the first agent to be created in the 2C series, followed by 2C-T-2 and 2C-T-7 and then the iodo analogue 2C-I (Meyer & Maurer, 2010).
    2) 2C-B: Is found in gelatin capsules containing 25 mg.
    3) 2C-D: It has partial agnonism at the alpha(1)-adrenergic receptors and has less hallucinogenic activity compared to other agents in the series (Meyer & Maurer, 2010).
    4) 2C-E: It has been used as an illicit drug in several countries (Meyer & Maurer, 2010).
    5) 2C-T-2: It has been marketed alone or sold in combination with other designer drugs (Meyer & Maurer, 2010).
    6) 2C-T-4: found in a sexual enhancing product called "Vanilla Aroma" (Miyajima et al, 2008).
    7) 2C-T-7: It is a sulfur analogue of 2C-B. Fatalities have been associated with this agent (Meyer & Maurer, 2010).
    8) The following are a list of agents associated with this series (Hill & Thomas, 2011):
    1) 2CB
    2) 2CI
    3) 2CD
    4) 2CT-2
    5) 2CT-7
    6) 2CE
    7) 2CP
    8) 2CN
    9) 2CT-8
    10) 2CT-9
    11) 2CT-21
    12) 2CF
    13) 2CC
    14) 2C-TFM
    4) RING SUBSTITUTED AMFETAMINE DERIVATIVES
    a) D SERIES
    1) The D series of substituted amfetamines have methoxy groups at positions 2 and 5 of the benzene rings with variable substitutions at the fourth position of the benzene ring. This substitution likely produces hallucinogenic effect via 5HT2a agonism. Onset can be delayed up to an hour, which may increase the risk of repeat dosing by inexperienced users. The D series is considered more potent and has a longer duration. They appear to have more vasoconstrictive properties, that can lead to more significant adverse events (ie, agitation, seizures, metabolic acidosis) and death (Hill & Thomas, 2011).
    2) The following are a list of agents associated with this series (Hill & Thomas, 2011):
    1) DOB
    2) DOC
    3) DOI
    4) DOM
    5) DON
    6) DOEF
    7) DOET
    8) DOPR
    9) DOTFM
    10) Aleph-1
    11) Aleph-6
    12) Aleph-7
    13) Ariadne
    14) Beatrice
    15) G3
    16) G5
    b) BENZODIFURANS
    1) Tetrahydrobenzodifuranyl and benzodifuranyl aminoalkanes (FLY and DragonFly) are potent hallucinogens due to their characteristic dihydrofuran or difuran rings. BromodragonFLY (DOB-Dragnonfly, 1-(8-bromobenzo[1,2-b;4,5-b']difuran-4-yl)-2-aminopropane) is one of the subgroups of phenethylamines. A typical dose of bromodragonFLY is 0.2 to 1 mg with an onset of up to 6 hours and a duration of 2 to 3 days. Adverse events related to the D series included agitation, tachycardia, mydriasis, hallucinations, severe limb ischaemia, seizures, and liver and renal failure (Hill & Thomas, 2011).
    c) OTHERS
    1) Other agents include paramethoxyamfetamine (PMA, 'death'), paramethoxymethamfetamine (PMMA and 4-methytrioamfetamine (4-MTA, 'flatliner') and appear to be more potent than other phenethylamines. Adverse events have resulted in severe hyperthermia likely due to serotonin toxicity (Hill & Thomas, 2011).
    2) The following are a list of agents associated with this series (Hill & Thomas, 2011):
    1) PMA
    2) PMMA
    3) 4-MTA
    4) TMA
    5) TMA-2
    6) 2MA
    7) 3MA

Life Support

    A) This overview assumes that basic life support measures have been instituted.

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Used primarily as drugs of abuse. Some related substances (e.g., peyote) are used legally in Native American religious ceremonies.
    B) PHARMACOLOGY: Phenylethylamines cause clinical effects by a complex and poorly understood stimulation of catecholamine, dopamine and serotonin activity. Naturally occurring phenylethylamines include mescaline, found in peyote (Lophophora williamsii) and other nonnative cactus species including the San Pedro cactus and Peruvian torch cactus (i.e., Trichocereus pachanoi and Trichocererus peruvianus). MDMA is a synthetic member of the class. Other common synthetic amphetamine analogues that are hallucinogenic include 4-bromo-2,5-dimethoxyphenethylamine and 2,5-dimethoxy-4-N-propylthiophenethylamine, among others. Although their mechanism of action of phenylethylamines is not completely understood, it is thought that all hallucinogens share a common site of action on central serotonin receptors, specifically 5-HT2 receptors. The dopaminergic system is also involved.
    C) TOXICOLOGY: It is an extension of the pharmacologic effects and can result in severe tachycardia, agitation, hyperthermia, seizures and end-organ damage. Serotonin syndrome can result from excessive serotonergic activity. Dopamine release contributes to the addictive and psychoactive properties of this class of substances.
    D) EPIDEMIOLOGY: Hallucinogens are a diverse group of substances that alter perception, thought and mood. Hallucinogens can be broken down into groups according to their chemical structure. One of the major classes of hallucinogens is the alkoxy ring-substituted phenylethylamines, otherwise known as hallucinogenic amphetamines. They may occur in the natural environment or as a synthetic agent. Poisoning is not common, but may be severe. Exposure may occur via oral, mucosal, parental, or inhalation routes depending on the substance.
    E) OTHER: For further detailed information on N-benzylpiperazine (BZP), dimethylamylamine (DMAA), and paramethoxyamphetamine (PMA) see the following managements: N-BENZYLPIPERAZINE, DIMETHYLAMYLAMINE and PARAMETHOXYAMPHETAMINE.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE POISONING: All of these agents have the potential to cause either pleasant or frightening hallucinations. Early sympathetic effects include mydriasis, dizziness, tachycardia and hypertension. Bruxism is common, especially in MDMA users. Nausea and vomiting are common with peyote in recreational doses or doses used for religious ceremonies and often precede the hallucinations. PMA intoxications are characterized by hypoglycemia, hyperkalemia and QRS interval prolongation.
    2) SEVERE POISONING: Severe effects may include tachypnea, hyperthermia, delirium, psychosis, seizures, coma and ventricular dysrhythmias. Rhabdomyolysis and renal failure may rarely develop in patients with prolonged agitation, hyperthermia, coma or seizures. Elevated blood pressure can lead to intracerebral hemorrhages. MDMA can cause inappropriate antidiuretic hormone release, which combined with often excessive water consumption while dancing at night clubs or warehouse parties, can result in profound and potentially fatal hyponatremia.
    3) ADVERSE EFFECTS (CHRONIC): Long-term abusers commonly exhibit chronic toxicity. Cardiovascular toxicities can include necrotizing vasculitis of small to medium arteries and may affect any organ system, aortic and mitral valve insufficiency, cardiomyopathy and pulmonary hypertension. Neurologic toxicity can include stroke and cognitive decline, presumably due to direct neurotoxicity. Long-term users of MDMA exhibit permanent destruction of serotonergic neurons.
    4) ONSET: Approximately 30 to 45 seconds after ingestion (on an empty stomach) of ecstasy a "rush" can occur, which lasts 15 to 30 minutes. This is followed by a sense of clarity and feelings of happiness. A booster dose may be taken at this point, to prolong these feelings. Thirty minutes to 3 hours after the initial ingestion a "plateau" phase occurs in which repetitive or trance-like movements become extremely pleasurable. Rhabdomyolysis can develop during this phase due to extended activity. The "coming down" phase occurs 3 to 6 hours after the initial ingestion, and can lead to negative feelings or emotions (eg, depression, anxiety). Symptoms may persist for several days.
    5) NOTE: The term "Ecstasy" which is most often associated with MDMA has also been linked to gamma-hydroxybutyrate (GHB), 'liquid ecstasy'; and 'herbal ecstasy', an ephedrine-containing preparation. Because these agents are pharmacologically different, it is necessary to correctly identify the suspected agent(s).
    6) USUAL DOSING: A usual dose of Ecstasy taken by young adults/teens is 1 to 2 mg/kg body weight (125 to 180 mg). "Candyflipping" is the intentional combination of ecstasy with LSD. Another method of use is called "stacking" in which 3 or more tablets of MDMA are taken at once; or mixing MDMA with alcohol, marijuana or other drugs of abuse (eg, ketamine, GHB, cocaine) in order to modulate the high. "Stacking" can increase the risk of overdose since MDMA, acting as a stimulant, can mask the sedative effects of alcohol or opiates.
    7) ADULTERANTS: Adulteration of so called "Ecstasy" (MDMA) tablets has included oriental herbal ephedrine (Ma Huang), ketamine, paracetamol, and caffeine. Concoctions of BZP plus TFMPP have been reportedly sold as "Ecstasy" and produced sympathomimetic toxicity and dissociative-like symptoms. So called brand names do not guarantee purity or concentration.
    8) BODY PACKERS/BODY STUFFERS: Please refer to the appropriate management if body packing or body stuffing is known/suspected.
    0.2.3) VITAL SIGNS
    A) WITH POISONING/EXPOSURE
    1) Hypertension, tachycardia, and tachypnea are common. In severe cases, hyperthermia, hypotension, and cardiovascular collapse may occur.
    0.2.20) REPRODUCTIVE
    A) Ecstasy has been associated with cardiovascular and musculoskeletal malformations in babies exposed in utero.

Laboratory Monitoring

    A) Monitor vital signs and mental status.
    B) Plasma levels are not clinically useful or readily available.
    C) Monitor sodium and renal function. Obtain an ECG and institute continuous cardiac monitoring in patients with moderate to severe toxicity (agitation, delirium, seizures, coma, hypotension).
    D) Monitor creatinine phosphokinase in patients with prolonged agitation, seizures or coma; monitor renal function and urine output in patients with rhabdomyolysis.
    E) Standard urine toxicology screen may not produce a positive result for many of the specific hallucinogenic amphetamines.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Patients are most likely to present with neuropsychiatric symptoms of psychosis. Hypertension and tachycardia are generally well tolerated and can be treated with benzodiazepines. Mildly intoxicated patients seem to do best in quiet, dark rooms with minimal stimulation. It may be helpful to remind the patient that they are experiencing a drug effect that will eventually wear off.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) The goal of treatment is to manage agitation and prevent end-organ damage. Orotracheal intubation for airway protection should be performed early. Consider activated charcoal in large overdoses of orally ingested drug (GI decontamination should be performed only in patients who can protect their airway or who are intubated). Severe delirium may develop and require large doses of benzodiazepines for sedation. Antipsychotics may be used as an adjunct treatment for agitation. Seizures may require aggressive use of benzodiazepines, propofol and/or barbiturates. Monitor for and treat dysrhythmias. Monitor core temperature and treat hyperthermia with aggressive benzodiazepine sedation to control agitation, external cooling. Clinical manifestations may be prolonged due to long half-life of many hallucinogenic amphetamines.
    C) DECONTAMINATION
    1) PREHOSPITAL: Not recommended because of the potential for agitation and seizures.
    2) HOSPITAL: Activated charcoal if recent, substantial oral ingestion, and patient able to protect airway. Administer to body stuffers that are not at risk for aspiration.
    D) AIRWAY MANAGEMENT
    1) Perform early in patients with severe intoxication (i.e., seizures, dysrhythmias, severe delirium or hyperthermia).
    E) ANTIDOTE
    1) None.
    F) ENHANCED ELIMINATION
    1) Hemodialysis and hemoperfusion are of no value.
    G) PATIENT DISPOSITION
    1) HOME CRITERIA: Asymptomatic adults may be monitored at home. However, it is unlikely that a patient will be asymptomatic.
    2) OBSERVATION CRITERIA: Patients with deliberate self-harm ingestions or children with any ingestion and symptomatic patients should be sent to a healthcare facility for observation for 6 to 8 hours.
    3) ADMISSION CRITERIA: Patients with significant persistent central nervous system toxicity (i.e., hallucinations, somnolence, delirium, coma), hyponatremia, or persistent tachycardia should be admitted. Patients with coma, seizures, dysrhythmias, serotonin syndrome or delirium should be admitted to an intensive care setting.
    4) CONSULT CRITERIA: Consult a poison center or medical toxicologist for assistance in managing patients with severe toxicity (i.e., seizures, dysrhythmias, severe delirium, coma), or in whom the diagnosis is not clear.
    H) PITFALLS
    1) Failure to control agitation and manage hyperthermia and seizures can result in death and irreversible end-organ damage. Patients with altered mentation should be ruled out for intracranial hemorrhage, infection, metabolic disturbance and hyponatremia.
    I) PHARMACOKINETICS
    1) Generally rapidly (20 to 60 min) absorbed, with an onset of one hour after a 75 to 100 mg dose of MDMA. Peak plasma levels usually occur 2 hours after oral administration. The amount of MDMA bound to plasma proteins is unknown. Large volumes of distribution. Elimination is by both hepatic and renal pathways. Elimination half-life of MDA is 9 to 19 hours, MDMA elimination is 7 to 10 hours in acid urine, and 16 to 31 hours in alkaline urine.
    J) TOXICOKINETICS
    1) Abusers will often "stack" doses of the drug, prolonging the apparent half-life and clinical effects. MDMA has nonlinear kinetics at increasing doses (increase in dose leads to disproportionate increases in serum concentrations).
    K) DIFFERENTIAL DIAGNOSIS
    1) Thyrotoxicosis, hypoglycemia, central nervous system infection, other sympathomimetic poisoning (such as cocaine), anticholinergic toxicity, mental illness presenting with mania or hallucinations, or ethanol/benzodiazepine/barbiturate withdrawal.

Range Of Toxicity

    A) Severe toxicity has developed at recreational doses in some individuals. Typical recreational doses include: DDPR 1 to 2 mg, DMA 20 to 50 mg, DOB 1 to 5 mg, DOET 0.75 to 4 mg, DOI 0.8 to 2 mg, DOM 2 to 15 mg, MDA 40 to 150 mg, MDMA 50 to 150 mg, MMDA 50 to 100 mg, DMMDA 2 to 3 mg, PMA 50 mg, TMA 50 to 150 mg, 2CB 16 to 30 mg, 2,4,5 TMA 20 mg, 2,4,6 TMA 30 to 40 mg.
    B) FATALITIES: Fatalities have been reported after ingestion of a single capsule of MDMA. Deaths have been associated with the development of hyperthermia following MDMA intoxication.
    1) An adult found at home survived a massive ingestion of ecstasy (50 tablets: each containing 50 to 150 mg of MDMA), oxazepam (10 mg), and alcohol (5 units).
    2) Deaths have also been associated with other agents and include 2C-T-7, Bromo-dragonfly and PMA/PMMA.
    C) SPECIAL SITUATION: Law enforcement and emergency medical services that enter clandestine phenylethylamine laboratories may suffer from mucous membrane and respiratory symptoms. Surface decontamination and symptomatic and supportive care is the treatment. Explosions in clandestine laboratories are common and put those in proximity at risk for burns and traumatic injuries. Children that present from clandestine laboratories are primarily at risk for abuse and neglect.

Summary Of Exposure

    A) USES: Used primarily as drugs of abuse. Some related substances (e.g., peyote) are used legally in Native American religious ceremonies.
    B) PHARMACOLOGY: Phenylethylamines cause clinical effects by a complex and poorly understood stimulation of catecholamine, dopamine and serotonin activity. Naturally occurring phenylethylamines include mescaline, found in peyote (Lophophora williamsii) and other nonnative cactus species including the San Pedro cactus and Peruvian torch cactus (i.e., Trichocereus pachanoi and Trichocererus peruvianus). MDMA is a synthetic member of the class. Other common synthetic amphetamine analogues that are hallucinogenic include 4-bromo-2,5-dimethoxyphenethylamine and 2,5-dimethoxy-4-N-propylthiophenethylamine, among others. Although their mechanism of action of phenylethylamines is not completely understood, it is thought that all hallucinogens share a common site of action on central serotonin receptors, specifically 5-HT2 receptors. The dopaminergic system is also involved.
    C) TOXICOLOGY: It is an extension of the pharmacologic effects and can result in severe tachycardia, agitation, hyperthermia, seizures and end-organ damage. Serotonin syndrome can result from excessive serotonergic activity. Dopamine release contributes to the addictive and psychoactive properties of this class of substances.
    D) EPIDEMIOLOGY: Hallucinogens are a diverse group of substances that alter perception, thought and mood. Hallucinogens can be broken down into groups according to their chemical structure. One of the major classes of hallucinogens is the alkoxy ring-substituted phenylethylamines, otherwise known as hallucinogenic amphetamines. They may occur in the natural environment or as a synthetic agent. Poisoning is not common, but may be severe. Exposure may occur via oral, mucosal, parental, or inhalation routes depending on the substance.
    E) OTHER: For further detailed information on N-benzylpiperazine (BZP), dimethylamylamine (DMAA), and paramethoxyamphetamine (PMA) see the following managements: N-BENZYLPIPERAZINE, DIMETHYLAMYLAMINE and PARAMETHOXYAMPHETAMINE.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE POISONING: All of these agents have the potential to cause either pleasant or frightening hallucinations. Early sympathetic effects include mydriasis, dizziness, tachycardia and hypertension. Bruxism is common, especially in MDMA users. Nausea and vomiting are common with peyote in recreational doses or doses used for religious ceremonies and often precede the hallucinations. PMA intoxications are characterized by hypoglycemia, hyperkalemia and QRS interval prolongation.
    2) SEVERE POISONING: Severe effects may include tachypnea, hyperthermia, delirium, psychosis, seizures, coma and ventricular dysrhythmias. Rhabdomyolysis and renal failure may rarely develop in patients with prolonged agitation, hyperthermia, coma or seizures. Elevated blood pressure can lead to intracerebral hemorrhages. MDMA can cause inappropriate antidiuretic hormone release, which combined with often excessive water consumption while dancing at night clubs or warehouse parties, can result in profound and potentially fatal hyponatremia.
    3) ADVERSE EFFECTS (CHRONIC): Long-term abusers commonly exhibit chronic toxicity. Cardiovascular toxicities can include necrotizing vasculitis of small to medium arteries and may affect any organ system, aortic and mitral valve insufficiency, cardiomyopathy and pulmonary hypertension. Neurologic toxicity can include stroke and cognitive decline, presumably due to direct neurotoxicity. Long-term users of MDMA exhibit permanent destruction of serotonergic neurons.
    4) ONSET: Approximately 30 to 45 seconds after ingestion (on an empty stomach) of ecstasy a "rush" can occur, which lasts 15 to 30 minutes. This is followed by a sense of clarity and feelings of happiness. A booster dose may be taken at this point, to prolong these feelings. Thirty minutes to 3 hours after the initial ingestion a "plateau" phase occurs in which repetitive or trance-like movements become extremely pleasurable. Rhabdomyolysis can develop during this phase due to extended activity. The "coming down" phase occurs 3 to 6 hours after the initial ingestion, and can lead to negative feelings or emotions (eg, depression, anxiety). Symptoms may persist for several days.
    5) NOTE: The term "Ecstasy" which is most often associated with MDMA has also been linked to gamma-hydroxybutyrate (GHB), 'liquid ecstasy'; and 'herbal ecstasy', an ephedrine-containing preparation. Because these agents are pharmacologically different, it is necessary to correctly identify the suspected agent(s).
    6) USUAL DOSING: A usual dose of Ecstasy taken by young adults/teens is 1 to 2 mg/kg body weight (125 to 180 mg). "Candyflipping" is the intentional combination of ecstasy with LSD. Another method of use is called "stacking" in which 3 or more tablets of MDMA are taken at once; or mixing MDMA with alcohol, marijuana or other drugs of abuse (eg, ketamine, GHB, cocaine) in order to modulate the high. "Stacking" can increase the risk of overdose since MDMA, acting as a stimulant, can mask the sedative effects of alcohol or opiates.
    7) ADULTERANTS: Adulteration of so called "Ecstasy" (MDMA) tablets has included oriental herbal ephedrine (Ma Huang), ketamine, paracetamol, and caffeine. Concoctions of BZP plus TFMPP have been reportedly sold as "Ecstasy" and produced sympathomimetic toxicity and dissociative-like symptoms. So called brand names do not guarantee purity or concentration.
    8) BODY PACKERS/BODY STUFFERS: Please refer to the appropriate management if body packing or body stuffing is known/suspected.

Vital Signs

    3.3.1) SUMMARY
    A) WITH POISONING/EXPOSURE
    1) Hypertension, tachycardia, and tachypnea are common. In severe cases, hyperthermia, hypotension, and cardiovascular collapse may occur.
    3.3.2) RESPIRATIONS
    A) WITH POISONING/EXPOSURE
    1) Increased respiratory rate is common (Melian et al, 2004; Henry et al, 1992; Watson et al, 1993).
    3.3.3) TEMPERATURE
    A) WITH POISONING/EXPOSURE
    1) SUMMARY
    a) Hyperthermia occurs in severe cases (Ghatol & Kazory, 2012; Vanden Eede et al, 2012; Melian et al, 2004; Caldicott et al, 2003; Refstad, 2003; Ginsberg et al, 1970; Brown et al, 1986; Russell et al, 1992; Woods & Henry, 1992; Singarajah & Lavies, 1992; Watson et al, 1993).
    1) Hyperthermia may be life threatening, and has been contributory to death (Iwersen & Schmoldt, 1996). Core temperatures above 42 degrees C may develop, and indicate a need for aggressive cooling measures and control of agitation (Chadwick et al, 1991; Screaton et al, 1992; Henry et al, 1992; Campkin & Davies, 1992; Logan et al, 1993; Tehan, 1993; Mueller & Korey, 1998).
    2) ETIOLOGY: Hyperthermia is a frequent clinical feature of MDMA toxicity with signs/symptoms similar to malignant hyperthermia which is biochemically caused by a rise of calcium ions in the myoplasm (Denborough & Hopkinson, 1997). An in-vitro muscle contracture study revealed a corresponding elevation of calcium within the cells following exposure to MDMA.
    3) INCIDENCE/FATALITY: In a retrospective chart review of 191 cases of MDMA exposure, 7 cases experienced hyperthermia with 1 death due to hyperthermia reported (Rella et al, 2000).
    4) COADMINISTRATION OF THC: In a study of 16 volunteers, the effects on heart rate and temperature were observed following MDMA (alone) or THC (alone) or a combination compared to placebo. Temperature was significantly higher in the MDMA-alone and MDMA + THC combination compared to THC alone and placebo. The addition of THC appeared to delay the onset and prolong the duration of temperature elevation compared to MDMA-alone. It was also found that epinephrine and norepinephrine levels were higher in MDMA + THC combination compared to THC-alone (Dumont et al, 2009).
    2) CASE REPORTS
    a) CASE SERIES/MDMA: In one case series, 6 patients developed severe hyperthermia following MDMA use. Three patients took MDMA during a rave party; the other 3 were exposed to MDMA outside of a rave setting and did not have vigorous muscle activity (seizures, dancing). The authors concluded that hyperthermia can occur in MDMA-exposed patients in a variety of settings. The following causes were proposed: hyperactivity and MDMA use during a rave party (high ambient temperatures, crowded conditions, dancing, and high fluid loss through sweating); muscle hyperactivity as a result of a seizure; direct effects of the drug on the serotonin, dopaminergic, and adrenergic systems; drug contaminants and coingestants (cocaine, methamphetamine, or other stimulant drugs); and individual idiosyncratic or genetic susceptibility (CYP2D6 polymorphism) to the effects of MDMA (Patel et al, 2005).
    b) CASE SERIES/PMA: Paramethoxyamphetamine (PMA), a methoxylated phenethylamine derivative that is structurally related to MDMA, produced severe hyperthermia in young adults; of the 7 cases reported all resulted in death (Refstad, 2003; Felgate et al, 1998).
    c) CASE REPORT/MDEA: Core body temperature was 42.5 degrees C in a young man exposed to methylene dioxyethylamphetamine (MDEA); hyperthermia (probably 5-HT mediated) was believed to be contributory to death. No relationship between high ambient temperature or intense physical activity with MDEA toxicity was noted in this case (Iwersen & Schmoldt, 1996).
    d) CASE REPORT/MIXED INGESTION: A young man died of disseminated intravascular coagulation, induced by hyperthermia following the combined ingestion of amphetamines (3,4-methylenedioxymethylamphetamine, 3,4-methylenedioxyamphetamine, and paramethoxyamphetamine). Postmortem serum PMA, MDMA, MDA, AMP levels were elevated (Dams et al, 2003).
    3.3.4) BLOOD PRESSURE
    A) WITH POISONING/EXPOSURE
    1) Hypertension is common (Lamberth et al, 2008; Melian et al, 2004; Russell et al, 1992; Henry et al, 1992; Logan et al, 1993).
    2) Hypotension and cardiovascular collapse may occur in severe poisoning (Henry et al, 1992; Campkin & Davies, 1992; Tehan, 1993; Watson et al, 1993).
    3.3.5) PULSE
    A) WITH POISONING/EXPOSURE
    1) Tachycardia is common (Melian et al, 2004; Henry et al, 1992; Screaton et al, 1992; Logan et al, 1993).

Heent

    3.4.2) HEAD
    A) WITH POISONING/EXPOSURE
    1) BRUXISM is anecdotally reported to be a very common effect of MDMA (Buchanan & Brown, 1988; Duxbury, 1993; McCann et al, 1996) and is likely caused by the interaction between dopaminergic and serotoninergic neurons. It can result in tooth damage following chronic use of MDMA (Dinis-Oliveira et al, 2010).
    2) TRISMUS is also reported (Duxbury, 1993; Jones et al, 1994; McCann et al, 1996). Bruxism also developed in 2 young adults after ingesting (1-(3-Trifluoromethylphenyl) piperazine (TFMPP) and 1-benzylpiperzine (BZP) (Wood et al, 2008).
    3) CHRONIC EFFECT: Idiopathic temporomandibular joint syndrome (TMJ) has been reported in part by the secondary effects of bruxism and trismus frequently observed following acute exposure (McCann et al, 1996).
    3.4.3) EYES
    A) WITH POISONING/EXPOSURE
    1) MYDRIASIS: Dilated pupils are virtually always present (Wood et al, 2007; Melian et al, 2004; Barrett & Taylor, 1993; Henry et al, 1992; Campkin & Davies, 1992; Ajaelo et al, 1998).
    a) TFMPP and BZP: Mydriasis developed in 3 young adults after ingesting 4 tablets containing 1-(3-trifluoromethylphenyl) piperazine (TFMPP) and 1-benzylpiperzine (BZP). In all cases, symptoms resolved by the following day (Wood et al, 2008).
    b) MDMA: Dilated and unreactive pupils were observed in a 11-month-old boy admitted with apparent seizures following the accidental ingestion of MDMA (confirmed by urine testing). The toddler recovered with supportive care (Garcia-Algar et al, 2005).
    2) KERATOPATHY: Eye pain, blurred vision, and diffuse, punctate epithelial erosions of the cornea have been reported in patients who ingested MDMA and then remained awake for long periods of time (O'Neill & Dart, 1993).
    3) NYSTAGMUS may develop (Caldicott et al, 2003; Brown & Osterloh, 1987).
    3.4.5) NOSE
    A) WITH POISONING/EXPOSURE
    1) DRYNESS: Mucous membranes are generally dry.

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) HYPERTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) SUMMARY
    1) Systolic and diastolic hypertension are common (Davies et al, 2014; Lamberth et al, 2008; Gee et al, 2005; Chang et al, 2005; Melian et al, 2004; Russell et al, 1992; Henry et al, 1992; Logan et al, 1993).
    b) MDMA
    1) CASE SERIES: In a double-blind, placebo-controlled trial, 8 healthy adults with occasional MDMA use (described as once or twice per month) were given either MDMA (0.5 and 1.5 mg/kg) or placebo with regular measurement of heart rate and blood pressure and an echocardiographic evaluation. Each patient was also given dobutamine (5, 20 and 40 mcg/kg) in a preliminary session, and echocardiographic effects were measured. It was found that MDMA (1.5 mg/kg) and dobutamine (20 and 40 mcg/kg) increased heart rate by 28 beats/min, systolic and diastolic blood pressure by 25 mmHg and 7 mmHg, respectively, and cardiac output (an increase of 2 L/min {same finding as 20 mcg/kg/min of dobutamine}); a MDMA dose of 0.5 mg/kg had NO cardiac effect. The authors concluded that a modest dose of MDMA produced dose-dependent increases in heart rate and blood pressure and myocardial oxygen consumption with no measurable inotropic effect (Lester et al, 2000).
    2) CASE REPORTS
    a) PEDIATRIC EXPOSURE: An 8-month-old boy presented with generalized seizure activity, tachycardia (210 beats/min), hypertension (125/70 mm Hg), and hyperthermia (38.9 degrees C) after accidentally ingesting one Ecstasy tablet. Treatment with benzodiazepines, body cooling and fluids resulted in a resolution of all abnormal parameters 6 hours after admission (Eifinger et al, 2008).
    b) PEDIATRIC EXPOSURE: A 2-year-old boy presented with normal vital signs after ingesting an unknown amount of MDMA, but later developed systolic hypertension of 115/61. The patient tested positive for MDMA the morning after hospital admission; however, the length of time between MDMA exposure to clinical presentation was unknown (Feldman & Mazor, 2007).
    c) PMA
    1) PARAMETHOXY-AMPHETAMINE (PMA): A 20-year-old man was admitted unconscious after ingesting PMA (confirmed by laboratory analysis) with a blood pressure of 171/148 mmHg, temperature of 42.8 degrees C, heart rate 90 beats/min and respiratory rate of 40 breaths/min. His course was complicated by hyperthermia, persistent coma, shock, hyperkalemia and severe coagulopathy and he died 10 days after admission (Lamberth et al, 2008).
    B) TACHYCARDIA
    1) WITH POISONING/EXPOSURE
    a) SUMMARY
    1) Tachycardia is common (Davies et al, 2014; Poon et al, 2010; Ovaska et al, 2008; Wood et al, 2007; Lamberth et al, 2008; Wood et al, 2008; Gee et al, 2005; Chang et al, 2005; Melian et al, 2004; Henry et al, 1992; Russell et al, 1992; Screaton et al, 1992; Jones et al, 1994; Engebretsen & Harris, 2001).
    b) MDMA
    1) Tachycardia, sometimes severe (160 beats/min or more) has been reported in severe and fatal overdoses of MDMA and PMA (Caldicott et al, 2003; Refstad, 2003; Henry et al, 1992; Campkin & Davies, 1992; Screaton et al, 1992).
    a) MDMA: Sinus tachycardia (190 beats/min) and a blood pressure of 100/50 mmHg were observed in an 11-month-old boy admitted with apparent seizures following the accidental ingestion of MDMA (confirmed by urine testing). The toddler recovered with supportive care (Garcia-Algar et al, 2005).
    2) INCIDENCE: In a retrospective chart review of 191 cases of MDMA exposure, tachycardia (22%) was the most commonly reported symptom (Rella et al, 2000).
    3) CASE REPORT/PEDIATRIC: An 8-month-old boy presented with generalized seizure activity, tachycardia (210 beats/min), hypertension (125/70 mm Hg), and hyperthermia (38.9 degrees C) after accidentally ingesting one Ecstasy tablet. Treatment with benzodiazepines, body cooling and fluids resulted in a resolution of all abnormal parameters 6 hours after admission (Eifinger et al, 2008).
    4) COADMINISTRATION OF THC
    a) In a study of 16 volunteers, the effects on heart rate and temperature were observed following MDMA (alone) or THC (alone) or a combination compared to placebo. In each setting, heart rate was increased with the administration of these agents compared to placebo. However, the coadministration of tetrahydrocannabinol (THC) and MDMA produced the highest increase in heart rate compared to MDMA-alone or THC-alone; the effects appear to be additive (Dumont et al, 2009).
    c) TFMPP
    1) Tachycardia and chest discomfort were reported in a 28-year-old man, with a history of substance abuse, after ingesting 1-(3-trifluoromethylphenyl)-piperazine (TFMPP; confirmed by laboratory analysis). Symptoms resolved spontaneously and the patient was discharged (Poon et al, 2010).
    C) VENTRICULAR HYPERTROPHY
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: In a matched, retrospective study using medical examiner data, 27 MDMA positive deaths and 135 matched MDMA negative deaths (mean age 20 years; range 16 to 33 years) were evaluated to determine if the use of MDMA is associated with myocardial hypertrophy. Multivariate analysis showed that MDMA-positive fatalities were more likely to have an enlarged heart (OR=18.3; 95% CI=3.6-1.6) with a mean heart weight of 315.7 and 277.2 g for MDMA-positive and MDMA-negative fatalities, respectively (Diff=38.5 g; 95% CI=18.3-8.7) (Patel et al, 2005a).
    D) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) Hypotension and cardiovascular collapse may occur in severe poisoning (Davies et al, 2014; Henry et al, 1992; Chadwick et al, 1991; Campkin & Davies, 1992; Tehan, 1993; Watson et al, 1993).
    E) CONDUCTION DISORDER OF THE HEART
    1) WITH POISONING/EXPOSURE
    a) SUMMARY: Cardiac dysrhythmias are common in patients with severe toxicity following MDMA overdose (Henry et al, 1992) or other hallucinogenic amphetamines (Lamberth et al, 2008; Tehan, 1993).
    b) MDMA
    1) ADULT/ADOLESCENTS
    a) CASE REPORT: A 19-year-old man presented in a coma with seizures, and hyperthermia (temperature 42.5 degrees C) 2 hours after ingesting ethanol and 3 ecstasy tablets (total dose unknown). Physical examination revealed bilateral mydriasis, myoclonus, a systolic murmur, and a thrill over the femoral arteries. He had a heart rate of 95 beats/min and arterial blood pressure of 125/45 mm Hg. An ECG revealed broad QRS complexes with short QT interval and a long PR interval, indicating hyperkalemia. Despite supportive therapy, including treatment with active cooling measures, sedation with midazolam, IV calcium gluconate, sodium bicarbonate, glucose, and insulin, he developed bradycardia and a P wave asystole. During cardiopulmonary resuscitation, he developed ventricular fibrillation that was successfully defibrillated. Laboratory results revealed hyperkalemia (peak potassium, 9.4 mmol/L after 30 minutes in the ICU), rhabdomyolysis (peak CK, 12,509 Units/L), myocardial damage, and syndrome of inappropriate antidiuretic hormone. He experienced another cardiac arrest after arrival in the ICU. He had pulmonary edema and pulseless electrical activity, alternating with sinus bradycardia (30 beats/min). Resuscitation efforts were unsuccessful and he died 2 hours later. It was suggested that the cause of death was a rapidly evolving hyperkalemia due to rhabdomyolysis (Vanden Eede et al, 2012).
    b) CASE REPORT: A 20-year-old man developed a wide complex tachycardia (180 to 200 beats/min) with hypotension (80 mm Hg systolic) after ingesting MDEA (Tehan, 1993).
    c) CASE REPORT: A 34-year-old man with a history of Wolff-Parkinson-White syndrome complained of palpitations after using MDMA (Suarez & Riemersma, 1988). The next morning he was found gasping and convulsing and was in ventricular fibrillation on arrival of the paramedics and could not be resuscitated.
    d) CASE REPORT: An 18-year-old collapsed after ingesting 150 mg of MDMA and an unknown amount of ethanol. She was in ventricular fibrillation on arrival of paramedics and could not be resuscitated (Dowling et al, 1987).
    e) CASE REPORT: A 16-year-old boy became bradycardic (48 beats/min) approximately 3 hours after having been found undressed in a local forest. The patient subsequently developed ventricular fibrillation and died, despite resuscitative efforts. Further investigation revealed that the patient had ingested 7 tablets suspected to be MDMA; however, lab analysis prior to death and at postmortem showed PMA concentrations of 4.8 mcmol/L and 5.0 mcmol/L, respectively (Refstad, 2003).
    f) CASE REPORT: A 16-year-old boy presented with hypotension and ventricular tachycardia. His course was complicated by severe hyperthermia, seizures, coma, metabolic acidosis, hypoglycemia, intractable hypotension, disseminated intravascular coagulation, rhabdomyolysis, and renal failure. He died 6 days after admission. MDMA was detected in his urine (Watson et al, 1993).
    g) CASE REPORT: A 19-year-old man became agitated, confused, hyperthermic, hypotensive (70/50 mmHg), and tachycardic (180 BPM) after ingesting 12 tablets of MDMA and dancing vigorously in a hot environment. After presentation to the hospital, the patient had a cardiac arrest with successful resuscitation. He became comatose necessitating mechanical ventilation. Laboratory data showed an elevated potassium level (6.3 mEq/L) and a pH level of 6.99 with bicarbonate level of 19 mEq/L (indicative of metabolic acidosis). The patient subsequently died after a second, asystolic cardiac arrest. It is believed that the rapid increase in extracellular potassium in combination with development of severe metabolic acidosis exacerbated the patient's cardiovascular effects resulting in asystolic death (Ravina et al, 2004).
    2) PEDIATRIC
    a) CASE REPORT: Approximately 40 minutes after ingesting a portion of an ecstasy pill, a 14-month-old child developed a generalized convulsion with opisthotonos, hyperthermia (38 degrees C), hypertension, tachycardia (130 beats/min), ventricular extrasystoles, tachypnea (50 breaths/min), and mydriasis. The urine levels of amphetamine/metamphetamine were greater than 16 mg/L 5 hours after ingestion. The serum level of MDMA was 0.591 mg/L 8 hours after ingestion. He continued to have hypertension, tachycardia and long periods of trigeminy, without hemodynamic repercussion during the first 12 hours. Following supportive care, the child recovered with no further sequelae (Melian et al, 2004).
    c) PMA
    1) PARAMETHOXY-AMPHETAMINE: A 20-year-old man was admitted unconscious after ingesting PMA (confirmed by laboratory analysis) with hypertension, tachycardia and ECG showing a prolonged QRS interval of 160 ms (reference range: less than 100 ms). The patient had a complex course which included hyperthermia, persistent coma, shock, hyperkalemia, hypoglycemia, rhabdomyolysis and severe coagulopathy and he died 10 days after admission (Lamberth et al, 2008).
    F) PERIPHERAL ISCHEMIA
    1) WITH POISONING/EXPOSURE
    a) Diffuse vascular spasm, resulting in limb ischemia, gangrene, and amputation have been reported following DOB ingestions (Bowen et al, 1983).
    G) DEAD - SUDDEN DEATH
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 34-year-old man with untreated Wolff-Parkinson-White syndrome died suddenly after use of MDMA. The MDMA level was 0.2 mg/dL in blood and 5 mg/dL in urine (Suarez & Riemersma, 1988).
    b) CASE REPORT: A 25-year-old man complained of pleuritic chest pain. He died while driving the following day. Autopsy revealed a 75% lesion of the left descending and left circumflex coronary artery and an area of pinpoint lumen in the right coronary artery without evidence of myocardial infarction. Postmortem screening revealed blood level of butalbital (0.8 mg/L) and MDEA (0.95 mg/L) (Dowling et al, 1987).
    H) DISSECTION OF AORTA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A previously healthy 29-year-old man died of a Type 1 aortic dissection (involving the superior and mesenteric arteries with resultant bowel ischemia), which had resulted from cardiac tamponade, approximately 48 hours after ingesting an ecstasy tablet and a large quantity of alcohol at a rave party. The patient initially felt well and experienced his first onset of symptoms (shortness of breath, abdominal pain, diarrhea, and vomiting) 36 hours after ingestion. The patient was normotensive with normal hematologic laboratory values with a WBC of 17,800/mL, and was thought to have gastroenteritis after passing a loose bloody stool. The patient refused further treatment and left the ED to return 8 hours later appearing seriously ill. The patient died 5 hours later despite aggressive resuscitation measures. Autopsy results revealed no evidence of hypertension, cardiovascular disease, or other risk factors associated with aortic dissection. Postmortem blood concentration of MDMA was 0.1 mg/L; the toxic screen was negative for alcohol or other agents at the time of autopsy (Duflou & Mark, 2000).
    I) HEART FAILURE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 23-year-old man developed tachycardia (160 beats/min) with hypertension (167 mmHg systolic) followed by hypotension (mean arterial pressure 57 mmHg) after ingesting MDMA. Pulmonary artery catheterization revealed low cardiac output (2.9 L/min) and pulmonary artery occlusion pressure (4 mmHg). Fluid resuscitation improved the pulmonary artery occlusion pressure without affecting cardiac output (Logan et al, 1993).
    J) CHEST PAIN
    1) WITH POISONING/EXPOSURE
    a) Chest pain which was not believed to be of cardiac origin has been reported in the setting of ecstasy use and physical exertion. Associated effects included mild tachycardia and hypertension, anxiety and diaphoresis (Rittoo et al, 1992).
    b) CASE REPORT: A 21-year-old man with no risk factors for ischemic heart disease presented agitated with chest pain, tachycardic (126 beats/min) and hypertensive (BP 213/109 mm Hg) a few hours after ingesting 3 tablets of "Head Candy" (diphenylprolinol plus glaucine). A toxicology screen identified a blood concentration of diphenylprolinol 0.17 mg/L and glaucine 0.10 mg/L. The patient made a full recovery by the next day with supportive treatment (Lidder et al, 2008).
    K) MYOCARDIAL INFARCTION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 27-year-old man presented to the ED with agitation, diaphoresis, nausea, epigastric and chest pain that began approximately 3 hours after consuming one bottle of whiskey and one-half of a tablet of MDMA. He had no prior history of chest pain or heart disease. An ECG showed ectopic atrial rhythm with ST elevation, Q wave, and T inversion in the inferior leads; CK MP and troponin I were elevated (peak 85.6 Units/L and 18.8 mg/dL, respectively). Plasma MDMA level was 1,100 ng/mL and a urine MDMA level was 96,800 ng/mL. Cardiac catheterization, performed 8 hours after the initial onset of chest pain, showed a thrombus in the right coronary artery. He was treated with an intravenous glycoprotein IIb/IIIa inhibitor and an intracoronary injection of urokinase and recovered. It is speculated that ingestion of MDMA produced coronary artery vasospasm and subsequent thrombus formation (Lai et al, 2003).
    b) CASE SERIES: Three patients presented with delayed acute coronary syndrome after ecstasy ingestion, including 2 patients with ST elevation myocardial infarction (Hoggett et al, 2012).
    1) The first patient, a 20-year-old man, presented with chest pain 3 days after receiving 10 Ecstasy tablets. Laboratory results revealed a Troponin I of 0.22 mcg/L (reference less than 0.04 mcg/L) at 2 hours after onset of the chest pain and a peak of 1.3 mcg/L at 24 hours. His echocardiogram was normal and he recovered without further sequela. Another patient, a 22-year-old man, presented with a 12-hour history of severe crushing central chest pain, nausea, dyspnea, and clamminess, 48 hours after ingesting 2 Ecstasy tablets. His vital signs included a heart rate of 80 beats/min and a blood pressure of 128/70 mmHg. An ECG revealed an acute left bundle branch block and concordant ST elevation through the lateral leads. He was treated with supportive care, including aspirin, clopidogrel, heparin, and morphine. In another ECG, 45 minutes later, reversal of the left bundle branch block was observed, but the ST elevation in leads V2-V6 persisted. A primary coronary angiography revealed structurally normal coronary arteries with sluggish flow secondary to thrombosis. Laboratory results revealed a Troponin I peak of 10 mcg/L. His symptoms improved and he was discharged home on clopidogrel and diltiazem (Hoggett et al, 2012).
    2) A 23-year-old woman, with a history of occasional Ecstasy and cocaine use, presented with insomnia and chest pain the day after ingesting 7 Ecstasy tablets over a 16-hour period with alcohol. Her vital signs included a heart rate of 123 beats/min and a blood pressure of 155/95 mmHg. He was treated with supportive care, including aspirin, diazepam, and sublingual nitrates and an ECG revealed sinus tachycardia. Laboratory results showed the Troponin I of 0.11 mcg/L. On day 2, normal wall motion with mild global ventricular systolic dysfunction were observed in an echocardiogram. On day 3, she developed severe central crushing chest pain and another ECG showed inferolateral ST segment elevation. She received aspirin, metoprolol, and ramipril at this time and her Troponin I peaked at 6.1 mcg/L. On day 4, she underwent coronary angiography after the recurrence of pain and ST elevation. Normal coronary arteries with no evidence of dissection was observed. She received aspirin, metoprolol, ramipril and was discharged home. On a 1-month follow-up, an echocardiogram was normal. Persistent inferior T wave inversion was observed on a follow-up ECG (Hoggett et al, 2012).
    L) MYOCARDITIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 16-year-old boy developed spontaneous pneumomediastinum and myocarditis after Ecstasy ingestion. A CT scan revealed pneumomediastinum with mediastinal air from the base of the heart up to the neck with diffusion to the right axilla. His troponin I level increased to 7.1 ng/mL with a creatine kinase level of 1023 Units/L and a creatine kinase MB of 53.8 Units/L. ECG revealed sinus tachycardia with slight, diffuse ST segment elevation, suggestive of pericarditis. Cardio-magnetic resonance imaging showed an inferolateral subepicardial delayed enhancement suggestive of myocarditis. Following supportive therapy, he recovered without further sequelae (Mortelmans et al, 2005).
    M) HEART VALVE DISORDER
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 33-year-old man, a chronic smoker and ecstasy user (several pills per week since the age of 17 years), presented with shortness of breath and chest pain. Physical examination revealed a systolic and diastolic mitral murmur and a mean blood pressure of 106/70 mmHg. An ECG revealed sinus rhythm (HR 91 beats/min) with a right bundle branch block. An echocardiography showed a slight left ventricular dilation (end-diastolic diameter of 55 mm), an ejection fraction of 55%, and a left atrial enlargement (area of 42 cm(2)) thick mitral valve with reduced opening of the mitral valve leaflets. There were moderate to severe mitral stenosis features with severe mitral regurgitation; other valves were normal. He also had pulmonary hypertension (40 mmHg) without right heart failure. Based on macroscopic analysis, a possible cardiac tumor was considered and a surgical procedure was performed. Because the valve and chordae tendineae were markedly thickened and fibrous, the chordae tendineae were retracted and a mitral valve replacement with a mechanical Saint Jude prosthesis was performed. Histopathology of the mitral valve revealed a huge fibrous thickening of the endocardium, with layers of collagen bundles and spindle cells. He did not have any common diseases (eg, carcinoid syndrome, Osler endocarditis, rheumatic fever, autoimmune disease) or used other drugs (eg, ergot derivatives, appetite-suppressants) that could have affected the valves. A month later, he presented with chest pain, fatigue, and weight loss. A paraprosthetic leak with prosthesis dehiscence was observed during echocardiography. After surgical fixation of his prosthesis, he recovered with no further complications (Montastruc et al, 2012).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) HYPERVENTILATION
    1) WITH POISONING/EXPOSURE
    a) An increased respiratory rate is often present (Lamberth et al, 2008; Henry et al, 1992; Watson et al, 1993).
    B) ACUTE LUNG INJURY
    1) WITH POISONING/EXPOSURE
    a) SUMMARY
    1) Pulmonary edema and ARDS may occur in severe intoxications (Lamberth et al, 2008; Chang et al, 2006; Curtis et al, 2003; Brown et al, 1986; Simpson & Rumack, 1981; Brown & Osterloh, 1987; Walubo & Seger, 1999) and has resulted in death (Albertson et al, 1995).
    b) MDMA
    1) CASE REPORT: Hyponatremia (initial serum sodium 124 mmol/L) with secondary cerebral edema and acute lung injury developed in a 17-year-old boy following suspected MDMA use. Clinical findings included coma, increased oxygen demands, and a chest x-ray that showed marked fluffy infiltrates consistent with acute lung injury. Despite aggressive supportive care the patient did not improve and died following withdrawal of life support. Toxicology screen was positive for MDMA and chlorxylenol (Engebretsen & Harris, 2001).
    2) CASE REPORT: A 23-year-old woman experienced generalized seizures and became comatose, necessitating mechanical ventilation, approximately 6 hours after ingesting 1 MDMA tablet while dancing at a discotheque. Following admission to the intensive care unit of the hospital, the patient developed severe hypoxemic respiratory failure diagnosed as acute respiratory distress syndrome. The patient became alert approximately 3.5 days postadmission and was discharged home with no sequelae (Ben-Abraham et al, 2003).
    c) PMA
    1) PARAMETHOXY-AMPHETAMINE: A 20-year-old man was admitted unconscious after ingesting PMA (confirmed by laboratory analysis) with hypertension, tachycardia and an ECG showing a prolonged QRS interval. Following intubation the patient had persistent hypoxia and an x-ray showed extensive bilateral airspace consolidation. The patient had an ongoing complex course which included hyperthermia, persistent coma, shock, hyperkalemia, rhabdomyolysis and severe coagulopathy. Five days after admission a head CT showed extensive cerebral edema. The patient died 10 days after admission (Lamberth et al, 2008).
    C) MEDIASTINAL EMPHYSEMA
    1) WITH POISONING/EXPOSURE
    a) SUMMARY: Infrequent reports of spontaneous pneumomediastinum have been reported in the literature following MDMA use (Levine et al, 1993; Quin et al, 1999).
    b) CASE REPORT: Spontaneous pneumomediastinum without associated esophageal perforation developed in a 17-year-old with protracted vomiting following ingestion of MDMA (Levine et al, 1993).
    c) CASE REPORT: A 25-year-old man developed spontaneous pneumomediastinum following the ingestion of 3 ecstasy tablets 14 hours previously. The patient denied injury or trauma to the chest. The patient was observed for 4 days and discharged with no sequelae. A temporal association between ecstasy ingestion and subsequent pneumomediastinum was suggested by the authors (Quin et al, 1999).
    d) CASE REPORT: A 16-year-old boy developed spontaneous pneumomediastinum and myocarditis after ecstasy ingestion. A CT scan revealed pneumomediastinum with mediastinal air from the base of the heart up to the neck with diffusion to the right axilla. His troponin I level increased to 7.1 ng/mL with a creatine kinase level of 1023 Units/L and a creatine kinase MB activity of 53.8 Units/L. ECG revealed sinus tachycardia with slight, diffuse ST segment elevation, suggestive of pericarditis. Cardio-magnetic resonance imaging showed an inferolateral subepicardial delayed enhancement suggestive of myocarditis. Following supportive therapy, he recovered without further sequelae (Mortelmans et al, 2005).
    D) PULMONARY ASPIRATION
    1) WITH POISONING/EXPOSURE
    a) Aspiration pneumonitis has been reported in patients who developed seizures and coma after MDMA use (Jones et al, 1994; Lehmann et al, 1995).
    E) ACUTE RESPIRATORY INSUFFICIENCY
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Apnea, respiratory acidosis, and central nervous system depression were reported in a 30-year-old man found at home following a massive ingestion of ecstasy (50 tablets), along with oxazepam and alcohol. Symptoms resolved, and the patient had a complete recovery following gastric decontamination and supportive care (Ramcharan et al, 1998).
    F) BRONCHOSPASM
    1) WITH POISONING/EXPOSURE
    a) An asthma related death secondary to the use of MDMA has been reported (Albertson et al, 1995).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) PSYCHOMOTOR AGITATION
    1) WITH POISONING/EXPOSURE
    a) Severe excitement and agitation can develop and is relatively common (Logan et al, 1993; Tehan, 1993; Watson et al, 1993).
    b) INCIDENCE: In a retrospective chart review of 191 cases of MDMA exposure, agitation (19%) was the second most commonly reported effect (Rella et al, 2000).
    1) A hospital in New Zealand reviewed 80 cases of 1-benzylpiperazine (BZP) exposures and found that agitation was reported in approximately 27% of cases (Gee et al, 2005).
    c) CASE REPORT: Hallucinations, confusion and agitation were reported in a 32-year-old woman who ingested approximately 200 mg of MDMA. Hypotension, tachycardia, hyperthermia, coma, pulmonary edema, and hypoxia rapidly ensued. Over the next 5 days complications developed, including rhabdomyolysis, leukocytosis, coagulopathy, toxic hepatitis, and a herpetic-like rash. Serum MDMA levels were 7 mcg/mL (Brown et al, 1986).
    B) SEIZURE
    1) WITH POISONING/EXPOSURE
    a) MDMA
    1) Seizures are a common complication of severe toxicity (Vanden Eede et al, 2012; Patel et al, 2005; Barrett & Taylor, 1993; Henry et al, 1992; Woods & Henry, 1992; Singarajah & Lavies, 1992; Buhrich et al, 1983; Chadwick et al, 1991; Shulgin, 1981; Russell et al, 1992; Campkin & Davies, 1992; Watson et al, 1993; Lehmann et al, 1995; Ajaelo et al, 1998).
    2) CASE REPORT/PEDIATRIC: Approximately 40 minutes after ingesting a portion of an ecstasy pill, a 14-month-old child developed a generalized convulsion with opisthotonos, hyperthermia (38 degrees Celsius), hypertension, tachycardia (130 beats/min), ventricular extrasystoles, tachypnea (50 breaths/min), and mydriasis. The urine levels of amphetamine/methamphetamine were greater than 16 mg/L 5 hours after ingestion. The serum level of MDMA was 0.591 mg/L 8 hours after ingestion. He continued to have hypertension, tachycardia and long periods of trigeminy, without hemodynamic repercussion during the first 12 hours. Following supportive care, the child recovered with no further sequelae (Melian et al, 2004).
    3) CASE REPORT/PEDIATRIC: A 20-month-old child accidentally ingested a MDMA tablet and began seizing along with consciousness disturbances approximately 50 minutes later. In the ED, the infant was restless, febrile, hypertensive, and tachycardic. With cooling, hydration, and supportive treatment, he made a full recovery 2 days later (Chang et al, 2005).
    b) DOB
    1) CASE SERIES: After ingesting a "hallucinogen LSD-like" drug, 2 men developed a rapid onset of hallucinations (within 15 minutes), vomiting, and coma. The first subject, a 28-year-old (body mass 113 kg) survived but developed serious convulsions. The second subject, a 29-year-old man (body mass 65 kg) died 6 days postingestion after developing generalized convulsions and metabolic acidosis (pH 6.6). Although immunoassay for amphetamines was negative in urine specimens, GC-MS showed 2,5-dimethoxy-4-bromoamphetamines (DOB) in the gastric and urine samples of both individuals. DOB serum concentration was 13 ng/mL for the surviving patient and 19 ng/mL for the deceased patient, respectively (Balikova, 2005).
    c) DOC
    1) CASE REPORT: A 20-year-old man collapsed and had tonic-clonic seizures after presumably ingesting DOI (2,5 dimethoxy-4-iodoamphetamine) at a rave party. He required intubation in the field and was admitted comatose with a Glasgow Coma Score of 3/15. However, he had normal reflexes and a brain CT was normal. He also developed tachycardia (152 beats/min), metabolic acidosis, hypotension and an elevated creatine kinase (peak: 4924 International Units/L) with no evidence of renal failure. Following supportive care, including IV hydration and prophylactic antibiotics, the patient was extubated the next day and discharged to home with no permanent sequelae. Serum and urine samples confirmed the presence MDMA and DOC (2,5 dimethoxy-4-chloroamphetamine), which could not be quantified (Ovaska et al, 2008).
    d) BROMO-DRAGONFLY
    1) 1-(8-bromobenzo[1,2-b; 4,5-b']difuran-4-yl)-2-aminopropane: An 18-year-old man presented with hallucinations, agitation and 2 self-terminating seizures as a result of ingesting Bromo-dragonfly and an unknown white powder 8 hours prior to presentation. Shortly after arrival, the patient had a self-terminating seizure with apnea lasting over 1 minute, followed by another generalized seizure treated with lorazepam. The patient was intubated due to decompensating respiratory function; but was successfully extubated 24 hours later. Toxicological screen revealed a Bromo-dragonfly concentration of 0.95 ng/ml, ketamine and cannabis (Wood et al, 2009).
    C) COMA
    1) WITH POISONING/EXPOSURE
    a) SUMMARY
    1) Coma may develop in severe cases (Vanden Eede et al, 2012; Lamberth et al, 2008; Campkin & Davies, 1992; Watson et al, 1993; Lehmann et al, 1995).
    b) MDMA
    1) CASE REPORT: A 30-year-old man was found at home in a coma, apneic, and had symmetrical convulsions following a massive ingestion of 50 tablets of ecstasy along with oxazepam and alcohol. Treatment included gastric decontamination and supportive care; recovery was complete within 2 days of ingestion (Ramcharan et al, 1998).
    2) CASE REPORTS: Coma was reported in 3 patients who ingested up to 3 MDMA tablets. The duration of coma ranged from approximately 1 to 3.5 days. The patients gradually recovered without neurologic sequelae (Ben-Abraham et al, 2003).
    3) CASE SERIES: After ingesting a "hallucinogen LSD-like" drug, 2 men developed a rapid onset of hallucinations (within 15 minutes), vomiting and coma. The first subject, a 28-year-old (body mass 113 kg) survived, but developed serious convulsions. The second subject, a 29-year-old man (body mass 65 kg) died 6 days postingestion after developing generalized convulsions and metabolic acidosis (pH 6.6). Although an immunoassay for amphetamines was negative in urine specimens, GC-MS showed 2,5-dimethoxy-4-bromoamphetamines (DOB) in the gastric and urine samples of both persons. DOB serum concentration was 13 ng/mL for the surviving patient and 19 ng/mL for the deceased patient, respectively (Balikova, 2005).
    c) PMA
    1) PARAMETHOXY-AMPHETAMINE: A 20-year-old man was admitted unconscious after ingesting PMA (confirmed by laboratory analysis) with hypertension, tachycardia and an ECG showing a prolonged QRS interval. The patient had a complex course which included hyperthermia, persistent coma, shock, hyperkalemia, rhabdomyolysis and severe coagulopathy. Five days after admission, the pupils were sluggish and the gag and cough reflex were absent. Laboratory studies included an ammonia level of 219 micromol/L (reference range: 10 to 50). A head CT showed extensive cerebral edema and the patient died 10 days after presentation (Lamberth et al, 2008).
    D) ISCHEMIC STROKE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 30-year-old woman complained of right sided weakness after an alcohol binge and ingesting ecstasy the night prior. Initial brain CT revealed left middle cerebral artery (MCA) infarction. Two weeks after symptom onset, a 4 vessel catheter angiography was performed, showing persistent left MCA stenosis. Follow-up studies at 3 months were completely normal. Amphetamine-induced vasculitis or vasospasm is the suspected pathogenesis of ischemic stroke in this patient (DeSilva et al, 2007).
    E) CEREBRAL THROMBOSIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 22-year-old woman developed severe headache, photophobia, nausea, transient right hemisensory loss and dysphasia secondary to cerebral venous sinus thrombosis after ingesting a tablet of MDMA and dancing for 8 hours without drinking any fluids (Rothwell & Grant, 1993).
    F) CEREBRAL HEMORRHAGE
    1) WITH POISONING/EXPOSURE
    a) COMBINED INGESTION
    1) CASE REPORT/MDA and 2C-I: A 39-year-old-woman presented with rapidly diminishing mental status, agitation, hypothermia, emesis, urinary incontinence, severe hypertension, vasoconstriction and extensor posturing after ingesting MDA (3,4-methylenedioxyamphetamine) and 2C-I (2,5-dimethoxy-4-iodophenethylamine). Urinary drug concentrations were MDA 5.56 mg/L and 2C-I 0.311 mg/L. A head CT showed massive intraventricular hemorrhaging and underlying Moyamoya. One month after ingestion, the patient developed sympathetic storming and seizures. A tracheostomy was placed for long-term mechanical ventilation. After 4 months in the hospital, the patient was transferred to a skilled nursing facility with quadriplegia and had a modest improvement in mental status (ie, she could follow commands), but could not speak. She remained severely disabled and required total care (Drees et al, 2009).
    b) OTHER
    1) CASE REPORT: A 25-year-old woman developed subarachnoid hemorrhage after taking 2.5 ecstasy tablets. Angiography revealed a left posterior communicating artery aneurysm (Gledhill et al, 1993).
    2) Four patients developed intracerebral hemorrhage related to the ingestion of ecstasy or amphetamine, 3 of whom had normal intracranial vessels on angiography (Silve & Harries, 1992).
    G) CEREBRAL INFARCTION
    1) WITH POISONING/EXPOSURE
    a) 4-BROMO-2,5-DIMETHOXYPHENETHYLAMINE (2C-B): A 43-year-old woman with seronegative polyarticular arthritis, obesity and hypothyroidism developed severe, pulsatile headaches with confusion 48 hours after ingesting 2C-B. She had progressive upper extremity weakness and was admitted to the hospital with profound quadriparesis with diffuse hyperreflexia and encephalopathy 3 weeks later. An MRI of the brain revealed diffusion-positive lesions with bilateral ischemia and a magnetic resonance angiography showed diffuse arterial irregularities. A cerebral angiography confirmed focal vascular narrowing in small, medium, and large caliber vessels and the cortical vessels showed no evidence of vasculitis. Serology testing was negative for infectious or autoimmune etiologies. One month after exposure, the patient had no spontaneous movements of her lower extremities and the upper extremities were limited to a shoulder shrug and she remained in an abulic state with significant perseveration. High-dose corticosteroid therapy was started with no clinical improvement. At 6 months, the patient had minimal clinical improvement; the patient could speak in brief sentences and move her hands. No other individuals that took 2C-B during this party developed these symptoms and this case may represent an idiosyncratic response resulting in diffuse cerebral vasculopathy (Ambrose et al, 2010).
    H) CEREBRAL ARTERY OCCLUSION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 35-year-old man developed a large cerebral infarction with dense right hemiparesis and dysphasia 36 hours after using MDMA (Manchanda & Connolly, 1993).
    b) CASE REPORT: A 28-year-old man developed cerebral infarction and right sided weakness 1 hour after using MDMA (Hanyu et al, 1995).
    I) CEREBRAL EDEMA
    1) WITH POISONING/EXPOSURE
    a) Cerebral edema has been found at autopsy (Chadwick et al, 1991; Parr et al, 1997; Walubo & Seger, 1999).
    b) Cerebral edema has also been reported in patients with severe hyponatremia and SIADH after MDMA abuse (Ghatol & Kazory, 2012; Holden & Jackson, 1996; Ajaelo et al, 1998; Engebretsen & Harris, 2001).
    c) CASE REPORT: A 27-year-old man presented to the ED comatose and tachycardic (112 beats/min) after injecting himself with approximately 0.5 g of heroin and approximately 1 g of cocaine and ingesting 3 tablets of MDMA (300 mg). Lab evaluation showed that the patient had rhabdomyolysis, lactic acidosis, hypoglycemia, and progressive fulminant hepatic failure. Liver transplantation was declined by the transplant surgeon due to the patient's history of illicit drug use. Despite continuous venovenous hemofiltration, arterial hyperammonemia occurred followed by grade IV encephalopathy. A cranial CT scan showed cerebral edema and transtentorial herniation. Due to possible irreversible neurological deterioration, experimental therapy using a fractionated plasma separation and adsorption system (FPSA/Prometheus), a hemodialysis-based system was initiated. Within 12 hours of FPSA initiation, the patient's arterial ammonia levels normalized. Within 12 hours of the second treatment period, the intracranial pressure fell from 47 mmHg initially to 27 mmHg. A repeat cranial CT scan, performed 4 days after the initial scan, showed that the cerebral edema was greatly reduced. The encephalopathy gradually resolved and the patient recovered following neurological rehab to treat sequelae of herniation (dysphagia, dysarthria, motor weakness, gait disturbances) and was discharged to home 6 weeks later to live independently (Kramer et al, 2003).
    d) CASE REPORT: A 20-year-old woman presented with an altered mental status and rapidly progressive drowsiness after ingesting 2 tablets of double-stack Ecstasy (273 mg each) a few hours prior to admission. She was hyperthermic (38.6 degrees C) and unable to respond to verbal or painful stimuli. Laboratory results showed hyponatremia (sodium 123 mmol/L) and lactic acidosis (lactic acid 6.2 mmol/L). A head CT scan showed severe parenchymal edema with effacement of basal cisterns and crowding at the level of the fourth ventricle due to mass effect. Following a diagnosis of cerebral edema due to MDMA-induced acute severe hyponatremia, she received a bolus of mannitol 20% (initial, 2 g/kg over 60 min; a second bolus of 0.5 g/kg a few hours later was given) and a normal saline infusion. Her neurological status, urine output, and serum sodium concentrations gradually improved. About a day later, a second head CT scan revealed significant improvement in cerebral edema with better visualization of the sulci. Her mental status and neurological examination, as well as serum sodium concentration (sodium 139 mmol/L), were normalized 2 days later (Ghatol & Kazory, 2012).
    J) LEUKOENCEPHALOPATHY
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Toxic leukoencephalopathy syndrome developed in a 17-year-old boy after ingesting an unknown amount of "molly" pills (MDMA, ecstasy). He presented with headaches, altered mental status, and double vision 2 days postingestion. CSF from a lumber puncture revealed mild lymphocytosis and elevated protein. Other lab results were normal. Diffuse signal abnormality in the midbrain, bilateral insulae, and basal ganglia bilaterally were observed in an MRI of the brain. It is suggested that leukoencephalopathy is resulted from serotonergic neuronal damage and myelin damage from oxidative stress (Ginat, 2015).
    K) PARALYSIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 14-year-old boy presented to the ED with sudden onset left-sided hemiplegia, right-sided hemiparesis, and headache. He initially denied any illicit drug use, but a urine toxicology screen revealed amphetamine and MDMA. After admission, the patient's status declined and he was intubated. Renal function deteriorated on hospital day 2, and a dilating spinal cord lesion was detected by MRI on hospital day 3. High-dose steroids and IV immunoglobulins were initiated in response to a suspected demyelinative process, but no improvement in symptoms was seen. Therapy was stopped, and the patient was extubated several days later. A follow-up MRI one month after admission revealed improvement in cervical spine edema. Over the following months, the child's condition improved, and he was transferred to a rehabilitation center. After 6 months of rehab, the patient had full resolution of the right hemiparesis with mild residual left hemiparesis (Goldstein et al, 2006).
    L) ABNORMAL GAIT
    1) WITH POISONING/EXPOSURE
    a) CHRONIC TOXICITY
    1) CASE REPORT: Parkinson-like symptoms (including postural instability and markedly abnormal gait) occurred in a 29-year-old man following ingestion of MDMA approximately 10 times over an 18 month period. Levodopa and pramipexole were tried unsuccessfully. It was speculated that the symptoms were a result of delayed neurotoxic effects of MDMA (Mintzer et al, 1999).
    M) DISSOCIATIVE DISORDER
    1) WITH POISONING/EXPOSURE
    a) TFMPP and BZP: Dissociative-type symptoms (eg, "insides had stopped working") developed in 3 young adults after ingesting 4 tablets containing 1-(3-trifluoromethylphenyl) piperazine (TFMPP) and 1-benzylpiperzine (BZP). In all cases, symptoms resolved by the following day (Wood et al, 2008).
    N) CLOUDED CONSCIOUSNESS
    1) WITH POISONING/EXPOSURE
    a) Preliminary data suggest that chronic use of MDMA may cause mild-to-moderate subclinical impairment in cognitive function, which may be related to deficits in serotonin (5-HT) function (Krystal et al, 1992).
    O) DYSTONIA
    1) WITH POISONING/EXPOSURE
    a) Acute dystonic reactions have been described after MDMA and MDEA use (Priori et al, 1995; Humphreys & Tanner, 1994).
    P) DOPAMINERGIC SYNAPTIC TRANSMISSION
    1) WITH POISONING/EXPOSURE
    a) CHRONIC EFFECTS
    1) SUMMARY: Recent evidence suggests that MDMA use can result in alterations and potential damage to serotonin (5-HT) neurons (Milroy, 1999; Pham & Puzantian, 2001; Reneman et al, 2006). Further studies have indicated that the effects of MDMA on 5-HT neurons in the human cortex may be reversible in individuals who have stopped using MDMA, but the effects of MDMA on memory function may be long-lasting (Reneman et al, 2001).
    b) In one study, ecstasy users had lower levels in the cerebrospinal fluid of 5-hydroxyindoleacetic acid (5-HIAA), a major metabolite of serotonin, which was considered to be a marker for serotonin terminal degeneration. In addition, in female MDMA users there was also a depletion in homovanillic acid, the main metabolite of dopamine. The authors concluded that MDMA can produce serotoninergic neuron damage (Milroy, 1999; Pham & Puzantian, 2001).
    c) Reneman et al (2001) studied the effects of MDMA on brain serotonin neurons using a radioligand that would bind with high affinity to serotonin transporters. The density of binding was calculated by single-photon-emission computed tomography (SPECT). The results indicated that heavy use of MDMA was associated with neurotoxic effects on serotonin neurons (women in this study were found to be more susceptible to the effects than men), and MDMA-induced neurotoxic changes in several brain regions of female ex-MDMA (no use in more than one year) users were reversible (Reneman et al, 2001).
    1) Similar work has been done by McCann et al (1998) using quantitative PET studies with a ligand selective for 5-HT transporters. They found that abstinent MDMA users showed decreased global and regional brain 5-HT transporter binding compared with controls. Decreases in 5-HT transporter binding positively correlated with the extent of previous MDMA use (McCann et al, 1998).
    2) In a small study by Obrocki et al (1999), positron emission tomography (PET) findings suggested that ecstasy use had lasting effects on central neuronal activity in humans (Obrocki et al, 1999).
    d) MDMA and its cogeners have been shown to induce reversible, dose-dependent depletion of serotonin and serotonin brain uptake sites in animals (Edwards, 1989; Peroutka, 1989). These neurotoxic effects may or may not occur in humans at prevalent doses; however, the possibility of their occurrence should remove the myth that these drugs are 'harmless'. This position is controversial; some argue that MDMA is no more neurotoxic than other widely-used drugs (Grob et al, 1990).
    e) One study in humans used serum prolactin levels and mood responses to intravenous L-tryptophan to compare known MDMA users to healthy controls. Suggestive, not significant, evidence of altered serotonin function in the users was found (Price et al, 1989). Krystal et al (1992) presented further evidence that chronic use of MDMA may cause serotonin (5-HT) deficits in humans(Krystal et al, 1992).
    f) CASE REPORTS
    1) Two cases of persistent neurobehavioral disturbance after MDMA ingestion have been reported (McCann & Ricaurte, 1992).
    2) A young woman developed symptoms (hyperthermia, confusion, and tachycardia) similar to serotonin syndrome following ingestion of MDMA. Despite aggressive management, the patient died 5 hours after admission from complications of hyperthermia and cardiac dysfunction (i.e., hypotension, malignant dysrhythmias) (Mueller & Korey, 1998).
    3) Ongoing interruption in serotonin transmission has been suggested in the role of discrete memory loss and learning deficits found in MDMA users (Klugman et al, 1999).
    Q) SCLEROSIS
    1) WITH POISONING/EXPOSURE
    a) HIPPOCAMPAL SCLEROSIS
    1) Gardner et all (2009) reported two cases of ecstacy ingestion that presented with tonic-clonic seizures, and MRI results showed hippocampal swelling that developed into sclerosis (Gardner et al, 2009).
    a) CASE REPORT: A 25-year-old man ingested 2 ecstacy tablets with 8 units of alcohol and experienced his first of two tonic-clonic seizures approximately 5 hours later. The following day he felt unwell, vomited and had the second seizure. A MRI 2 days after the seizures showed right hippocampal swelling and high signal, and a follow-up MRI 3 months later revealed right hippocampal atrophy and high signal, consistent with sclerosis. At the time of follow-up, the patient had remained seizure free and asymptomatic.
    b) CASE REPORT: A 25-year-old woman took 1 ecstasy tablet and 5 units of alcohol and presented 11 hours later following a 1-minute seizure. The patient presented confused and complained of headache and vomiting. A MRI 2 days after the ingestion showed right hippocampal swelling as well as signal change. The patient regained normal cognition on the third day, and returned to work with no sequelae. A follow-up MRI 3 months later revealed right hippocampal atrophy and ongoing high signal.
    R) AMNESIA
    1) WITH POISONING/EXPOSURE
    a) In a small study of chronic methamphetamine users (self reported use of an average of 2.4 times a month; potency of MDMA pills was unknown), progressive or permanent memory impairment may occur. This study found that MDMA is associated with different aspects of memory decline, but further study is needed (Anon, 2001).
    b) A study was conducted to determine working memory performance in pure MDMA users as compared with polyvalent MDMA users (concomitant use of MDMA with amphetamines or marijuana). Although results of the study showed no significant difference between the 2 groups in the cognitive performance of specific working memory tasks, cortical activation patterns, observed via an MRI, showed significantly lower responses in the pure MDMA users, primarily in the striate cortex, compared with polyvalent users, and higher responses in the premotor cortex, compared with polyvalent users. The results suggest that an altered cortical response may appear before significant impairments in cognitive performance become apparent. Because pure MDMA users denied concomitant use of any other drug, it is suggested that the signal changes may be due to prior MDMA use and may indicate its neurotoxic potential, although further research is needed (Daumann et al, 2003).
    c) CASE REPORT: A young adult was admitted to the ED unconscious after ingesting alcohol, cocaine and ecstasy. He was intubated and 2 days after extubation the patient was confused; oxygenation and vital signs were normal. Memory loss was suspected and a MR scan showed bilateral hippocampal damage, which can impair the formation of new memories (long-term memory is usually intact) (Foex et al, 2010).
    3.7.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) SEROTONIN SYNDROME
    a) SUMMARY: Based on animal studies, there is evidence that MDMA causes dose-related reductions of brain serotonin (5-HT) and 5-hydroxyindoleacetic acid (5-HIAA) (Steele et al, 1994).
    b) Monkeys dosed at 10 mg/kg twice daily for 4 days showed no measurable behavioral response to MDMA, but forebrain serotonin levels were reduced by 80% one month after dosing.
    c) MDMA-treated rats exhibited the serotonin motor syndrome within 30 minutes of dosing. Four weeks later, the behavioral tests of these rats were back to normal despite a 50% decrease in serotonin levels in the frontal cortex (Slikker et al, 1989).
    d) Another study of rats dosed with MDA and MDMA found a disruption of behavior patterns similar to that of hallucinogenic phenylethylamine derivatives (Gold et al, 1988).
    2) ALTERED NEUROTRANSMITTER LEVEL
    a) Rhesus monkeys injected with 10 mg/kg MDMA twice daily for 4 days experienced a decrease in serotonin uptake sites in cerebral cortex and striatum, as well as a decrease in blood serotonin levels (Insel et al, 1989).
    b) Battaglia et al (1988) found that reduction of serotonergic uptake sites (20% to 30% less than controls) was dependent on the MDMA dose in guinea pigs and rats, but no depletion was found in mice (Battaglia et al, 1988).
    c) After stopping MDMA treatment, animals recovered almost all of the lost uptake sites within one year. These effects may be directly due to MDMA, or may be due to metabolites.
    d) DEPLETION OF SEROTONIN: A single injection of MDA at 10 mg/kg (3 to 5 times the dose used by humans) caused a 32% decrease of the serotonin level in the hippocampus of rats (Ricaurte et al, 1985).
    1) MDMA given orally to rats demonstrated the same dose-related depletion of serotonin in the hippocampus (Finnegan et al, 1988). Battaglia et al (1988) found that brain serotonin levels in rats decreased 20% after only 2 doses of 10 mg/kg MDMA (Battaglia et al, 1988).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) NAUSEA AND VOMITING
    1) WITH POISONING/EXPOSURE
    a) SUMMARY
    1) Nausea is common, with an onset of 30 minutes and lasting another 30 minutes (Hayner & McKinney, 1986). Vomiting may be protracted in some cases. Vomiting and diarrhea may also occur (Gee et al, 2010).
    b) MDMA
    1) INCIDENCE: In a retrospective chart review of 191 cases of MDMA exposure, nausea and vomiting (12%) were frequently reported (Rella et al, 2000).
    B) ACUTE PANCREATITIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 2-year-old boy who ingested an unknown amount of MDMA presented with elevations in the following pancreatic enzymes: amylase 501 International Units/L and lipase 4291 Units/L. The patient tested positive for MDMA the morning after hospital admission; however, the length of time between MDMA exposure to clinical presentation is unknown. One month after hospital discharge, the patient was reported to be clinically normal; however, his pancreatic enzymes remained slightly elevated (Feldman & Mazor, 2007).
    C) PNEUMORETROPERITONEUM
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Spontaneous pneumomediastinum and pneumoretroperitoneum without associated esophageal perforation developed in a 17-year-old with protracted vomiting following ingestion of MDMA (Levine et al, 1993).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) LIVER DAMAGE
    1) WITH POISONING/EXPOSURE
    a) Jaundice, elevated serum transaminase levels and bilirubin, and hepatomegaly have been reported in patients with acute MDMA and MDA toxicity (Ben-Abraham et al, 2003; Barrett & Taylor, 1993; Henry et al, 1992; Jones et al, 1994).
    b) PEDIATRIC EXPOSURE: A 2-year-old boy ingested an unknown amount of MDMA and presented with elevations in the following liver enzymes: aspartate aminotransferase 12,463 IU/L, alanine aminotransferase 4606 IU/L, gamma-glutamyltransferase 232 IU/L, alkaline phosphatase 442 IU/L. His reported bilirubin level was 1.5/0.5 mg/dL. One month after hospital discharge, the patient was reported to be clinically normal; however, his hepatic enzymes remained slightly elevated (Feldman & Mazor, 2007).
    B) TOXIC HEPATITIS
    1) WITH POISONING/EXPOSURE
    a) MDMA
    1) Hepatitis has developed in patients after recreational use of MDMA (Shearman et al, 1992; Gorard et al, 1992; Dykhuizen et al, 1995). Fatal fulminant hepatitis occurred in a young adult following MDMA exposure despite medical treatment (Colak et al, 2011).
    2) ETIOLOGY: The potential risk for liver injury is likely multifactorial. Several possible explanations have been suggested in the literature: toxic hepatitis due to MDMA or its metabolites or to an unknown contaminant introduced during manufacture; idiosyncratic toxic hepatitis due to genetic predisposition; hemodynamic alteration in liver circulation; induction of isoenzymes of CYP450; or acute hyperthermic hepatotoxicity (Jones & Simpson, 1999; Garbino et al, 2001). Toxicity can produce asymptomatic hepatitis to acute hepatic failure (Colak et al, 2011).
    3) ONSET: Signs of hepatic toxicity may develop a few days up to 1 to 2 weeks after exposure (Colak et al, 2011).
    4) CASE REPORT: A 19-year-old man presented severely jaundiced 2 weeks after consuming 2 ecstasy tablets. He had an enlarged liver and spleen, but all serologic markers for liver diseases were negative. The patient had no other source or exposure to explain his acute hepatitis. With supportive care, he made a full recovery and was discharged on hospital day 20. All laboratory parameters normalized by 2 months (Brncic et al, 2006).
    b) MDA
    1) CASE SERIES: Six patients developed symptoms of hepatotoxicity with cholestatic jaundice following exposure to 4,4'-methylenedianiline (MDA). Although hepatic enzymes normalized over a period of 6 weeks in all patients, the authors suggested that, theoretically, the decline in acetylcholinesterase activity may result in severe liver damage leading to fulminant hepatic failure following recreational use (Tillman et al, 1997).
    C) HEPATIC FAILURE
    1) WITH POISONING/EXPOSURE
    a) SUMMARY
    1) Hepatic failure may occur with severe exposure to these agents (Lamberth et al, 2008; Garbino et al, 2001; Kramer et al, 2003).
    b) MDMA
    1) CASE REPORT: Necrotic toxic hepatitis developed in a 19-year-old man following ingestion of 1.5 tablets of ecstasy (MDMA) and alcohol. The patient reported an occasional use of recreational drugs. Symptoms progressed and the patient's condition deteriorated over a 3 week period resulting in fulminant hepatic failure which required auxiliary liver transplantation. The patient was discharged to home 20 days after surgery (Garbino et al, 2001).
    2) CASE REPORT: Fulminant hepatic failure occurred in a 27-year-old man following injection of 0.5 g heroin, 1 g cocaine, and ingesting 3 MDMA tablets (300 mg). Liver transplantation was declined by the transplant surgeon due to the patient's history of illicit drug use. Initiation of experimental therapy using a fractionated plasma separation and adsorption system (FPSA/Promethus), a hemodialysis-based system, resulted in the gradual recovery of hepatic function (Kramer et al, 2003).
    c) PMA
    1) PARAMETHOXY-AMPHETAMINE: A 20-year-old man was admitted unconscious after ingesting PMA (confirmed by laboratory analysis) with hypertension, tachycardia and an ECG showing a prolonged QRS interval. The patient had a complex course which included hyperthermia, persistent coma, shock, hyperkalemia, rhabdomyolysis and severe coagulopathy. This was followed by renal and hepatic failure and extensive cerebral edema. Laboratory studies included a bilirubin of 412 micromol/L (reference range: 2 to 20) and ALT of 3961 Units/L (reference range: less than 55). He died 10 days after presentation (Lamberth et al, 2008).
    3.9.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) HEPATOTOXICITY
    a) The metabolism of 3,4-methylenedioxymethamphetamine (MDMA) and 3,4-methylenedioxyamphetamine (MDA) can produce highly reactive hepatotoxic compounds N-methyl-alpha-methyldopamine (N-Me-alpha-MeDA) and alpha-methyldopamine (alpha-MeDA), and alpha-MeDA (Carvalho et al, 2004).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) ABNORMAL URINE
    1) WITH POISONING/EXPOSURE
    a) PEDIATRIC EXPOSURE: A 2-year-old boy ingested an unknown amount of MDMA and presented with 1+ proteinuria, 2+ ketonuria, and 3+ occult blood with more than 100 red blood cells per high power field. The patient tested positive for MDMA the morning after hospital admission; however, the length of time between MDMA exposure to clinical presentation was unknown (Feldman & Mazor, 2007).
    B) MYOGLOBINURIA
    1) WITH POISONING/EXPOSURE
    a) Myoglobinuria secondary to rhabdomyolysis has been reported (Ginsberg et al, 1970; Brown et al, 1986).
    C) ACUTE RENAL FAILURE SYNDROME
    1) WITH POISONING/EXPOSURE
    a) SUMMARY
    1) Acute renal failure has been reported in patients who develop rhabdomyolysis and/or disseminated intravascular coagulation associated with BZP, MDA, MDEA, or MDMA (Davies et al, 2014; Alansari & Hamilton, 2006; Barrett & Taylor, 1993; Henry et al, 1992; Fahal et al, 1992; Watson et al, 1993; Walubo & Seger, 1999).
    b) MDMA
    1) CASE REPORT: DIC and acute renal failure were reported in a 21-year-old man after taking 30 "hits" (a typical "hit" of MDMA is 100 mg, but can range from 50 to 250 mg) of ecstasy. The patient was extubated on day 25, but required daily hemodialysis, and permanent neurological impairment was present (Shannon, 2000).
    2) PEDIATRIC EXPOSURE: A 2-year-old boy ingested an unknown amount of MDMA and presented with abnormal urine findings (ie, 1+ proteinuria, 2+ ketonuria, and 3+ occult blood with more than 100 red blood cells per high power field) and developed mild DIC and muscle injury. The patient tested positive for MDMA the morning after hospital admission; however, the length of time between MDMA exposure to clinical presentation was unknown (Feldman & Mazor, 2007).
    D) RETENTION OF URINE
    1) WITH POISONING/EXPOSURE
    a) Urinary retention has been reported after large doses of MDMA (Bryden et al, 1995).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) Metabolic (lactic) acidosis may occur in severe cases (Ghatol & Kazory, 2012; Ovaska et al, 2008; Balikova, 2005; Ben-Abraham et al, 2003; Chadwick et al, 1991; Barrett & Taylor, 1993; Logan et al, 1993; Tehan, 1993; Webb & Williams, 1993; Watson et al, 1993).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) DISSEMINATED INTRAVASCULAR COAGULATION
    1) WITH POISONING/EXPOSURE
    a) Prolonged INR or PT and PTT, thrombocytopenia, anemia, and elevated fibrin degradation products have been observed in severe and fatal poisonings (Brown et al, 1986; Ginsberg et al, 1970; Chadwick et al, 1991; Campkin & Davies, 1992; Screaton et al, 1992; Henry et al, 1992; Barrett & Taylor, 1993; Tehan, 1993; Watson et al, 1993; Fineschi & Masti, 1996; Walubo & Seger, 1999).
    b) CASE REPORT: A 23-year-old man presented with tachycardia, hypotension, tachypnea, a Glasgow Coma Scale (GCS) of 12/15 and fever (rectal temperature of 108.9 degrees F; 42.7 degrees C) after ingesting 3 MDMA pills at a concert. He was also combative, tremulous, hypertonic and hyper-reflexic. Following supportive care, including ventilatory support, his temperature decreased to 99 degrees F (37.2 degree C) within 2 hours. He also received sodium polystyrene and calcium gluconate after laboratory results revealed hyperkalemia (potassium 6.2 mmol/L). Following further supportive care, including IV midazolam and propofol drips, his rigidity decreased and tachycardia improved. However, he developed severe rhabdomyolysis, multiorgan failure, and mild disseminated intravascular coagulopathy on day 2. Laboratory results revealed a peak CK of 130,350 Units/L, AST 3011 Units/L, ALT 314 Units/L, albumin 1.6 g/dL, creatinine 9.6 mg/dL, phosphate 8 mg/dL, platelet count 54 K/mcL, INR 1.44, activated partial thromboplastin time (aPTT) 42.7 and D-dimer concentrations of 1799 ng/mL. Following aggressive supportive care, including several sessions of hemodialysis (a total of 5 inpatient sessions and 2 outpatient sessions), he had a full recovery within 3 weeks (Davies et al, 2014).
    c) CASE REPORT: DIC and acute renal failure were reported in a 21-year-old man after taking 30 "hits" (a typical "hits" of MDMA is 100 mg, but can range from 50 to 250 mg) of ecstasy. Permanent neurological sequelae was reported (Shannon, 2000).
    d) CASE REPORT: A young man died of disseminated intravascular coagulation (DIC), induced by hyperthermia following the combined ingestion of amphetamines (3,4-methylenedioxymethylamphetamine, 3,4-methylenedioxyamphetamine, and paramethoxyamphetamine) (Dams et al, 2003).
    e) PEDIATRIC EXPOSURE: A 2-year-old boy ingested an unknown amount of MDMA and developed mild disseminated intravascular coagulation (ie, elevated prothrombin time of 15.3 seconds, INR 1.2 (normal), partial thromboplastin time was low at 22 seconds and D-dimers were severely elevated at more than 20 mcg/mL). The following day, the patient developed anemia with a hematocrit of 28% and his prothrombin time increased to 16.6 seconds. The patient tested positive for MDMA; however, the length of time between MDMA exposure to clinical presentation was unknown. After supportive care, the patient recovered and was discharged (Feldman & Mazor, 2007).
    B) THROMBOCYTOPENIC DISORDER
    1) WITH POISONING/EXPOSURE
    a) Thrombocytopenia has been reported following overdoses of MDMA (Campkin & Davies, 1992; Henry et al, 1992).
    C) APLASTIC ANEMIA
    1) WITH POISONING/EXPOSURE
    a) Two cases of aplastic anemia associated with MDMA use have been reported (Marsh et al, 1994).
    D) THROMBOTIC THROMBOCYTOPENIC PURPURA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Two days after using MDMA, a 37-year-old occasional MDMA user presented with fever, upper abdominal pain, and petechiae on his limbs. Urinalysis revealed macrohematuria, proteinuria (total protein, 5564 mg/L), red blood cell casts, and acanthocytes. Following the diagnosis of rapid-progressive glomerulonephritis, he was admitted to the ICU. Laboratory results revealed elevated serum creatinine 159 mcmol/L (normal range, 59 to 104 mcmol/L) and BUN 12.3 mmol/L (normal range, 3.8 to 8 mmol/L), severe thrombocytopenia (platelet count, 23,000/ mcL), and anemia (hemoglobin 6 g/dL) with signs of hemolysis: schistocytes in peripheral blood smear 1.3%, lactate dehydrogenase 21,257 International Units (normal range, 135 to 225 IU/L), haptoglobin less than 0.1 g/L (normal range, 0.3 to 2 g/L), free hemoglobin in plasma 44 mcmol/L (normal range, less than 3 mcmol/L). His condition deteriorated and he became confused and obtunded, necessitating orotracheal intubation. He underwent continuous renal replacement therapy (CRRT) after he developed oliguria and metabolic acidosis. At this time, his platelet count was 5000/mcL and a diagnosis of thrombotic thrombocytopenia purpura (TTP) was made. Despite using plasma exchange, he developed a severe lactic acidosis (blood lactate peaking at 19 mmol/L). He experienced a total atrioventricular block 16 hours after the ICU admission followed by asystole. An ECG revealed an acute myocardial infarction with wide-spread ST-segment elevation. He died despite intensive resuscitation measures. Postmortem laboratory analysis revealed an ADAMTS 13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13) activity of 10% (reference range, 50% to 110%) and an ADAMTS 13 antigen of 0.14 mcg/mL (reference range, 0.5 to 1.6 mcg/mL), supporting the diagnosis of TTP (de Fallois et al, 2015).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) EXCESSIVE SWEATING
    1) WITH POISONING/EXPOSURE
    a) Pallor, diaphoresis, and piloerection may be present (Campkin & Davies, 1992; Woods & Henry, 1992; Henry et al, 1992; Watson et al, 1993).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) HYPERREFLEXIA
    1) WITH POISONING/EXPOSURE
    a) Muscle spasms, tremors, and hyperreflexia are common; clonus, myoclonus, opisthotonus, rigidity, akinesia, and rhabdomyolysis may also occur.
    b) Muscle tension and/or jaw clenching were described by 100% of 44 users of MDMA during recreational intoxication (Siegel, 1986).
    c) CASE REPORT/MDMA: Repetitive and jerky movements were associated with hyperreflexia and muscle rigidity in an 11-month-old boy admitted with apparent seizures following the accidental ingestion of MDMA (confirmed by urine testing). The toddler recovered with supportive care (Garcia-Algar et al, 2005).
    B) RHABDOMYOLYSIS
    1) WITH POISONING/EXPOSURE
    a) SUMMARY
    1) Rhabdomyolysis is a common complication in patients who develop hyperthermia, or prolonged seizures, coma, or muscular hyperactivity (Ovaska et al, 2008; Lamberth et al, 2008; Brown & Osterloh, 1987; Campkin & Davies, 1992; Screaton et al, 1992; Singarajah & Lavies, 1992; Henry et al, 1992; Barrett & Taylor, 1993; Logan et al, 1993; Watson et al, 1993; Lehmann et al, 1995).
    b) MDMA
    1) CASE REPORT: A 19-year-old man presented in a coma with seizures, and hyperthermia (temperature 42.5 degrees C) 2 hours after ingesting ethanol and 3 ecstasy tablets (total dose unknown). Physical examination revealed bilateral mydriasis, myoclonus, a systolic murmur, and a thrill over the femoral arteries. He had a heart rate of 95 beats/min and arterial blood pressure of 125/45 mm Hg. An ECG revealed broad QRS complexes with short QT interval and a long PR interval, indicating hyperkalemia. Despite supportive therapy, including treatment with active cooling measures, sedation with midazolam, IV calcium gluconate, sodium bicarbonate, glucose, and insulin, he developed bradycardia and a P wave asystole. During cardiopulmonary resuscitation, he developed ventricular fibrillation that was successfully defibrillated. Laboratory results revealed hyperkalemia (peak potassium, 9.4 mmol/L after 30 minutes in the ICU), rhabdomyolysis (peak CK, 12,509 Units/L), myocardial damage, and syndrome of inappropriate antidiuretic hormone. He experienced another cardiac arrest after arrival in the ICU. He had pulmonary edema and pulseless electrical activity, alternating with sinus bradycardia (30 beats/min). Resuscitation efforts were unsuccessful and he died 2 hours later. It was suggested that the cause of death was a rapidly evolving hyperkalemia due to rhabdomyolysis (Vanden Eede et al, 2012).
    2) CASE REPORT: A 23-year-old man presented with tachycardia, hypotension, tachypnea, a Glasgow Coma Scale (GCS) of 12/15 and fever (rectal temperature of 108.9 degrees F; 42.7 degrees C) after ingesting 3 MDMA pills at a concert. He was also combative, tremulous, hypertonic and hyper-reflexic. Following supportive care, including ventilatory support, his temperature decreased to 99 degrees F (37.2 degree C) within 2 hours. He also received sodium polystyrene and calcium gluconate after laboratory results revealed hyperkalemia (potassium 6.2 mmol/L). Following further supportive care, including IV midazolam and propofol drips, his rigidity decreased and tachycardia improved. However, he developed severe rhabdomyolysis, multiorgan failure, and mild disseminated intravascular coagulopathy on day 2. Laboratory results revealed a peak CK of 130,350 Units/L, AST 3011 Units/L, ALT 314 Units/L, albumin 1.6 g/dL, creatinine 9.6 mg/dL, phosphate 8 mg/dL, platelet count 54 K/mcL, INR 1.44, activated partial thromboplastin time (aPTT) 42.7 and D-dimer concentrations of 1799 ng/mL. Following aggressive supportive care, including several sessions of hemodialysis (a total of 5 inpatient sessions and 2 outpatient sessions), he had a full recovery within 3 weeks (Davies et al, 2014).
    3) CASE REPORT: Severe rhabdomyolysis as evidenced by an initial serum creatine kinase (CK) of 6,111 Units/L was observed in a 21-year-old man after ingesting ecstasy (7 tablets), amphetamine and alcohol. CK peaked at 122,341 Units/L on hospital day 10, remaining above 1000 until day 26, and returned to normal after 45 days. Progressive muscle damage was evidenced by severe weakness and ongoing ventilatory support (weaning attempts failed). After a complex hospital course, the patient was discharged to home doing well on hospital day 80 (Murthy et al, 1997).
    4) CASE REPORTS: Rhabdomyolysis was reported in 3 people following ingestion of up to 3 MDMA tablets. The serum creatine kinase levels ranged from 800 Units/L on hospital day 1 (approximately 6 hours postingestion) to 225,000 Units/L on hospital day 3. All three patients completely recovered without sequelae (Ben-Abraham et al, 2003).
    5) CASE REPORT/PEDIATRIC: An 8-month-old boy presented with generalized seizure activity, tachycardia (210 beats/min), hypertension (BP 125/70 mmHg), and hyperthermia (38.9 degrees C) after accidentally ingesting one ecstasy tablet. Creatine kinase (CK) level was also elevated and peaked 6 hours after admission at 1681 Units/L. Treatment with benzodiazepines, body cooling and fluids stabilized the child's clinical status within 6 hours of admission, and CK normalized 5 days after discharge (Eifinger et al, 2008).
    6) PEDIATRIC EXPOSURE: A 2-year-old boy ingested an unknown amount of MDMA and presented with elevated creatine phosphokinase level of 2854 U/L. The patient tested positive for MDMA the morning after hospital admission; however, the length of time between MDMA exposure to clinical presentation was unknown. After supportive care, the patient recovered and was discharged (Feldman & Mazor, 2007).
    c) PMA
    1) PARAMETHOXY-AMPHETAMINE: A 20-year-old man was admitted unconscious after ingesting PMA (confirmed by laboratory analysis) with hypertension, tachycardia and an ECG showing a prolonged QRS interval. The patient had a complex course which included hyperthermia, persistent coma, shock, hyperkalemia, rhabdomyolysis and severe coagulopathy. Several days after admission, his laboratory studies included a creatine kinase of 58358 Units/L (reference range: 20 to 200), and a troponin I level of 85.8 (reference range: less than 0.06). Further multiorgan failure included renal and hepatic failure and extensive cerebral edema. He died 10 days after presentation (Lamberth et al, 2008).
    d) DOC
    1) CASE REPORT: A 20-year-old man collapsed and had tonic-clonic seizures after presumably ingesting DOI (2,5 dimethoxy-4-iodoamphetamine) at a rave party. He required intubation in the field and was admitted comatose with a Glasgow Coma Score of 3/15. However, he had normal reflexes and a brain CT was normal. He also developed tachycardia (152 beats/min), metabolic acidosis, hypotension and an elevated creatine kinase (peak: 4924 International Units/L) with no evidence of renal failure. Following supportive care, including IV hydration and prophylactic antibiotics, the patient was extubated the next day and discharged to home with no permanent sequelae. Serum and urine samples confirmed the presence MDMA and DOC (2,5 dimethoxy-4-chloroamphetamine), which could not be quantified (Ovaska et al, 2008).
    C) COMPARTMENT SYNDROME
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 46-year-old man injected 3 ecstasy tablets (crushed and mixed with tap water) into his right groin and subsequently developed right posterior thigh compartment syndrome. The patient underwent a posterior compartment fasciotomy 6 hours after the injection, and the fasciotomy had to be extended to the mid-thigh level 48 hours later secondary to right foot-drop with infra-malleolar parasthesia. At a 15 month follow-up, the patient had residual damage with limited flexion, extension, and sensation in his right leg and foot. The authors suggested that the injected powdered ecstasy into the femoral artery embolized and occluded blood supply to the posterior thigh (Swan et al, 2006).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) ADRENAL HEMORRHAGE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 2-year-old child ingested an unknown amount of MDMA and developed right adrenal hemorrhage revealed by an abdominal computed tomography. The patient tested positive for MDMA the morning after hospital admission; however, the length of time between MDMA exposure to clinical presentation was unknown (Feldman & Mazor, 2007).
    B) ABNORMAL ANTI-DIURETIC HORMONE
    1) WITH POISONING/EXPOSURE
    a) SUMMARY: Hyponatremia, secondary to syndrome of inappropriate antidiuretic hormone secretion (SIADH) has been reported after MDMA abuse (Farah & Farah, 2008; Ajaelo et al, 1998; Holden & Jackson, 1996).
    b) PATHOLOGY: This syndrome can produce euvolemic hyponatremia and is caused by nonosmotic, non-extracellular fluid volume-mediated factors. MDMA can stimulate CNS release of serotonin and inhibits serotonin reuptake resulting in a rapid increase in serotonin levels. Animal studies have shown that serotonin stimulates the release of antidiuretic hormone (Zenenberg & Goldfarb, 2000).
    c) CASE REPORT: A 19-year-old woman developed seizures and mental status depression (GCS 11) after ingesting MDMA. Her serum sodium was 115 mmol/L, serum osmolality 253 mosm/kg, urine osmolarity 522 mosm/kg and urine sodium 162 mmol/L. She recovered after treatment with 3% NaCl at 50 mL/hr and furosemide 40 mg twice a day (Ajaelo et al, 1998).
    d) CASE REPORT: Increased arginine release (plasma arginine vasopressin 4.5 pmol/L) was the primary toxicity, with water intoxication a secondary factor, in the development of ecstasy-induced hyponatremia in a 20-year-old woman after one tablet (Holden & Jackson, 1996).
    e) CASE SERIES: In human volunteers, low doses of MDMA (47.5 mg) induced increased secretion of arginine vasopressin and a small fall in plasma sodium concentration (Henry & Hill, 1998).
    C) HYPOGLYCEMIA
    1) WITH POISONING/EXPOSURE
    a) Hypoglycemia has been reported, but is not common with these agents (Lamberth et al, 2008; Ben-Abraham et al, 2003; Chadwick et al, 1991; Webb & Williams, 1993; Watson et al, 1993).
    b) PMA intoxications are characterized by hypoglycemia, hyperkalemia and QRS interval prolongation (Lamberth et al, 2008).
    D) INCREASED OXYTOCIN LEVEL
    1) WITH THERAPEUTIC USE
    a) In a study of 15 healthy volunteers with a history of regular ecstasy use, an oral dose of MDMA (100 mg) or a matched placebo were administered and then evaluated for prosocial effects at frequent intervals (15, 60, 105, 150, 240 and 300 min). It was noted that subjective increases in prosocial feelings occurred about 60 minutes after administration of MDMA. An increase in plasma concentrations of oxytocin were correlated with an increase in subjective prosocial feelings. Plasma oxytocin concentrations were significantly elevated compared to placebo. Mean plasma oxytocin concentrations were 0.8 pmol/L at baseline to an average maximum concentration of 34.3 pmol/L at 110 min after administration (Dumont et al, 2009a).
    3.16.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) GLUCOCORTICOIDS INCREASED
    a) MDMA: When dosed at 10 mg/kg intraperitoneally in rats, MDMA elevated serum corticosteroid and prolactin levels within 1 hour of administration (Nash et al, 1988).

Reproductive

    3.20.1) SUMMARY
    A) Ecstasy has been associated with cardiovascular and musculoskeletal malformations in babies exposed in utero.
    3.20.2) TERATOGENICITY
    A) CONGENITAL ANOMALY
    1) A prospective follow-up of 136 babies exposed to ecstasy in utero suggested that the drug may be associated with an increased risk of congenital defects (15.4%). A significant increase in cardiovascular anomalies (26 per 1000 livebirths {95% CI}) and musculoskeletal anomalies (38 per 1000 livebirths {95% CI}) were observed (McElhatton et al, 1999).
    B) MOTOR DELAYS
    1) A prospective, longitudinal cohort design study evaluated 28 MDMA-exposed and 68 non-MDMA-exposed infants from birth to 2 years of age. Overall, no differences were observed between MDMA-exposed and non-MDMA-exposed infants at birth. However, prenatal MDMA exposure was associated with fine and gross motor delays in the first 2 years of life (Singer et al, 2015).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor vital signs and mental status.
    B) Plasma levels are not clinically useful or readily available.
    C) Monitor sodium and renal function. Obtain an ECG and institute continuous cardiac monitoring in patients with moderate to severe toxicity (agitation, delirium, seizures, coma, hypotension).
    D) Monitor creatinine phosphokinase in patients with prolonged agitation, seizures or coma; monitor renal function and urine output in patients with rhabdomyolysis.
    E) Standard urine toxicology screen may not produce a positive result for many of the specific hallucinogenic amphetamines.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) Monitor sodium and renal function.
    2) Monitor creatinine phosphokinase in patients with prolonged agitation, seizures or coma; monitor renal function and urine output in patients with rhabdomyolysis.
    3) Monitor fluid status, which may include hemodynamic pressure monitoring, to evaluate and guide fluid therapy in severe overdose.
    4.1.3) URINE
    A) TOXICOLOGY SCREEN
    1) 2C-T-4
    a) CASE REPORT: A urine test (Triage(R) was unable to detect any psychoactive substances in an adult that had ingested 2C-T-4 (2-[2,5-dimethoxy-4-isopropylsulfanylphenyl] ethanamine. However, the substance can be detected by gas-chromatography-mass spectrometry (Miyajima et al, 2008).
    4.1.4) OTHER
    A) OTHER
    1) MONITORING
    a) Obtain an ECG and institute continuous cardiac monitoring in patients with moderate to severe toxicity (i.e., agitation, delirium, seizures, coma, hypotension).
    b) Central venous or pulmonary artery pressure monitoring may be required in significant exposures.

Methods

    A) MULTIPLE ANALYTICAL METHODS
    1) STANDARD TOXICOLOGY SCREENING
    a) Standard urine toxicology screen may not produce a positive result for many of the specific hallucinogenic amphetamines.
    b) The usual toxicology screen fails to detect MDMA unless large doses have been ingested. A positive test will show up positive for the amphetamine drug class (Schwartz & Miller, 1997).
    c) Hallucinogenic amines and their metabolites can be detected by thin layer chromatography, ultraviolet spectroscopy, gas chromatography, and gas-liquid chromatography and liquid chromatography tandem mass spectrometry. Immunoassays are less sensitive than other methods. Qualitative analysis is sufficient to determine or confirm the identity of the intoxicant.
    2) IMMUNOASSAYS
    a) The EMIT(R) amphetamine assay responds to MDA, MDMA, and MDEA, but is less sensitive than for amphetamine. Minimal detectable concentrations are 13 mg/L for MDA, 5 to 8 mg/L for MDMA, 4 to 7 mg/L for MDEA, and 0.3 mg/L for amphetamine (Bost, 1988).
    1) A monoclonal immunoassay for amphetamine/methamphetamine can detect MDMA, but it is recommended to confirm any positive immunoassay screening tests by means of gas chromatography/mass spectrometry (Schwartz & Miller, 1997; Rome, 2001).
    2) Amphetamine immunoassays have limited ability to detect MDMA. High concentrations of MDMA (3,4-methylenedioxymethamphetamine) in the urine are needed to elicit a response on amphetamine immunoassays. For common monoclonal amphetamine immunoassays (eg, EMIT, FPIA, RIA) the sensitivity for MDMA is approximately 50% less than for amphetamines and methamphetamines (Moeller et al, 2008). Three monoclonal antibodies tests can specifically test for amphetamine, methamphetamine and MDMA at one time. CEDIA-Amp/MDMA (MDMA as the testing calibrator for MDMA and d-MA), CEDIA-Amp/MDMA (d-MA as the testing calibrator) and OnLine-Amp/MDMA can detect low levels of both MDMA and MA at low concentrations (Hsu et al, 2003).
    3) LIQUID CHROMATOGRAPHY TANDEM MASS SPECTROMETRY
    a) The following hallucinogenic amphetamines can be detected using LC-MS/MS (liquid chromatography tandem mass spectrometry) (Wohlfarth et al, 2010):
    1) 2,5-DMA: (1-(2,3-dimethophenyl)propan-2-amine)
    2) 2C-B: (2-(4-bromo-2,5-dimethoxyphenyl)ethanamine)
    3) 2C-D: (2-(2,5 dimethoxy-4-methylphenyl)ethanamine)
    4) 2C-H: (2-(2,5-dimethoxyphenyl)ethanamine)
    5) 2C-I: (2-(4-iodo-2,5-dimethoxyphenyl)ethanamine)
    6) 2C-P: (2-2,5-diemethoxy-4-prophylphenyl)ethanamine)
    7) 2C-T-2: (2-[4-(ethylthio)-2,5-dimethoxyphenyl]ethanamine)
    8) 2C-T-4:(2-[4-(isopropylthio)-2,5-dimethoxyphenyl]ethanamine)
    9) 2C-T-7: (2-[2,5-dimethoxy-40(propylthio)phenyl]ethanamine)
    10) 3,4,5-TMA: (1-(3,4,5-trimethoxyphenyl)propan-2-amine)
    11) 3,4-DMA: (1-(3,4-dimethoxyphenyl)propan-2-amine)
    12) 4-MTA: (1-4-(methylthio)phenyl]propan-2-amine)
    13) DOB: (1-4-bromo-2,5-dimethoxypehnyl)propan-2-amine)
    14) DOET: (1-(4-ethyl-2,5-dimethoxyphenyl)propan-2-amine)
    15) DOM: (1-(2,5-dimethoxy-4-methylphenyl)propan-2-amine)
    16) Ethylamphetamine: (N-ethyl-1-phenylpropan-2-amine)
    17) MDDMA: (1-(1,3-benxodioxyl-5-yl)-N,N-dimethylpropan-2-amine)
    18) PMA: (1-(4-methoxyphenyl)propan-2-amine)
    19) PMMA: (1-(4-methoxyphenyl)-N-methylpropan-2-amine)
    20) TMA-6: (1-(2,4,6-trimethoxyphenyl)propan-2-amine)
    b) Liquid-chromatography tandem mass spectrometry method was able to detect and quantify both MDA and 2CI in urine; however, this method can only detect the parent drug of 2C-I, while 2C-I is present in conjugated and metabolized forms in the urine (Drees et al, 2009).
    c) Liquid chromatography-sonic spray ionization-mass spectrometry (LC-SSI-MS) (after pretreatment with liquid-liquid extraction) was used to screen for the presence of MDMA in blood and urine and amphetamine in urine (Dams et al, 2003).
    4) GAS CHROMATOGRAPHY/MASS SPECTROMETRY
    a) MDMA
    1) MDMA: Thin-layer chromatography can detect MDMA metabolites in the urine (Schwartz & Miller, 1997).
    b) 2C-T-4
    1) 2C-T-4: Gas chromatography-mass spectometry can detect 2C-T-4; however it is difficult to detect when screened with Triage(R) (Miyajima et al, 2008).
    c) 2C-T-7
    1) 2,5-dimethoxy-4-n-propylthiophenethylamine (2C-T-7): This agent is structurally and pharmacodynamically similar to methylenedioxymethamphetamine (MDMA). A 20-year-old man died after insufflating 35 mg of 2C-T-7. Gas chromatography with nitrogen-phosphorus detection (GC-NPD) and electron ionization GC-mass spectrometry (MS) utilizing selected ion monitoring (GC-MSD) were used to analyze postmortem blood, urine and liver samples (The limits of detection and quantitation in blood were 6.0 and 15.6 ng/mL for both GC-NPD and GC-MS, respectively). The following methods were used: the addition of trimethoxyamphetamine (TMA) as internal standard; alkalinization with ammonium hydroxide; liquid-liquid extraction with n-chlorobutane; a 1:4 aqueous homogenate pretreated with dilute perchloric acid, centrifuged, and the supernatant was extracted to facilitate recovery from liver. To prevent loss of drug by evaporation, 0.1% hydrochloride acid in methanol was added during the final concentration step (Curtis et al, 2003).
    d) BZP
    1) BZP: GC-MS was used to detect 1-benzylpiperazine (BZP) in a young adult after ingesting 5 tablets (Wood et al, 2007).
    e) DOB
    1) Balikova (2005) reported two cases of severe intoxication with 2,5-dimethoxy-4-bromoamphetamines (DOB). Although immunoassay for amphetamines was negative in urine specimens of 2 men, GC-MS showed DOB in the gastric and urine samples. DOB serum concentrations were 13 ng/mL for the surviving patient and 19 ng/mL for the deceased patient (Balikova, 2005).
    f) MDE
    1) An assay for simultaneous quantification of the enantiomers of 3,4-methylenedioxy-N-ethylamphetamine (MDE) and MDA (a proposed metabolite of MDE) has been developed. The method uses derivatization with an optically pure reagent and analysis by gas chromatography (Hegadoren et al, 1995).
    g) PMA AND PMMA
    1) PMA, PMMA, and MDMA: Gas chromatography-mass spectrometry was used to determine the quantitation of PMA, PMMA, and MDMA in the postmortem blood of 3 individuals (Johansen et al, 2003).
    h) PPP
    1) ALPHA-PYRROLIDINOPROPIOPHENONE: In one animal study, PPP metabolites could be detected in rat urine using solid-phase extraction, trimethylsilylation and gas chromatography-mass spectrometry (GC-MS) (the limit of detection for the parent compound: 100 ng/mL). To detect other designer drugs of the pyrrolidinophenone type such as MOPPP, MDPPP, MPHP, and MPPP, the same detection procedure can be used (Springer et al, 2003).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) Patients with significant persistent central nervous system toxicity (i.e., hallucinations, somnolence, delirium, coma), hyponatremia, or persistent tachycardia should be admitted. Patients with coma, seizures, dysrhythmias, serotonin syndrome or delirium should be admitted to an intensive care setting.
    6.3.1.2) HOME CRITERIA/ORAL
    A) Asymptomatic adults may be monitored at home. However, it is unlikely that a patient will be asymptomatic.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Consult a poison center or medical toxicologist for assistance in managing patients with severe toxicity (i.e., seizures, dysrhythmias, severe delirium, coma), or in whom the diagnosis is not clear.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Patients with deliberate self-harm ingestions or children with any ingestion and symptomatic patients should be sent to a healthcare facility for observation for 6 to 8 hours.

Monitoring

    A) Monitor vital signs and mental status.
    B) Plasma levels are not clinically useful or readily available.
    C) Monitor sodium and renal function. Obtain an ECG and institute continuous cardiac monitoring in patients with moderate to severe toxicity (agitation, delirium, seizures, coma, hypotension).
    D) Monitor creatinine phosphokinase in patients with prolonged agitation, seizures or coma; monitor renal function and urine output in patients with rhabdomyolysis.
    E) Standard urine toxicology screen may not produce a positive result for many of the specific hallucinogenic amphetamines.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) Prehospital gastrointestinal decontamination is not recommended because of the potential for agitation and seizures.
    6.5.2) PREVENTION OF ABSORPTION
    A) SUMMARY
    1) Gastrointestinal decontamination is often NOT needed because the patient presents several hours after ingestion when absorption is complete. Give activated charcoal if recent ingestion or possibility of coingestants, and only if the patient can protect the airway or is intubated.
    B) ACTIVATED CHARCOAL
    1) CHARCOAL ADMINISTRATION
    a) Consider administration of activated charcoal after a potentially toxic ingestion (Chyka et al, 2005). Administer charcoal as an aqueous slurry; most effective when administered within one hour of ingestion.
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    6.5.3) TREATMENT
    A) PSYCHOMOTOR AGITATION
    1) INDICATION
    a) If patient is severely agitated, sedate with IV benzodiazepines.
    2) DIAZEPAM DOSE
    a) ADULT: 5 to 10 mg IV initially, repeat every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) CHILD: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    3) LORAZEPAM DOSE
    a) ADULT: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed (Manno, 2003).
    b) CHILD: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    4) Extremely large doses of benzodiazepines may be required in patients with severe intoxication in order to obtain adequate sedation. Titrate dose to clinical response and monitor for hypotension, CNS and respiratory depression, and the need for endotracheal intubation.
    5) Phenothiazines are not routinely recommended due to undesirable side effects (orthostatic hypotension and mental status changes).
    a) There are conflicting animal data on deleterious and beneficial effects of chlorpromazine. Chlorpromazine was shown to enhance the toxicity of some of these compounds (Grinspoon and Headblom, 1975) (Snyder et al, 1970) (Solursch and Clement, 1968).
    b) CHLORPROMAZINE
    1) In a dog study where chlorpromazine 10 mg/kg was given intravenously immediately following a lethal dose of MDA (20 mg/kg intravenously), all dogs survived for 48 hours. Chlorpromazine protected against acidosis, seizures, and hyperthermia (Davis et al, 1986).
    c) HALOPERIDOL
    1) Haloperidol has been protective in mice against lethality and hyperthermia caused by some but not all hallucinogenic amphetamines (Nichols et al, 1975; Davis & Borne, 1984; Paton et al, 1975).
    2) Dose: Up to 0.1 mg/kg
    3) There are no data in humans. The mechanism of action is presumed to be dopaminergic blockade. Because haloperidol lowers seizure threshold and predisposes to hyperthermia, benzodiazepines are preferred.
    6) Phenobarbital has been protective against lethality in mice but there are no human data (Thiessen & Cook, 1973b) (David & Borne, 1984) (Paton et al, 1975).
    B) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2010; Chin et al, 2008).
    5) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    6) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    7) CASE REPORT: Thiopentone 400 mg followed by neuromuscular blockade with an atracurium infusion were successfully used to control status epilepticus that was refractory to treatment with diazepam (Singarajah & Lavies, 1992).
    8) CASE REPORT: A chlormethiazole infusion (10 mg/kg/hour) was successfully used to treat seizures unresponsive to diazepam and haloperidol in a 13 month-old-child (Russell et al, 1992).
    9) RECURRING SEIZURES
    a) If seizures are not controlled by the above measures, patients will require endotracheal intubation, mechanical ventilation, continuous EEG monitoring, a continuous infusion of an anticonvulsant, and may require neuromuscular paralysis and vasopressor support. Consider continuous infusions of the following agents:
    1) MIDAZOLAM: ADULT DOSE: An initial dose of 0.2 mg/kg slow bolus, at an infusion rate of 2 mg/minute; maintenance doses of 0.05 to 2 mg/kg/hour continuous infusion dosing, titrated to EEG (Brophy et al, 2012). PEDIATRIC DOSE: 0.1 to 0.3 mg/kg followed by a continuous infusion starting at 1 mcg/kg/minute, titrated upwards every 5 minutes as needed (Loddenkemper & Goodkin, 2011).
    2) PROPOFOL: ADULT DOSE: Start at 20 mcg/kg/min with 1 to 2 mg/kg loading dose; maintenance doses of 30 to 200 mcg/kg/minute continuous infusion dosing, titrated to EEG; caution with high doses greater than 80 mcg/kg/minute in adults for extended periods of time (ie, longer than 48 hours) (Brophy et al, 2012); PEDIATRIC DOSE: IV loading dose of up to 2 mg/kg; maintenance doses of 2 to 5 mg/kg/hour may be used in older adolescents; avoid doses of 5 mg/kg/hour over prolonged periods because of propofol infusion syndrome (Loddenkemper & Goodkin, 2011); caution with high doses greater than 65 mcg/kg/min in children for extended periods of time; contraindicated in small children (Brophy et al, 2012).
    3) PENTOBARBITAL: ADULT DOSE: A loading dose of 5 to 15 mg/kg at an infusion rate of 50 mg/minute or lower; may administer additional 5 to 10 mg/kg. Maintenance dose of 0.5 to 5 mg/kg/hour continuous infusion dosing, titrated to EEG (Brophy et al, 2012). PEDIATRIC DOSE: A loading dose of 3 to 15 mg/kg followed by a maintenance dose of 1 to 5 mg/kg/hour (Loddenkemper & Goodkin, 2011).
    4) THIOPENTAL: ADULT DOSE: 2 to 7 mg/kg, at an infusion rate of 50 mg/minute or lower. Maintenance dose of 0.5 to 5 mg/kg/hour continuous infusing dosing, titrated to EEG (Brophy et al, 2012)
    b) Endotracheal intubation, mechanical ventilation, and vasopressors will be required (Brophy et al, 2012) and consultation with a neurologist is strongly advised.
    c) Neuromuscular paralysis (eg, rocuronium bromide, a short-acting nondepolarizing agent) may be required to avoid hyperthermia, severe acidosis, and rhabdomyolysis. If rhabdomyolysis is possible, avoid succinylcholine chloride, because of the risk of hyperkalemic-induced cardiac dysrhythmias. Continuous EEG monitoring is mandatory if neuromuscular paralysis is used (Manno, 2003).
    C) HYPERTENSIVE EPISODE
    1) SUMMARY: Hypertension often resolves once the patient is less agitated (ie, following sedative use). If hypertension persists, use of nitroprusside or a calcium channel blocker is suggested; use of beta-blockers is generally contraindicated since these agents may worsen vasospasm and result in hypertension (Shannon, 2000). As hypotension may develop later, a short acting titratable agent is preferred for treating hypertension.
    2) Monitor vital signs regularly. For mild/moderate hypertension without evidence of end organ damage, pharmacologic intervention is generally not necessary. Sedative agents such as benzodiazepines may be helpful in treating hypertension and tachycardia in agitated patients, especially if a sympathomimetic agent is involved in the poisoning.
    3) For hypertensive emergencies (severe hypertension with evidence of end organ injury (CNS, cardiac, renal), or emergent need to lower mean arterial pressure 20% to 25% within one hour), sodium nitroprusside is preferred. Nitroglycerin and phentolamine are possible alternatives.
    4) SODIUM NITROPRUSSIDE/INDICATIONS
    a) Useful for emergent treatment of severe hypertension secondary to poisonings. Sodium nitroprusside has a rapid onset of action, a short duration of action and a half-life of about 2 minutes (Prod Info NITROPRESS(R) injection for IV infusion, 2007) that can allow accurate titration of blood pressure, as the hypertensive effects of drug overdoses are often short lived.
    5) SODIUM NITROPRUSSIDE/DOSE
    a) ADULT: Begin intravenous infusion at 0.1 microgram/kilogram/minute and titrate to desired effect; up to 10 micrograms/kilogram/minute may be required (American Heart Association, 2005). Frequent hemodynamic monitoring and administration by an infusion pump that ensures a precise flow rate is mandatory (Prod Info NITROPRESS(R) injection for IV infusion, 2007). PEDIATRIC: Initial: 0.5 to 1 microgram/kilogram/minute; titrate to effect up to 8 micrograms/kilogram/minute (Kleinman et al, 2010).
    6) SODIUM NITROPRUSSIDE/SOLUTION PREPARATION
    a) The reconstituted 50 mg solution must be further diluted in 250 to 1000 mL D5W to desired concentration (recommended 50 to 200 mcg/mL) (Prod Info NITROPRESS(R) injection, 2004). Prepare fresh every 24 hours; wrap in aluminum foil. Discard discolored solution (Prod Info NITROPRESS(R) injection for IV infusion, 2007).
    7) SODIUM NITROPRUSSIDE/MAJOR ADVERSE REACTIONS
    a) Severe hypotension; headaches, nausea, vomiting, abdominal cramps; thiocyanate or cyanide toxicity (generally from prolonged, high dose infusion); methemoglobinemia; lactic acidosis; chest pain or dysrhythmias (high doses) (Prod Info NITROPRESS(R) injection for IV infusion, 2007). The addition of 1 gram of sodium thiosulfate to each 100 milligrams of sodium nitroprusside for infusion may help to prevent cyanide toxicity in patients receiving prolonged or high dose infusions (Prod Info NITROPRESS(R) injection for IV infusion, 2007).
    8) SODIUM NITROPRUSSIDE/MONITORING PARAMETERS
    a) Monitor blood pressure every 30 to 60 seconds at onset of infusion; once stabilized, monitor every 5 minutes. Continuous blood pressure monitoring with an intra-arterial catheter is advised (Prod Info NITROPRESS(R) injection for IV infusion, 2007).
    9) PHENTOLAMINE/INDICATIONS
    a) Useful for severe hypertension, particularly if caused by agents with alpha adrenergic agonist effects usually induced by catecholamine excess (Rhoney & Peacock, 2009).
    10) PHENTOLAMINE/ADULT DOSE
    a) BOLUS DOSE: 5 to 15 mg IV bolus repeated as needed (U.S. Departement of Health and Human Services, National Institutes of Health, and National Heart, Lung, and Blood Institute, 2004). Onset of action is 1 to 2 minutes with a duration of 10 to 30 minutes (Rhoney & Peacock, 2009).
    b) CONTINUOUS INFUSION: 1 mg/hr, adjusted hourly to stabilize blood pressure. Prepared by adding 60 mg of phentolamine mesylate to 100 mL of 0.9% sodium chloride injection; continuous infusion ranging from 12 to 52 mg/hr over 4 days has been used in case reports (McMillian et al, 2011).
    11) PHENTOLAMINE/PEDIATRIC DOSE
    a) 0.05 to 0.1 mg/kg/dose (maximum of 5 mg per dose) intravenously every 5 minutes until hypertension is controlled, then every 2 to 4 hours as needed (Singh et al, 2012; Koch-Weser, 1974).
    12) PHENTOLAMINE/ADVERSE EFFECTS
    a) Adverse events can include orthostatic or prolonged hypotension, tachycardia, dysrhythmias, angina, flushing, headache, nasal congestion, nausea, vomiting, abdominal pain and diarrhea (Rhoney & Peacock, 2009; Prod Info Phentolamine Mesylate IM, IV injection Sandoz Standard, 2005).
    13) CAUTION
    a) Phentolamine should be used with caution in patients with coronary artery disease because it may induce angina or myocardial infarction (Rhoney & Peacock, 2009).
    14) LABETALOL
    a) INTRAVENOUS INDICATIONS
    1) Consider if severe hypertension is unresponsive to short acting titratable agents such as sodium nitroprusside. Although labetalol has mixed alpha and beta adrenergic effects (Pearce & Wallin, 1994), it should be used cautiously if sympathomimetic agents are involved in the poisoning, as worsening hypertension may develop from alpha adrenergic effects.
    b) ADULT DOSE
    1) INTRAVENOUS BOLUS: Initial dose of 20 mg by slow IV injection over 2 minutes. Repeat with 40 to 80 mg at 10 minute intervals. Maximum total dose: 300 mg. Maximum effects on blood pressure usually occur within 5 minutes (Prod Info Trandate(R) IV injection, 2010).
    2) INTRAVENOUS INFUSION: Administer infusion after initial bolus, until desired blood pressure is reached. Administer IV at 2 mg/min of diluted labetalol solution (1 mg/mL or 2 mg/3 mL concentrations); adjust as indicated and continue until adequate response is achieved; usual effective IV dose range is 50 to 200 mg total dose; maximum dose: 300 mg. Prepare 1 mg/mL concentration by adding 200 mg labetalol (40 mL) to 160 mL of a compatible solution and administered at a rate of 2 mL/min (2 mg/min); also can be mixed as an approximate 2 mg/3 mL concentration by adding 200 mg labetalol (40 mL) to 250 mL of solution and administered at a rate of 3 mL/min (2 mg/min) (Prod Info Trandate(R) IV injection, 2010). Use of an infusion pump is recommended (Prod Info Trandate(R) IV injection, 2010).
    c) PEDIATRIC DOSE
    1) INTRAVENOUS: LOADING DOSE: 0.2 to 1 mg/kg, may repeat every 5 to 10 minutes (Hari & Sinha, 2011; Flynn & Tullus, 2009; Temple & Nahata, 2000; Fivush et al, 1997; Fivush et al, 1997; Bunchman et al, 1992). Maximum dose: 40 mg/dose (Hari & Sinha, 2011; Flynn & Tullus, 2009). CONTINUOUS INFUSION: 0.25 to 3 mg/kg/hour IV (Hari & Sinha, 2011; Flynn & Tullus, 2009; Temple & Nahata, 2000; Fivush et al, 1997; Miller, 1994; Deal et al, 1992; Bunchman et al, 1992).
    d) ADVERSE REACTIONS
    1) Common adverse events include postural hypotension, dizziness; fatigue; nausea; vomiting, sweating, and flushing (Pearce & Wallin, 1994).
    e) PRECAUTIONS
    1) Contraindicated in patients with bronchial asthma, congestive heart failure, greater than first degree heart block, cardiogenic shock, or severe bradycardia or other conditions associated with prolonged or severe hypotension. In patients with pheochromocytoma, labetalol should be used with caution because it has produced a paradoxical hypertensive response in some patients with this tumor (Prod Info Trandate(R) IV injection, 2010).
    2) Use caution in hepatic disease or intermittent claudication; effects of halothane may be enhanced by labetalol (Prod Info Trandate(R) IV injection, 2010). Labetalol should be stopped if there is laboratory evidence of liver injury or jaundice (Prod Info Trandate(R) IV injection, 2010).
    f) MONITORING PARAMETER
    1) Monitor blood pressure frequently during initial dosing and infusion (Prod Info Trandate(R) IV injection, 2010).
    15) ALPHA BLOCKERS: Phentolamine and phenoxybenzamine have been shown to block the pressor response in mice.
    16) BETA BLOCKERS: May lead to increased hypertension due to unopposed alpha stimulation, unless a vasodilator is given concurrently. May be used with nitroprusside. A short acting cardioselective agent such as esmolol is preferred.
    a) PINDOLOL: In a double-blind, placebo-controlled, crossover study, the use of pindolol (20 mg), a non-selective beta-blocker, on the cardiovascular response to MDMA (1.6 mg/kg) was evaluated in 16 subjects. Patients underwent 4 different treatment conditions (placebo-placebo, pindolol-placebo, placebo-MDMA or pindolol-MDMA) and a 2-week washout time between sessions (treatment duration of 6 to 10 weeks per subject). Vital signs were monitored in all patients during the treatment period. Pindolol prevented MDMA-induced increases in heart rate (HR peak value of 84 +/- 13 beats/min after MDMA vs 69 +/- 7 beats/min after pindolol-MDMA), but did not affect the mean arterial blood pressure (peak mean arterial blood pressure value of 115 +/- 11 mmHg after MDMA vs 114 +/- 11 mmHg after pindolol-MDMA) or change any adverse effects caused by MDMA use (Hysek et al, 2010).
    D) BODY TEMPERATURE ABOVE REFERENCE RANGE
    1) Severe hyperthermia is frequently associated with a fatal outcome (Henry et al, 1992) and should be treated aggressively.
    2) Place patient in a cool room.
    3) Minimize physical activity, sponge patient with tepid to cool water, and use fans to maximize evaporative heat loss.
    4) Place patient on a hypothermia blanket.
    5) Other methods include intubation and cool air ventilation, and gastric lavage with iced saline.
    6) Large doses of benzodiazepines may be needed to control neuromuscular hyperactivity.
    7) Immersion in ice water makes monitoring and resuscitation more difficult. It should be reserved for severe hyperthermia not responding to the above therapies.
    8) DANTROLENE
    a) Most patients respond well to sedation with benzodiazepines and aggressive cooling measures. Dantrolene may be considered in patients failing these therapies.
    b) A number of case reports describe the use of IV dantrolene (total dose: 1 to 10 mg/kg IV) to treat MDMA-induced hyperthermia by reducing the patient's muscular hyperactivity (Grunau et al, 2010; Singarajah & Lavies, 1992; Woods & Henry, 1992; Henry et al, 1992; Campkin & Davies, 1993; Webb & Williams, 1993; Logan et al, 1993; Tehan, 1993; Tehan, 1993; Barrett, 1993; Wake, 1995). In one systematic review, the most common dose used was 1 mg/kg or 80 mg IV and repeated as needed (Grunau et al, 2010).
    1) REVIEW: In a systematic review of the literature, 71 case reports of MDMA-induced hyperpyrexia were reviewed to assess the safety and efficacy of dantrolene. In both groups, dantrolene (n=26) and no dantrolene groups (n=45), patients were young adults and there were no reports of cyproheptadine use. Overall, survival was higher in the dantrolene group (21/26) compared with no dantrolene group (25/45). In particular, survival rates were higher among patients with extreme (greater than or equal to 42 degrees C) and severe (greater than or equal to 40 degrees C) elevations in temperature in the dantrolene group (8/13 and 10/10 survived, respectively) compared with no dantrolene group (no survivors (n=4) and 15/27 survived, respectively). More severe sequelae was also reported in the no dantrolene group. The most common dose for dantrolene was 1 mg/kg or 80 mg IV and repeated as needed (Grunau et al, 2010).
    9) NEUROMUSCULAR BLOCKADE
    a) The use of nondepolarizing neuromuscular blocking agents has been proposed to treat hyperthermia induced by hallucinogenic amphetamines (Barrett, 1992). In one case of MDEA toxicity anesthesia with propofol, atracurium and alfentanyl was associated with increasing hyperthermia which reversed on administration of dantrolene and active cooling measures (Tehan, 1993). Muscle biopsy later revealed no predisposition to malignant hyperthermia.
    10) CARVEDILOL
    a) An animal study evaluated the therapeutic potential of carvedilol for the prevention of reversal of MDMA-induced hyperthermia and rhabdomyolysis. Rats were treated with either nonselective (beta1 + beta2) adrenergic receptor antagonists propranolol or nadolol, or the alpha1 + beta 1,2,3 adrenergic receptor antagonist carvedilol before or after a thermogenic challenge of MDMA. MDMA caused a 35-fold increase in norepinephrine levels, a 20-fold increase in epinephrine levels, and a 2.4-fold increase in dopamine levels. There was no effect on the thermogenic response when propranolol (10 mg/kg IP) or nadolol (10 mg/kg IP) were administered 30 minutes before MDMA. However, this hyperthermic response was completely prevented when carvedilol (5 mg/kg IP) was administered 15 minutes before or after MDMA. When carvedilol was administered 60 minutes after MDMA, it completely reversed established hyperthermia and significantly attenuated the rise in creatine kinase induced by MDMA (Sprague et al, 2005).
    1) Jackson (2005) suggested that the required dose of carvedilol to duplicate the above findings in clinical care might be considerably higher than that currently used for cardiovascular indications in humans (Jackson, 2005).
    E) TACHYARRHYTHMIA
    1) Sedation with benzodiazepines to control agitation is sufficient in the vast majority of cases. Administer oxygen and intravenous fluids and correct hyperthermia as clinically indicated. If severe tachycardia persists and is associated with hemodynamic compromise or myocardial ischemia, additional therapy may be required, but this is unusual. Small incremental doses of labetalol may be useful because of combined alpha and beta blocking effects. A short acting agent such as esmolol may also be considered, however esmolol carries the risk of inducing hypertension due to unopposed alpha agonist effects of amphetamines in this setting.
    2) LABETALOL
    a) INTRAVENOUS INDICATIONS
    1) Consider if severe hypertension is unresponsive to short acting titratable agents such as sodium nitroprusside. Although labetalol has mixed alpha and beta adrenergic effects (Pearce & Wallin, 1994), it should be used cautiously if sympathomimetic agents are involved in the poisoning, as worsening hypertension may develop from alpha adrenergic effects.
    b) ADULT DOSE
    1) INTRAVENOUS BOLUS: Initial dose of 20 mg by slow IV injection over 2 minutes. Repeat with 40 to 80 mg at 10 minute intervals. Maximum total dose: 300 mg. Maximum effects on blood pressure usually occur within 5 minutes (Prod Info Trandate(R) IV injection, 2010).
    2) INTRAVENOUS INFUSION: Administer infusion after initial bolus, until desired blood pressure is reached. Administer IV at 2 mg/min of diluted labetalol solution (1 mg/mL or 2 mg/3 mL concentrations); adjust as indicated and continue until adequate response is achieved; usual effective IV dose range is 50 to 200 mg total dose; maximum dose: 300 mg. Prepare 1 mg/mL concentration by adding 200 mg labetalol (40 mL) to 160 mL of a compatible solution and administered at a rate of 2 mL/min (2 mg/min); also can be mixed as an approximate 2 mg/3 mL concentration by adding 200 mg labetalol (40 mL) to 250 mL of solution and administered at a rate of 3 mL/min (2 mg/min) (Prod Info Trandate(R) IV injection, 2010). Use of an infusion pump is recommended (Prod Info Trandate(R) IV injection, 2010).
    c) PEDIATRIC DOSE
    1) INTRAVENOUS: LOADING DOSE: 0.2 to 1 mg/kg, may repeat every 5 to 10 minutes (Hari & Sinha, 2011; Flynn & Tullus, 2009; Temple & Nahata, 2000; Fivush et al, 1997; Fivush et al, 1997; Bunchman et al, 1992). Maximum dose: 40 mg/dose (Hari & Sinha, 2011; Flynn & Tullus, 2009). CONTINUOUS INFUSION: 0.25 to 3 mg/kg/hour IV (Hari & Sinha, 2011; Flynn & Tullus, 2009; Temple & Nahata, 2000; Fivush et al, 1997; Miller, 1994; Deal et al, 1992; Bunchman et al, 1992).
    d) ADVERSE REACTIONS
    1) Common adverse events include postural hypotension, dizziness; fatigue; nausea; vomiting, sweating, and flushing (Pearce & Wallin, 1994).
    e) PRECAUTIONS
    1) Contraindicated in patients with bronchial asthma, congestive heart failure, greater than first degree heart block, cardiogenic shock, or severe bradycardia or other conditions associated with prolonged or severe hypotension. In patients with pheochromocytoma, labetalol should be used with caution because it has produced a paradoxical hypertensive response in some patients with this tumor (Prod Info Trandate(R) IV injection, 2010).
    2) Use caution in hepatic disease or intermittent claudication; effects of halothane may be enhanced by labetalol (Prod Info Trandate(R) IV injection, 2010). Labetalol should be stopped if there is laboratory evidence of liver injury or jaundice (Prod Info Trandate(R) IV injection, 2010).
    f) MONITORING PARAMETER
    1) Monitor blood pressure frequently during initial dosing and infusion (Prod Info Trandate(R) IV injection, 2010).
    3) TACHYCARDIA SUMMARY
    a) Evaluate patient to be sure that tachycardia is not a physiologic response to dehydration, anemia, hypotension, fever, sepsis, or hypoxia. Sinus tachycardia does not generally require treatment unless hemodynamic compromise develops.
    b) If therapy is required, a short acting, cardioselective agent such as esmolol is generally preferred (Prod Info BREVIBLOC(TM) intravenous injection, 2012).
    c) ESMOLOL/ADULT LOADING DOSE
    1) Infuse 500 micrograms/kilogram (0.5 mg/kg) IV over 1 minute (Neumar et al, 2010).
    d) ESMOLOL/ADULT MAINTENANCE DOSE
    1) Follow loading dose with infusion of 50 mcg/kg per minute (0.05 mg/kg per minute) (Neumar et al, 2010).
    2) EVALUATION OF RESPONSE: If response is inadequate, infuse second loading bolus of 0.5 mg/kg over 1 minute and increase the maintenance infusion to 100 mcg/kg (0.1 mg/kg) per minute. Reevaluate therapeutic effect, increase in the same manner if required to a maximum infusion rate of 300 mcg/kg (0.3 mg/kg) per minute (Neumar et al, 2010).
    3) The manufacturer recommends that a maximum of 3 loading doses be used (Prod Info BREVIBLOC(TM) intravenous injection, 2012).
    4) END POINT OF THERAPY: As the desired heart rate or blood pressure is approached, omit loading dose and adjust maintenance infusion as required (Prod Info BREVIBLOC(TM) intravenous injection, 2012).
    e) CAUTION
    1) Esmolol is a short acting beta-adrenergic blocking agent with negative inotropic effects. Esmolol should be avoided in patients with asthma, obstructive airway disease, decompensated heart failure and pre-excited atrial fibrillation (wide complex irregular tachycardia) or atrial flutter (Neumar et al, 2010).
    4) PINDOLOL: In a double-blind, placebo-controlled, crossover study, the use of pindolol (20 mg), a non-selective beta-blocker, on the cardiovascular response to MDMA (1.6 mg/kg) was evaluated in 16 subjects. Patients underwent 4 different treatment conditions (placebo-placebo, pindolol-placebo, placebo-MDMA or pindolol-MDMA) and a 2-week washout time between sessions (treatment duration of 6 to 10 weeks per subject). Vital signs were monitored in all patients during the treatment period. Pindolol prevented MDMA-induced increases in heart rate (HR peak value of 84 +/- 13 beats/min after MDMA vs 69 +/- 7 beats/min after pindolol-MDMA), but did not affect the mean arterial blood pressure (peak mean arterial blood pressure value of 115 +/- 11 mmHg after MDMA vs 114 +/- 11 mmHg after pindolol-MDMA) or change any adverse effects caused by MDMA use (Hysek et al, 2010).
    F) VENTRICULAR ARRHYTHMIA
    1) VENTRICULAR DYSRHYTHMIAS SUMMARY
    a) Obtain an ECG, institute continuous cardiac monitoring and administer oxygen. Evaluate for hypoxia, acidosis, and electrolyte disorders (particularly hypokalemia, hypocalcemia, and hypomagnesemia). Lidocaine and amiodarone are generally first line agents for stable monomorphic ventricular tachycardia, particularly in patients with underlying impaired cardiac function. Amiodarone should be used with caution if a substance that prolongs the QT interval and/or causes torsades de pointes is involved in the overdose. Unstable rhythms require immediate cardioversion.
    2) LIDOCAINE
    a) LIDOCAINE/DOSE
    1) ADULT: 1 to 1.5 milligrams/kilogram via intravenous push. For refractory VT/VF an additional bolus of 0.5 to 0.75 milligram/kilogram can be given at 5 to 10 minute intervals to a maximum dose of 3 milligrams/kilogram (Neumar et al, 2010). Only bolus therapy is recommended during cardiac arrest.
    a) Once circulation has been restored begin a maintenance infusion of 1 to 4 milligrams per minute. If dysrhythmias recur during infusion repeat 0.5 milligram/kilogram bolus and increase the infusion rate incrementally (maximal infusion rate is 4 milligrams/minute) (Neumar et al, 2010).
    2) CHILD: 1 milligram/kilogram initial bolus IV/IO; followed by a continuous infusion of 20 to 50 micrograms/kilogram/minute (de Caen et al, 2015).
    b) LIDOCAINE/MAJOR ADVERSE REACTIONS
    1) Paresthesias; muscle twitching; confusion; slurred speech; seizures; respiratory depression or arrest; bradycardia; coma. May cause significant AV block or worsen pre-existing block. Prophylactic pacemaker may be required in the face of bifascicular, second degree, or third degree heart block (Prod Info Lidocaine HCl intravenous injection solution, 2006; Neumar et al, 2010).
    c) LIDOCAINE/MONITORING PARAMETERS
    1) Monitor ECG continuously; plasma concentrations as indicated (Prod Info Lidocaine HCl intravenous injection solution, 2006).
    3) AMIODARONE
    a) AMIODARONE/INDICATIONS
    1) Effective for the control of hemodynamically stable monomorphic ventricular tachycardia. Also recommended for pulseless ventricular tachycardia or ventricular fibrillation in cardiac arrest unresponsive to CPR, defibrillation and vasopressor therapy (Link et al, 2015; Neumar et al, 2010). It should be used with caution when the ingestion involves agents known to cause QTc prolongation, such as fluoroquinolones, macrolide antibiotics or azoles, and when ECG reveals QT prolongation suspected to be secondary to overdose (Prod Info Cordarone(R) oral tablets, 2015).
    b) AMIODARONE/ADULT DOSE
    1) For ventricular fibrillation or pulseless VT unresponsive to CPR, defibrillation, and a vasopressor therapy give an initial dose of 300 mg IV followed by 1 dose of 150 mg IV. For stable ventricular tachycardias: Infuse 150 milligrams over 10 minutes, and repeat if necessary. Follow by a 1 milligram/minute infusion for 6 hours, then a 0.5 milligram/minute. Maximum total dose over 24 hours is 2.2 grams (Neumar et al, 2010).
    c) AMIODARONE/PEDIATRIC DOSE
    1) Infuse 5 milligrams/kilogram as a bolus for pulseless ventricular tachycardia or ventricular fibrillation; may repeat twice up to 15 mg/kg. Infuse 5 milligrams/kilogram over 20 to 60 minutes for perfusing tachycardias. Maximum single dose is 300 mg. Routine use with other drugs that prolong the QT interval is NOT recommended (Kleinman et al, 2010).
    d) ADVERSE EFFECTS
    1) Hypotension and bradycardia are the most common adverse effects (Neumar et al, 2010).
    G) HYPOTENSIVE EPISODE
    1) SUMMARY
    a) Infuse 10 to 20 milliliters/kilogram of isotonic fluid and keep the patient supine. If hypotension persists, administer dopamine or norepinephrine. Consider central venous pressure monitoring to guide further fluid therapy.
    2) DOPAMINE
    a) DOSE: Begin at 5 micrograms per kilogram per minute progressing in 5 micrograms per kilogram per minute increments as needed (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). If hypotension persists, dopamine may need to be discontinued and a more potent vasoconstrictor (eg, norepinephrine) should be considered (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    b) CAUTION: If ventricular dysrhythmias occur, decrease rate of administration (Prod Info dopamine hcl, 5% dextrose IV injection, 2004). Extravasation may cause local tissue necrosis, administration through a central venous catheter is preferred (Prod Info dopamine hcl, 5% dextrose IV injection, 2004).
    3) NOREPINEPHRINE
    a) PREPARATION: 4 milligrams (1 amp) added to 1000 milliliters of diluent provides a concentration of 4 micrograms/milliliter of norepinephrine base. Norepinephrine bitartrate should be mixed in dextrose solutions (dextrose 5% in water, dextrose 5% in saline) since dextrose-containing solutions protect against excessive oxidation and subsequent potency loss. Administration in saline alone is not recommended (Prod Info norepinephrine bitartrate injection, 2005).
    b) DOSE
    1) ADULT: Dose range: 0.1 to 0.5 microgram/kilogram/minute (eg, 70 kg adult 7 to 35 mcg/min); titrate to maintain adequate blood pressure (Peberdy et al, 2010).
    2) CHILD: Dose range: 0.1 to 2 micrograms/kilogram/minute; titrate to maintain adequate blood pressure (Kleinman et al, 2010).
    3) CAUTION: Extravasation may cause local tissue ischemia, administration by central venous catheter is advised (Peberdy et al, 2010).
    H) VASOSPASM
    1) Heparinization and intravenous or intraarterial nitroprusside have been used with success (Bowen et al, 1983).
    I) RHABDOMYOLYSIS
    1) SUMMARY: Early aggressive fluid replacement is the mainstay of therapy and may help prevent renal insufficiency. Diuretics such as mannitol or furosemide may be added if necessary to maintain urine output but only after volume status has been restored as hypovolemia will increase renal tubular damage. Urinary alkalinization is NOT routinely recommended.
    2) Initial treatment should be directed towards controlling acute metabolic disturbances such as hyperkalemia, hyperthermia, and hypovolemia. Control seizures, agitation, and muscle contractions (Erdman & Dart, 2004).
    3) FLUID REPLACEMENT: Early and aggressive fluid replacement is the mainstay of therapy to prevent renal failure. Vigorous fluid replacement with 0.9% saline (10 to 15 mL/kg/hour) is necessary even if there is no evidence of dehydration. Several liters of fluid may be needed within the first 24 hours (Walter & Catenacci, 2008; Camp, 2009; Huerta-Alardin et al, 2005; Criddle, 2003; Polderman, 2004). Hypovolemia, increased insensible losses, and third spacing of fluid commonly increase fluid requirements. Strive to maintain a urine output of at least 1 to 2 mL/kg/hour (or greater than 150 to 300 mL/hour) (Walter & Catenacci, 2008; Camp, 2009; Erdman & Dart, 2004; Criddle, 2003). To maintain a urine output this high, 500 to 1000 mL of fluid per hour may be required (Criddle, 2003). Monitor fluid input and urine output, plus insensible losses. Monitor for evidence of fluid overload and compartment syndrome; monitor serum electrolytes, CK, and renal function tests.
    4) DIURETICS: Diuretics (eg, mannitol or furosemide) may be needed to ensure adequate urine output and to prevent acute renal failure when used in combination with aggressive fluid therapy. Loop diuretics increase tubular flow and decrease deposition of myoglobin. These agents should be used only after volume status has been restored, as hypovolemia will increase renal tubular damage. If the patient is maintaining adequate urine output, loop diuretics are not necessary (Vanholder et al, 2000).
    5) URINARY ALKALINIZATION: Alkalinization of the urine is not routinely recommended, as it has never been documented to reduce nephrotoxicity, and may cause complications such as hypocalcemia and hypokalemia (Walter & Catenacci, 2008; Huerta-Alardin et al, 2005; Brown et al, 2004; Polderman, 2004). Retrospective studies have failed to demonstrate any clinical benefit from the use of urinary alkalinization (Brown et al, 2004; Polderman, 2004; Homsi et al, 1997).
    J) FLUID/ELECTROLYTE BALANCE REGULATION
    1) Hyponatremia, if secondary to SIADH from MDMA, may not respond to administration of 0.9% NaCl. Fluid restriction is generally the treatment for SIADH. If hemodynamic instability is present, then fluid administration is necessary. If severe or life threatening hyponatremia develops, slow correction with 3% NaCl may be considered (Ajaelo et al, 1998).
    2) MONITORING PARAMETERS - Monitor serum sodium concentrations. Central venous pressure monitoring and/or pulmonary capillary wedge pressure may be indicated to evaluate extracellular fluid volume; urine osmolarity is generally elevated in hyponatremia secondary to SIADH and may NOT be clinically useful in assessing extracellular fluid volume. Likewise, urine sodium concentrations can be misleading because alterations are generally not evident for as long as 24 hours after acute exposure (Zeneberg & Goldfarb, 2000).
    K) CYPROHEPTADINE
    1) SEROTONIN ANTAGONISTS
    a) Since many of the toxic reactions to MDMA (hyperthermia, tachycardia, DIC, renal failure) resemble the serotonin syndrome, it has been suggested, although clinical data are lacking, that the use of nonselective serotonin antagonists such as methysergide or cyproheptadine may be useful as adjunctive treatment in MDMA overdoses (Ames & Wirshing, 1993; Friedman, 1993).
    L) SEROTONIN SYNDROME
    1) SUMMARY
    a) Benzodiazepines are the mainstay of therapy. Cyproheptadine, a 5-HT antagonist, is also commonly used. Severe cases have been managed with benzodiazepine sedation and neuromuscular paralysis with non-depolarizing agents(Claassen & Gelissen, 2005).
    2) HYPERTHERMIA
    a) Control agitation and muscle activity. Undress patient and enhance evaporative heat loss by keeping skin damp and using cooling fans.
    b) MUSCLE ACTIVITY: Benzodiazepines are the drug of choice to control agitation and muscle activity. DIAZEPAM: ADULT: 5 to 10 mg IV every 5 to 10 minutes as needed, monitor for respiratory depression and need for intubation. CHILD: 0.25 mg/kg IV every 5 to 10 minutes; monitor for respiratory depression and need for intubation.
    c) Non-depolarizing paralytics may be used in severe cases.
    3) CYPROHEPTADINE
    a) Cyproheptadine is a non-specific 5-HT antagonist that has been shown to block development of serotonin syndrome in animals (Sternbach, 1991). Cyproheptadine has been used in the treatment of serotonin syndrome (Mills, 1997; Goldberg & Huk, 1992). There are no controlled human trials substantiating its efficacy.
    b) ADULT: 12 mg initially followed by 2 mg every 2 hours if symptoms persist, up to a maximum of 32 mg in 24 hours. Maintenance dose 8 mg orally repeated every 6 hours (Boyer & Shannon, 2005).
    c) CHILD: 0.25 mg/kg/day divided every 6 hours, maximum dose 12 mg/day (Mills, 1997).
    4) HYPERTENSION
    a) Monitor vital signs regularly. For mild/moderate asymptomatic hypertension, pharmacologic intervention is usually not necessary.
    5) HYPOTENSION
    a) Administer 10 to 20 mL/kg 0.9% saline bolus and place patient supine. Further fluid therapy should be guided by central venous pressure or right heart catheterization to avoid volume overload.
    b) Pressor agents with dopaminergic effects may theoretically worsen serotonin syndrome and should be used with caution. Direct acting agents (norepinephrine, epinephrine, phentolamine) are theoretically preferred.
    c) NOREPINEPHRINE
    1) PREPARATION: Add 4 mL of 0.1% solution to 1000 mL of dextrose 5% in water to produce 4 mcg/mL.
    2) INITIAL DOSE
    a) ADULT: 2 to 3 mL (8 to 12 mcg)/minute.
    b) ADULT or CHILD: 0.1 to 0.2 mcg/kg/min. Titrate to maintain adequate blood pressure.
    3) MAINTENANCE DOSE
    a) 0.5 to 1 mL (2 to 4 mcg)/minute.
    6) SEIZURES
    a) DIAZEPAM
    1) MAXIMUM RATE: Administer diazepam IV over 2 to 3 minutes (maximum rate: 5 mg/min).
    2) ADULT DIAZEPAM DOSE: 5 to 10 mg initially, repeat every 5 to 10 minutes as needed. Monitor for hypotension, respiratory depression and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after diazepam 30 milligrams.
    3) PEDIATRIC DIAZEPAM DOSE: 0.2 to 0.5 mg/kg, repeat every 5 minutes as needed. Monitor for hypotension, respiratory depression and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after diazepam 10 milligrams in children over 5 years or 5 milligrams in children under 5 years of age.
    4) RECTAL USE: If an intravenous line cannot be established, diazepam may be given per rectum (not FDA approved), or lorazepam may be given intramuscularly.
    b) LORAZEPAM
    1) MAXIMUM RATE: The rate of IV administration of lorazepam should not exceed 2 mg/min (Prod Info Ativan(R), 1991).
    2) ADULT LORAZEPAM DOSE: 2 to 4 mg IV. Initial doses may be repeated in 10 to 15 minutes, if seizures persist (Prod Info ATIVAN(R) injection, 2003).
    3) PEDIATRIC LORAZEPAM DOSE: 0.1 mg/kg IV push (range: 0.05 to 0.1 mg/kg; maximum dose 4 mg); may repeat dose in 5 to 10 minutes if seizures continue. It has also been given rectally at the same dose in children with no IV access (Sreenath et al, 2010; Chin et al, 2008; Wheless, 2004; Qureshi et al, 2002; De Negri & Baglietto, 2001; Mitchell, 1996; Appleton, 1995; Giang & McBride, 1988).
    c) RECURRING SEIZURES
    1) If seizures cannot be controlled with diazepam or recur, give phenobarbital or propofol.
    d) PHENOBARBITAL
    1) SERUM LEVEL MONITORING: Monitor serum levels over next 12 to 24 hours for maintenance of therapeutic levels (15 to 25 mcg/mL).
    2) ADULT PHENOBARBITAL LOADING DOSE: 600 to 1200 mg of phenobarbital IV initially (10 to 20 mg/kg) diluted in 60 mL of 0.9% saline given at 25 to 50 mg/minute.
    3) ADULT PHENOBARBITAL MAINTENANCE DOSE: Additional doses of 120 to 240 mg may be given every 20 minutes.
    4) MAXIMUM SAFE ADULT PHENOBARBITAL DOSE: No maximum safe dose has been established. Patients in status epilepticus have received as much as 100 mg/min until seizure control was achieved or a total dose of 10 mg/kg.
    5) PEDIATRIC PHENOBARBITAL LOADING DOSE: 15 to 20 mg/kg of phenobarbital intravenously at a rate of 25 to 50 mg/min.
    6) PEDIATRIC PHENOBARBITAL MAINTENANCE DOSE: Repeat doses of 5 to 10 mg/kg may be given every 20 minutes.
    7) MAXIMUM SAFE PEDIATRIC PHENOBARBITAL DOSE: No maximum safe dose has been established. Children in status epilepticus have received doses of 30 to 120 mg/kg within 24 hours. Vasopressors and mechanical ventilation were needed in some patients receiving these doses.
    8) NEONATAL PHENOBARBITAL LOADING DOSE: 20 to 30 mg/kg IV at a rate of no more than 1 mg/kg/min in patients with no preexisting phenobarbital serum levels.
    9) NEONATAL PHENOBARBITAL MAINTENANCE DOSE: Repeat doses of 2.5 mg/kg every 12 hours may be given; adjust dosage to maintain serum levels of 20 to 40 mcg/mL.
    10) MAXIMUM SAFE NEONATAL PHENOBARBITAL DOSE: Doses of up to 20 mg/kg/min up to a total of 30 mg/kg have been tolerated in neonates.
    11) CAUTION: Adequacy of ventilation must be continuously monitored in children and adults. Intubation may be necessary with increased doses.
    7) CHLORPROMAZINE
    a) Chlorpromazine is a 5-HT2 receptor antagonist that has been used to treat cases of serotonin syndrome (Graham, 1997; Gillman, 1996). Controlled human trial documenting its efficacy are lacking.
    b) ADULT: 25 to 100 mg intramuscularly repeated in 1 hour if necessary.
    c) CHILD: 0.5 to 1 mg/kg repeated as needed every 6 to 12 hours not to exceed 2 mg/kg/day.
    8) NOT RECOMMENDED
    a) BROMOCRIPTINE: It has been used in the treatment of neuroleptic malignant syndrome but is NOT RECOMMENDED in the treatment of serotonin syndrome as it has serotonergic effects (Gillman, 1997). In one case the use of bromocriptine was associated with a fatal outcome (Kline et al, 1989).
    M) FULMINANT HEPATIC FAILURE
    1) FPSA/PROMETHEUS: A 27-year-old man presented to the ED comatose and tachycardic (HR 112 beats/min) after injecting himself with approximately 0.5 grams of heroin and approximately 1 gram of cocaine and ingesting 3 tablets of MDMA (300 mg). Lab evaluation showed that the patient had rhabdomyolysis, lactic acidosis, hypoglycemia, and progressive fulminant hepatic failure. Liver transplantation was declined by the transplant surgeon due to the patient's history of illicit drug use. Despite continuous venovenous hemofiltration, arterial hyperammonemia occurred followed by grade IV encephalopathy. A cranial CT scan showed cerebral edema and transtentorial herniation. Due to possible irreversible neurological deterioration, experimental therapy using a fractionated plasma separation and adsorption system (FPSA/Prometheus), a hemodialysis-based system, was initiated. An activated charcoal column in the FPSA circuit was used and changed 1 to 2 times daily. The blood flow rate was 180 to 220 mL/min, the dialysate flow was 300 mL/min, and dialysate sodium was 142 mmol/L in order to avoid exacerbation of the cerebral edema.
    a) Within 12 hours of FPSA initiation, the patient's arterial ammonia levels normalized. Within 12 hours of the second treatment period, the intracranial pressure fell from 47 mm Hg initially to 27 mm Hg. A repeat cranial CT scan, performed 4 days after the initial scan, showed that the cerebral edema was greatly reduced, but several small ischemic lesions were identified. The encephalopathy gradually resolved and the patient recovered following neurological rehab to treat sequelae of herniation (ie, dysphagia, dysarthria, motor weakness and gait disturbances) and was discharged to home six weeks later to live independently (Kramer et al, 2003).
    2) In an in vitro model, alpha MeDA (a major metabolite of MDA) caused marked depletion of glutathione and loss of hepatocellular viability(Carvalho et al, 2004). This suggests that acetylcysteine might be useful in treating or preventing MDA-induced hepatotoxicity; however this has not been evaluated in clinical studies.
    N) ANGIOEDEMA
    1) ICATIBANT: A 30-year-old man who was an occasional user of MDMA and cannabis, presented with progressive dyspnea, dysphagia, and difficulty with phonation an hour after ingesting an unknown amount of MDMA. Physical examination revealed an edematous uvula, without bronchospasms or rashes. Treatment with methylprednisolone and dexclorpheniramine did not improve his symptoms. Urine toxicology testing was positive for methamphetamine and MDMA. His symptoms worsened 4 hours later and despite further treatment with IM epinephrine and IV hydrocortisone, his symptoms persisted. At this time, angioedema mediated with bradykinin was suspected and he was treated with 30 mg of icatibant SubQ which resulted in an improvement in his condition within 30 minutes. A decrease in the size of uvula was noted in 2 hours. He recovered completely 8 hours after receiving icatibant. Complement C3 and C4 levels and tryptase were normal 3 days after presentation (Escalante et al, 2015).

Enhanced Elimination

    A) LACK OF EFFECT
    1) Hemodialysis and hemoperfusion are not of value.
    B) OSMOTIC DIURESIS
    1) Osmotic diuresis with mannitol was used to treat a patient with MDMA-associated symptomatic hyponatremia (Ghatol & Kazory, 2012).
    a) CASE REPORT: A 20-year-old woman presented with an altered mental status and rapidly progressive drowsiness after ingesting 2 tablets of double-stack Ecstasy (273 mg each) a few hours prior to admission. She was hyperthermic (38.6 degrees C) and unable to respond to verbal or painful stimuli. Laboratory results showed hyponatremia (sodium 123 mmol/L) and lactic acidosis (lactic acid 6.2 mmol/L). A head CT scan showed severe parenchymal edema with effacement of basal cisterns and crowding at the level of the fourth ventricle due to mass effect. Following a diagnosis of cerebral edema due to MDMA-induced acute severe hyponatremia, she received a bolus of mannitol 20% (initial, 2 g/kg over 60 min; a second bolus of 0.5 g/kg a few hours later was given) and a normal saline infusion. Her neurological status, urine output, and serum sodium concentrations gradually improved. About a day later, a second head CT scan revealed significant improvement in cerebral edema with better visualization of the sulci. Her mental status and neurological examination, as well as serum sodium concentration (sodium 139 mmol/L), were normalized 2 days later (Ghatol & Kazory, 2012).

Case Reports

    A) ADULT
    1) MDMA
    a) MDMA, 3,4-methylenedioxymethamphetamine, is the N-methyl derivative of MDA. In low doses, amphetamine-like effects and changes in consciousness occur. Severe anxiety reactions, paranoia, sleep disturbances, hallucinations, depression, and fearfulness have been reported.
    b) A 39-year-old woman died from myocardial damage and multiorgan failure 7 days after ingesting one pill of MDMA. Serum blood level of MDMA was 1.2 mg/L on presentation (Sano et al, 2009).
    c) A death has been reported in a psychoanalyst with preexisting heart disease 30 to 60 minutes after taking 200 mg.
    d) Hallucinations, confusion, and agitation were reported in a 32-year-old woman who ingested approximately 200 mg of MDMA. Hypotension, tachycardia, hyperthermia, coma, pulmonary edema, and hypoxia rapidly ensued. Over the next 5 days complications developed, including rhabdomyolysis, leukocytosis, coagulopathy, toxic hepatitis, and a herpetic-like rash. Serum MDMA level was 7 mcg/mL (Brown et al, 1986).
    e) Twelve hours after ingestion of an unknown quantity of MDMA, a 53-year-old man became acutely hyperthermic (107.2 degrees F) with signs of rhabdomyolysis. The patient died 5 days after exposure secondary to multiorgan failure which included adult respiratory distress syndrome, disseminated intravascular coagulation, and acute renal failure. Postmortem MDMA plasma concentration was 3.05 mg/L (Walubo & Seger, 1999).
    2) 4-METHYL AMINOREX
    a) A 37-year-old man was found dead with a plastic bag of white powder in his pocket. The bag was found to contain 4-methyl aminorex; the cause of death was accidental overdose leading to cerebral and pulmonary edema, respiratory paralysis, and death. Drug concentrations present in blood and urine were: diazepam 0.8 mg/L (therapeutic level) and 4-methyl aminorex in blood 21.3 mg/L and urine 12.3 mg/L (Davis & Brewster, 1988).
    3) COMBINED INGESTION (MDA and 2C-I)
    a) A 39-year-old-woman presented with rapidly diminishing mental status, agitation, hypothermia, emesis, urinary incontinence, severe hypertension, vasoconstriction and exterior posturing after ingesting MDA (3,4-methylenedioxyamphetamine) and 2C-I (2,5-dimethoxy-4-iodophenethylamine). Drug concentrations measured in the urine were: MDA 5.56 mg/L and 2C-I 0.311mg/L. A head CT showed massive intraventricular hemorrhaging and underlying Moyamoya. One month after ingestion, the patient developed sympathetic storming and seizures. A tracheostomy was placed for long-term mechanical ventilation. After 4 months in the hospital, the patient was transferred to a skilled nursing facility with quadriplegia and minimal mental improvement. She remained severely disabled (Drees et al, 2009).
    B) PEDIATRIC
    1) Approximately 40 minutes after ingesting a portion of an ecstasy pill, a 14-month-old child developed a generalized convulsion with opisthotonos, hyperthermia (38 degrees C), hypertension, tachycardia (130 beats/min), ventricular extrasystoles, tachypnea (50 breaths/min), and mydriasis. The urine levels of amphetamine/methamphetamine was greater than 16 mg/L 5 hours after ingestion. The serum level of MDMA was 0.591 mg/L 8 hours after ingestion. He continued to have hypertension, tachycardia and long periods of trigeminy, without hemodynamic repercussion during the first 12 hours. Following supportive care, the child recovered with no further sequelae (Melian et al, 2004).
    2) An 8-month-old boy presented with generalized seizure activity, tachycardia (210 beats/min), hypertension (BP 125/70 mmHg), and hyperthermia (38.9 degrees C) after accidentally ingesting 1 ecstasy tablet. Treatment with benzodiazepines, body cooling and fluids resulted in a resolution of all abnormal parameters 6 hours after admission (Eifinger et al, 2008).

Summary

    A) Severe toxicity has developed at recreational doses in some individuals. Typical recreational doses include: DDPR 1 to 2 mg, DMA 20 to 50 mg, DOB 1 to 5 mg, DOET 0.75 to 4 mg, DOI 0.8 to 2 mg, DOM 2 to 15 mg, MDA 40 to 150 mg, MDMA 50 to 150 mg, MMDA 50 to 100 mg, DMMDA 2 to 3 mg, PMA 50 mg, TMA 50 to 150 mg, 2CB 16 to 30 mg, 2,4,5 TMA 20 mg, 2,4,6 TMA 30 to 40 mg.
    B) FATALITIES: Fatalities have been reported after ingestion of a single capsule of MDMA. Deaths have been associated with the development of hyperthermia following MDMA intoxication.
    1) An adult found at home survived a massive ingestion of ecstasy (50 tablets: each containing 50 to 150 mg of MDMA), oxazepam (10 mg), and alcohol (5 units).
    2) Deaths have also been associated with other agents and include 2C-T-7, Bromo-dragonfly and PMA/PMMA.
    C) SPECIAL SITUATION: Law enforcement and emergency medical services that enter clandestine phenylethylamine laboratories may suffer from mucous membrane and respiratory symptoms. Surface decontamination and symptomatic and supportive care is the treatment. Explosions in clandestine laboratories are common and put those in proximity at risk for burns and traumatic injuries. Children that present from clandestine laboratories are primarily at risk for abuse and neglect.

Therapeutic Dose

    7.2.1) ADULT
    A) GENERAL/SUMMARY
    1) MDMA - A typical dose or "hit" of MDMA (ecstasy) is 100 milligrams, but can range from 50 to 250 milligrams (1 to 4 milligrams/kilogram). Doses of 50 milligrams have reportedly provided the desired CNS effects (e.g., feeling relaxed, lucid, peaceful) (Shannon, 2000).
    2) Unlike other street drugs, MDMA is usually available as pharmaceutical-grade tablets that are illegally brought into the US; however "home made" ecstasy may contain a range of other substances (Shannon, 2000).
    B) SPECIFIC SUBSTANCE
    1) SUMMARY - Representative hallucinogenic amphetamines and their usual oral doses are listed below.
    a) Because of their sympathomimetic effects, the toxic to "therapeutic" ratio is quite low.
    b) Deaths which occur soon after administration are presumably due to cardiac dysrhythmias, seizures, and CNS depression. Late death (24 to 48 hours after administration) results from a syndrome resembling malignant hyperthermia.
    2)
    TABLE 1: (Glennon & Rosecrans, 1982)
    ABBREV CHEMICAL NAMEHALLUCINOGENICDOSE (mg)
    DDPR4-propyl DMA1 to 2
    DMA(1)2,5-dimethoxyamphetamine20 to 50
    DOB(8)4-bromo-DMA1 to 5
    DOET(2)4-ethyl-DMA0.75 to 4
    DOI4-iodo-DMA0.8 to 2
    DOM(3)4-methyl-DMA2 to 15
    MDA(4)3,4-methylenedioxyamphetamine40 to 150
    MDEA(7)N-ethyl MDA .
    MDMA3,4-methylenedioxymethamphetamine50 to 150.
    MMDA(5)3-methoxy-4,5-MDA50 to 100
    DMMDA(6)2,5-dimethoxy MDA2 to 3
    PMAp-methoxyamphetamine50
    TMA3,4,5-trimethoxyamphetamine50 to 150
    2CB2,5-Dimethoxy-4-bromo-phenethylamine16 to 30
    2,3,4 TMA2,3,4 trimethoxyamphetamineinactive at low doses
    2,4,5 TMA2,4,5-trimethoxyamphetamine20
    2,4,6 TMA2,4,6 trimethoxyamphetamine30 to 40
    (1) 2,4-DMA is slightly less potent.
    (2) 4-propyl DMA (DOP) is similar in potency.
    (3) X-alkyl and N-alkyl analogs are also active.
    (4) N-alkyl analogs are similar in potency (e.g., MDE or n-ethyl-MDA).
    (5) 2-methoxy MDA's are 10 to 20 times more potent.
    (6) DMMDA's with methoxy and methylenedioxy groups at other positions are less potent.
    (7) MDEA is reported to besimilar to MDMA with more rapid onset and shorter duration. (Stone et al, 1987).
    (8) DOB is reported to have an onset of one hour and duration of 12 to 24 hours.

Minimum Lethal Exposure

    A) SPECIFIC SUBSTANCE
    1) MDA
    a) Deaths have been reported following ingestions of up to 1200 mg, resulting in blood levels of more than 100 mcg percent (Climko et al, 1986-7).
    2) MDMA/MDEA
    a) CASE REPORTS
    1) A 29-year-old man with no history of heart disease, hypertension or other cardiovascular risk factors died of a Type 1 aortic dissection approximately 48 hours after ingesting 1 ecstasy tablet and a large quantity of alcohol. Initial symptoms did not begin until approximately 36 hours after exposure (Duflou & Mark, 2000).
    2) An adult with AIDS died at a party within 4.5 hours of ingesting 2.5 tablets (total dose 180 mg) of MDMA and alcohol. The patient collapsed and immediate resuscitation attempts were unsuccessful. The authors suggested that ritonavir (recently added to the patient's drug regimen), a potential inhibitor of CYP2D6, increased the MDMA level to a toxic concentration (4.56 mg/L at autopsy versus an anticipated level of 0.5 mg/L for a 180 mg dose) (Henry & Hill, 1998).
    3) An 18-year-old woman who ingested an estimated 150 mg of MDMA in combination with alcohol developed ventricular fibrillation and died; postmortem MDMA levels were 1 mg/L (Dowling et al, 1987).
    4) Four other deaths associated with MDMA or MDEA had other causes or contributing factors (electrical shock, trauma, asthma, cardiomyopathy) (Dowling et al, 1987).
    5) A 16-year-old girl died of hyperpyrexia, coagulopathy, and rhabdomyolysis after ingestion of one tablet of "ecstasy" (strength unknown) (Chadwick et al, 1991).
    6) An 18-year-old man died following ingestion of 3 tablets of "ecstasy". Serum MDMA concentration was 1.26 mg/L on admission (Campkin & Davies, 1992).
    7) In a series of 7 fatalities related to MDMA use, the doses ingested ranged from 1 to 5 tablets (unknown dose in 2 patients). MDMA plasma concentration ranged from 0.11 mg/L to 1.26 mg/L (Henry et al, 1992).
    3) 2C-T-7
    a) CASE REPORT: A 20-year-old man died after insufflating 35 mg of 2C-T-7. Postmortem values were as follows: heart blood 57 ng/mL; femoral blood 100 ng/mL; urine 1120 ng/mL; liver 854 ng/g. 2,5-dimethoxy-4-n-propylthiophenethylamine (2C-T-7) is structurally and pharmacodynamically similar to methylenedioxymethamphetamine (MDMA) (Curtis et al, 2003).
    4) BROMO-DRAGONFLY
    a) CASE REPORT: An 18-year-old woman was found dead by her boyfriend after they both had ingested 1 mL each of an LSD-like liquid that was later identified as Bromo-Dragonfly. The concentration of the liquid was determined to be 0.69 mg/mL; ingestion amount was estimated to be 700 mcg of Bromo-Dragonfly. Postmortem concentrations of the substance were: femoral blood 4.7 +/- 0.7 mcg/kg; urine 22 +/- 2 mcg/kg, and it was also detected in the victim's liver blood (Andreasen et al, 2009).
    5) PMA/PMMA
    a) Investigations of fatal ingestions of PMA or PMMA have concluded that doses of 50 mg or greater can result in spontaneous and life-threatening hypertension and hyperthermia (Becker et al, 2003).
    b) CASE SERIES: Paramethoxyamphetamine (PMA), a methoxylated phenethylamine derivative that is structurally related to MDMA, produced severe hyperthermia and CNS effects in young adults; of the 6 cases reported all resulted in death (Felgate et al, 1998).
    c) CASE SERIES: Thirty-two cases of PMA-related deaths have been reported. Blood concentrations ranged from 0.2 mg/L to 5.7 mg/L, and the deaths which involved ingestion of PMA had a dosage range of 50 mg to 90 mg (Caldicott et al, 2003).

Maximum Tolerated Exposure

    A) SUMMARY
    1) MDMA: Low doses of ecstasy (approximately 50 mg) can produce undesirable effects (nausea, diaphoresis, anorexia, tremor, myoclonus, tics, nystagmus, hypertension, urinary retention, ataxia) (Shannon, 2000).
    2) An acute single dose of 200 mg or greater is considered an overdose of MDMA. Clinically, this resembles an acute methamphetamine overdose with the following symptoms possible: agitation, delirium, paranoia, tachycardia, hypertension, hyperthermia, marked diaphoresis, vomiting, diarrhea, abdominal pain, seizures, hypoglycemia, cerebral hemorrhage, rhabdomyolysis, acute renal failure, and DIC. Two common complications of MDMA overdose are hepatotoxicity and hyponatremia (Shannon, 2000).
    B) SPECIFIC SUBSTANCE
    1) MDMA
    a) CASE REPORTS
    1) ADULT
    a) Ingestion of a presumed "therapeutic" dose of 60 to 65 mg by an adult resulted in a severe, nearly fatal reaction (Hayner & McKinney, 1986). A similar severe reaction was noted in a 32-year-old woman who ingested 200 mg (Brown et al, 1986).
    b) A 30-year-old man found at home survived a massive overdose of ecstasy (50 tablets), along with oxazepam 10 mg, and alcohol 5 units (Ramcharan et al, 1998).
    c) A 23-year-old man presented with tachycardia, hypotension, tachypnea, a Glasgow Coma Scale (GCS) of 12/15 and fever (rectal temperature of 108.9 degrees F; 42.7 degrees C) after ingesting 3 MDMA pills at a concert. He was also combative, tremulous, hypertonic and hyper-reflexic. Despite supportive care, he developed severe rhabdomyolysis, multiorgan failure, and mild disseminated intravascular coagulopathy on day 2. Following further aggressive supportive care, including several sessions of hemodialysis (a total of 7 sessions), he had a full recovery within 3 weeks (Davies et al, 2014).
    2) PEDIATRIC
    a) INFANT: A 13-month-old child developed hypertonia, muscular hyperactivity, seizures, hypertension, tachycardia, and hyperthermia after ingesting a single capsule of MDMA (Russell et al, 1992).
    b) CASE REPORT: Approximately 40 minutes after ingesting a portion of an ecstasy pill, a 14-month-old child developed a generalized convulsion with opisthotonos, hyperthermia (38 degrees C), hypertension, tachycardia (130 beats/min), ventricular extrasystoles, tachypnea (50 breaths/min), and mydriasis. The urine levels of amphetamine/methamphetamine were greater than 16 mg/L 5 hours after ingestion. The serum level of MDMA was 0.591 mg/L 8 hours after ingestion. He continued to have hypertension, tachycardia and long periods of trigeminy, without hemodynamic repercussion during the first 12 hours. Following supportive care, the child recovered with no further sequelae (Melian et al, 2004).
    b) CASE SERIES
    1) In a retrospective review of a poison center database, 191 cases of MDMA exposure (defined as either the primary or secondary drug used) were reported during the years 1993 to 1999. Of the cases reported, 123 were males and 66 females (2 patients' gender was unknown) with a median age of 22 years (range: 18 to 25 years). Fifty-two cases (27%) experienced moderate to major toxicity (included 1 death from hyperthermia), and minor or no toxicity were reported in 139 cases (73%). The most common effects were tachycardia (22%), agitation (19%), and nausea and vomiting (12%), with the most serious complications being hyperthermia and hyponatremia. Of note, as of 1997, GHB (gamma-hydroxybutyrate) and ketamine were common coingestants (Rella et al, 2000).
    2) 2CB
    a) CASE REPORT: A 27-year-old healthy man developed auditory hallucinations and paranoid delusions (hearing voices and insistent that someone was following him) 2 days after intentionally ingesting a 2C-B (4-bromo-2, 5-dimethoxy-beta-phenethylamine) tablet for recreational use. He was admitted with a self-inflicted head injury after severely banging his head against a wall. Treatment included antipsychotics, but hallucinations persisted for approximately 5 days when the patient reported improvement with less anxiety (Huang & Bai, 2011).
    b) CASE REPORT: A 43-year-old woman with seronegative polyarticular arthritis, obesity and hypothyroidism developed severe, pulsatile headaches with confusion 48 hours after ingesting the liquid form of 2C-B. She had progressive upper extremity weakness and was admitted to the hospital with profound quadriparesis with diffuse hyperreflexia and encephalopathy 3 weeks later. An MRI of the brain revealed diffusion-positive lesions with bilateral ischemia and a magnetic resonance angiography showed diffuse arterial irregularities. A cerebral angiography confirmed focal vascular narrowing in small, medium, and large caliber vessels and the cortical vessels showed no evidence of vasculitis. At 6 months, the patient had minimal clinical improvement (Ambrose et al, 2010).

Serum Plasma Blood Concentrations

    7.5.1) THERAPEUTIC CONCENTRATIONS
    A) THERAPEUTIC CONCENTRATION LEVELS
    1) MDMA
    a) After ingestion of 50 mg MDMA by an adult, a peak plasma MDMA level of 0.106 mg/L was measured 2 hours postingestion. Peak level of methylenedioxyamphetamine, the major metabolite, was 0.028 mg/L at 4 hours postingestion (Verebey et al, 1988).
    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) SPECIFIC SUBSTANCE
    a) MDMA
    1) A 19-year-old man developed coma, seizures, hyperthermia, rhabdomyolysis, and hyperkalemia after ingesting ethanol and 3 ecstasy tablets (total dose unknown). Serum toxicology results revealed an elevated MDMA concentration of 1.5 mg/L. Despite supportive care, he had a cardiac arrest. Resuscitation efforts were unsuccessful and he died 2 hours later (Vanden Eede et al, 2012).
    2) In two cases of severe reactions, including hyperthermia, hypotension, coma, and pulmonary edema, serum MDMA levels were 6.5 to 7 mcg/mL (Brown et al, 1986; Hayner & McKinney, 1986).
    3) Death occurred in a 16-year-old girl whose blood MDMA level was 0.424 mg/L (Chadwick et al, 1991).
    4) A 30-year-old man survived a massive overdose at home of Ecstasy (50 tablets), oxazepam 10 mg, and alcohol 5 units. The patient's serum MDMA level 60 hours after ingestion was 0.8 mg/L (Ramcharan et al, 1998).
    5) At necropsy, blood concentrations of MDMA were 4.56 mg/L and 0.36 mg/L for 3,4-methylenedioxyamphetamine with an ethanol level of 0.24 g/L in an adult following ingestion of 2.5 tablets of Ecstasy (total dose approximately 180 mg MDMA) and alcohol at a party. The authors speculated that ritonavir (recently added to the patient's drug regimen) increased serum concentrations of MDMA (Henry & Hill, 1998).
    6) Postmortem serum concentrations were 3.05 mg/L in a 53-year-old man 5 days after ingesting an unknown quantity of MDMA (Walubo & Seger, 1999).
    7) A postmortem plasma concentration of 3.65 mcg/mL was reported in a 19-year-old man following ingestion of 12 MDMA tablets (Ravina et al, 2004).
    8) At more than 48 hours postingestion, postmortem serum concentration was 0.1 mg/L in a 29-year-old man after taking 1 Ecstasy tablet and a large amount of alcohol. At the time of autopsy, no alcohol or other agents were detected in the blood (Duflou & Mark, 2000).
    9) The tissue distribution in a fatal acute overdose involving MDMA were as follows (Rohrig & Prouty, 1992)
    TISSUEMILLIGRAMS/LITER (MDMA)
    Heart Blood10.9
    Femoral Blood2.8
    Liver20.2
    Brain13.7

    10) Postmortem serum MDMA concentrations of an 18-year-old man and a 31-year-old man, obtained from peripheral blood samples, were 0.304 mcg/mL and 26.059 mcg/mL (via subclavian blood), respectively, and 13.508 mcg/mL (from the 31-year-old man via femoral blood). In contrast, the serum MDMA concentrations found in cardiac blood and various organs (lung lobes, liver, and stomach) were much higher, with right atrial blood concentrations of 0.416 mcg/mL and 57.297 mcg/ml, respectively, right lung upper lobe concentrations of 3.031 mcg/g and 79.225 mcg/g, respectively, liver concentrations of 4.867 mcg/g and 103.497 mcg/g, respectively, and the stomach contents concentration of 10.519 mcg/g and 2310.709 mcg/g, respectively.
    a) The difference in MDMA concentrations between peripheral blood sampling and blood sampling centrally indicate that postmortem redistribution of MDMA into cardiac blood may occur and; therefore, may be misleading in interpreting potentially toxic or lethal MDMA levels. It is suggested that postmortem peripheral blood sampling, for accurate determination of MDMA concentrations, should remain as the recommended procedure and blood sampling should not be obtained centrally in the body (De Letter et al, 2004).
    11) A young man died of disseminated intravascular coagulation, induced by hyperthermia following the combined ingestion of amphetamines (3,4-methylenedioxymethylamphetamine, 3,4-methylenedioxyamphetamine, and paramethoxyamphetamine). Postmortem serum PMA, MDMA, MDA, AMP levels were as follows (Dams et al, 2003)
    SAMPLEPMAMDMAMDAAMP
    Femoral blood (mcg/L)16341129436198
    Vena Iliaca blood (mcg/L)16181421493203
    Vitreous humor (mcg/L)21011633577292
    Urine (mcg/L)932791369522
    Bile (mcg/L)50,01225,42011,6559425
    Liver (mcg/kg)89046657744857
    Spleen (mcg/kg)439030501454666
    Stomach content (mcg/L)73,10333,16814,3085478

    12) The authors proposed that blood sampled from femoral vein should be considered as the "gold standard" for blood sampling; however, if this sample is not available, iliac vein blood can also be used (Dams et al, 2003).
    13) In 5 Ecstasy ingestion cases that all resulted in death, antemortem and postmortem MDMA and MDA blood samples were obtained. The mean antemortem concentrations for MDMA and MDA were 1.84 mg/L and 0.06 mg/L, respectively. Results from postmortem sampling were overall higher compared to antemortem values, and they also showed wide variation in concentration values dependent on anatomical site. Mean postmortem concentrations for MDMA and MDA from peripheral sites were 2.9 mg/L and 0.13 mg/L, respectively. The highest postmortem concentrations were measured in postmortem heart blood. The author concluded that MDMA and MDA postmortem concentrations are not directly comparable with antemortem findings, but it is clear that MDMA and MDA exhibit postmortem redistribution (Elliott, 2005).
    b) 2C-T-7
    1) A 20-year-old man died after insufflating 35 mg of 2C-T-7. Postmortem values were: heart blood 57 ng/mL; femoral blood 100 ng/mL; urine 1120 ng/mL; and liver 854 ng/g (Curtis et al, 2003).
    c) BROMO-DRAGONFLY
    1) Postmortem concentrations of Bromo-Dragonfly following ingestion of approximately 700 mcg in an 18-year-old girl were as follows: femoral blood 4.7 +/- 0.7 mcg/kg and urine 22 +/- 2 mcg/kg (Andreasen et al, 2009).
    d) DOB
    1) CASE SERIES: After ingesting a "hallucinogen LSD-like" drug, 2 men developed a rapid onset of hallucinations (within 15 minutes), vomiting, and coma. The first subject, a 28-year-old (body mass 113 kg) survived, but developed serious convulsions. The second subject, a 29-year-old man (body mass 65 kg) died 6 days postingestion after developing generalized convulsions and metabolic acidosis (pH 6.6). Although immunoassay for amphetamines was negative in urine specimens, GC-MS showed 2,5-dimethoxy-4-bromoamphetamines (DOB) in the gastric and urine samples of both persons. DOB serum concentrations were 13 ng/mL for the surviving patient and 19 ng/mL for the deceased patient (Balikova, 2005).
    e) MDA
    1) The ingestion of 5 tablets of MDA, in a 20-year-old man, resulted in hyperpyrexia, muscle rigidity, dilated pupils, opisthotonic posturing, profuse sweating, and tonic-clonic seizures. Serum and urine MDA levels were 1.51 and 48.6 mg/L, respectively (Woods & Henry, 1992).
    2) In 5 Ecstasy ingestion cases that all resulted in death, antemortem and postmortem MDMA and MDA blood samples were obtained. The mean antemortem concentrations for MDMA and MDA were 1.84 mg/L and 0.06 mg/L, respectively. Results from postmortem sampling were overall higher compared to antemortem values, and they also showed wide variation in concentration values dependent on anatomical site. Mean postmortem concentrations for MDMA and MDA from peripheral sites were 2.9 mg/L and 0.13 mg/L, respectively. The highest postmortem concentrations were measured in postmortem heart blood. The author concluded that MDMA and MDA postmortem concentrations are not directly comparable with antemortem findings, but it is clear that MDMA and MDA exhibit postmortem redistribution (Elliott, 2005).
    3) MDA and 2C-I: A woman developed a massive intraventricular hemorrhage and permanent quadriplegia after ingesting MDA and 2C-I. Liquid-chromatography tandem mass spectrometry method was able to detect and quantify both MDA (5.56 mg/L) and 2CI (0.311 mg/L) in urine. The exact toxic dose is unknown for 2C-I. However, the MDA concentration was within range of urine concentrations (2 to 175 mg/L; average 108 mg/L MDA) in 12 fatalities (Drees et al, 2009).
    f) PMA
    1) Caldicott et al (2003) at the time of article publication identified 32 cases of PMA-related deaths and blood concentrations ranged from 0.2 mg/L to 5.7 mg/L (Caldicott et al, 2003).
    2) A postmortem serum concentration of 5.0 mcmol/L was reported in a 16-year-old boy who ingested 7 tablets of PMA (Refstad, 2003).
    3) Postmortem blood concentrations of PMA and PMMA in a 20-year-old man, following ingestion of an unknown amount, were 3.4 mg/kg and 3.3 mg/kg, respectively. In a 24-year-old man, the postmortem blood concentrations of PMA and PMMA, following ingestion of an unknown amount, were 0.78 mg/kg and 0.68 mg/kg, respectively (Johansen et al, 2003).
    g) PMMA
    1) The postmortem blood concentrations of PMA, PMMA, and other drugs were determined in 8 fatalities (age range: 14 to 25 years) associated with PMMA use. PMMA was found to be higher in postmortem blood and urine compared to PMA. The mean PMA and PMMA postmortem blood concentrations were 0.213 (+/- 0.144) and 4.312 (+/- 4.806) mcg/mL, respectively. Postmortem urine samples also contained high amounts of PMA and PMMA with the PMA-to-PMMA concentration ratios ranging from 0.019 to 0.237. Other agents detected included MDA, MDMA, ketamine, norketamine, hydroxymidazolam, methamphetamine, and pentobarbital (Lin et al, 2007).
    h) U4EuH
    1) Blood and urine levels in a fatal reported case were 21.3 and 12.3 mg/L, respectively. Autopsy reports showed pulmonary and cerebral edema (Davis & Brewster, 1988).

Toxicologic Mechanism

    A) The hallucinogenic methoxylated and/or methylenedioxy derivatives of amphetamines have properties in common with amphetamine and mescaline (3,4,5-trimethoxyphenylethylamine).
    1) Their ability to stimulate both the sympathetic and central nervous systems ultimately results from their structural similarity to the endogenous catecholamines, epinephrine, norepinephrine, and dopamine.
    2) Methoxylation and methylenedioxylation of the catechol ring is responsible for their hallucinogenic activity.
    B) Although their exact mechanism of action remains uncertain, proposed mechanisms include:
    1) Sympathetic receptor stimulation (false transmitter)
    2) Indirect receptor stimulation by causing the release of endogenous neurotransmitters
    3) Inhibition of monoamine oxidase (responsible for catecholamine degradation)
    4) Inhibition of catecholamine reuptake by presynaptic neurons (primary mechanism for terminating the effects of neurotransmitters)
    5) Biotransformation of these agents to indolamines related to serotonin (5-hydroxytryptamine)
    1) (Fellows & Bernheim, 1950; Schmidt et al, 1986).
    C) RATS
    1) MDA, MDMA, and MDEA produce toxicity to serotonergic neurons in the rat brain.
    a) 5-hydroxytryptamine levels are markedly reduced following acute administration of 10 mg/kg (Stone et al, 1986; Commins et al, 1987; Schmidt, 1987a).
    b) MDMA was neurotoxic in rats given parenteral doses approximately 4 times the human oral dose (Schmidt et al, 1986).
    c) A biphasic effect on serotonergic neurons has been observed with MDMA. An initial reversible depletion of serotonin was followed later by a partially reversible neurotoxic effect on the nerve terminal.
    d) The doses in this study were 4 to 8 times the human oral dose (Schmidt, 1987).
    2) The ability of hallucinogenic amphetamines to release dopamine from rat striatal slices decreased as the size of the N-alkyl substituent increased. Thus the dopamine-releasing potency was greatest with MDA, less with MDMA, and least with MDEA. Thus amphetamine-like side effects may be less with the latter 2 agents (Schmidt, 1987a).
    3) HEPATOTOXICITY: The metabolism of 3,4-methylenedioxymethamphetamine (MDMA) and 3,4-methylenedioxyamphetamine (MDA) can produce highly reactive hepatotoxic compounds: N-methyl-alpha-methyldopamine (N-Me-alpha-MeDA), alpha-methyldopamine (alpha-MeDA), and alpha-MeDA. One rat study proposed that MDA and alpha-MeDA may induce toxicity by disrupting thiol homeostasis due to conjugation of glutathione with alpha-MeDA, resulting in loss of protein function, decreases in the antioxidant enzyme activities, and cell death (Carvalho et al, 2004).
    D) PRIMATES
    1) Single doses of MDMA have also been shown to produce long-lasting serotonin depletion (30% of neurons for up to 2 weeks) when administered orally to monkeys at near usual human doses (5 mg/kg) (usual doses in humans are 1 to 4 mg/kg) (Ricaurte et al, 1988).
    2) Repeated administration of subcutaneous doses of MDMA twice daily to monkeys for 4 days resulted in a dose-dependent depletion of serotonin in the somatosensory cortex.
    a) A dose of 2.5 and 5 mg/kg produced 44% and 90% depletion, respectively. Serotonin was also depleted in the cerebral cortex, with a 44% depletion at 2.5 mg/kg. Neither dopamine nor norepinephrine were depleted. Axonal damage to serotonin nerve fibers in the cerebral cortex was demonstrated. Cell body damage was also shown (Ricaurte et al, 1988a).
    E) PMA
    1) Paramethoxyamphetamine (PMA) is a methoxylated phenethylamine derivative. It is structurally related to MDA, MDMA, MDEA, and mescaline and has been sold under the name "Ecstasy" in Australia. It does have hallucinogenic properties. Animal studies have demonstrated that PMA exerts it's effects primarily through serotonin release and reuptake and an ability to selectively inhibit serotonin metabolism. PMA can increase extracellular levels of serotonin more than MDMA, although the toxicity profile of PMA (agitation, delirium, hyperthermia) appears similar (Felgate et al, 1998; Caldicott et al, 2003).

Physical Characteristics

    A) MDMA: Powder
    B) "Ecstasy": Crystals

Molecular Weight

    A) MDMA: 193.25

Clinical Effects

    11.1.2) BOVINE/CATTLE
    A) Poisoning is uncommon but not rare. Signs in small animals include hyperthermia, hyperventilation, tachycardia, mydriasis, lactic acidosis, hypoglycemia, hypertension, arrythmias, vomiting, diarrhea, and dysuria. Renal failure may occur secondary to rhabdomyolysis. Seizures and coma may occur (Beasley et al, 1989).

Treatment

    11.2.1) SUMMARY
    A) GENERAL TREATMENT
    1) Begin treatment immediately.
    2) Keep animal warm and do not handle unnecessarily.
    3) Sample vomitus, blood, urine, and feces for analysis.
    4) ANIMAL POISON CONTROL CENTERS
    a) ASPCA Animal Poison Control Center, An Allied Agency of the University of Illinois, 1717 S. Philo Rd, Suite 36, Urbana, IL 61802, website www.aspca.org/apcc
    b) It is an emergency telephone service which provides toxicology information to veterinarians, animal owners, universities, extension personnel and poison center staff for a fee. A veterinary toxicologist is available for consultation.
    c) The following 24-hour phone number is available: (888) 426-4435. A fee may apply. Please inquire with the poison center. The agency will make follow-up calls as needed in critical cases at no extra charge.
    5) Due to lack of reports of large animal intoxication with this substance, the following sections address small animals (dogs and cats) only. In the case of a poisoning involving large animals, consult a veterinary poison control center.
    11.2.2) LIFE SUPPORT
    A) GENERAL
    1) MAINTAIN VITAL FUNCTIONS: Secure airway, supply oxygen, and begin supportive fluid therapy if necessary.
    11.2.4) DECONTAMINATION
    A) GASTRIC DECONTAMINATION
    1) GENERAL TREATMENT
    a) EMESIS/GASTRIC LAVAGE - Emesis may be initiated only if in the early period after ingestion, or if the animal is not overly stimulated. Lavage is preferred if the animal is stimulated (administer appropriate sedation) or if emesis is ineffective.
    1) If within 2 hours of exposure, induce emesis with 1 to 2 milliliters/kilogram syrup of ipecac per os. Dogs may vomit more readily with 1 tablet (6 milligrams) apomorphine diluted in 3 to 5 milliliters water and instilled into the conjunctival sac or per os.
    2) Dogs may also be given apomorphine intravenously at 40 micrograms/kilogram. Do not use an emetic if the animal is hypoxic. In the absence of a gag reflex or if vomiting cannot be induced, place a cuffed endotracheal tube and begin gastric lavage.
    3) Pass large bore stomach tube and instill 5 to 10 milliliters/kilogram water or lavage solution, then aspirate. Repeat 10 times (Kirk, 1986).
    b) ACTIVATED CHARCOAL - Administer activated charcoal, 2 grams/kilogram per os or via stomach tube. Avoid aspiration by proper restraint, careful technique, and if necessary tracheal intubation.
    c) CATHARTIC - Administer a dose of a saline cathartic such as magnesium or sodium sulfate (sodium sulfate dose is 1 gram/kilogram). If access to these agents is limited, give 5 to 15 milliliters magnesium oxide (Milk of Magnesia) per os for dilution.
    11.2.5) TREATMENT
    A) GENERAL TREATMENT
    1) MAINTAIN VITAL FUNCTIONS - as necessary.
    B) DOG
    1) DIAZEPAM - 0.5 milligram/kilogram intravenous bolus; may repeat dose as needed for CNS effects. Give slowly over 1 to 2 minutes.
    C) CAT
    1) PHENOBARBITAL may be used to control CNS signs. Dose: 5 to 30 milligrams/kilogram over 5 to 10 minutes intravenously.
    D) OTHER
    1) PHENOBARBITAL may be used to control CNS signs. Dose: 5 to 30 milligrams/kilogram over 5 to 10 minutes intravenously.
    E) REFRACTORY SEIZURES - Consider anaesthesia or heavy sedation. Administer pentobarbital to DOGS & CATS at a dose of 3 to 15 milligrams/kilogram intravenously slowly to effect. May need to repeat in 4 to 8 hours. Be sure to protect the airway.
    F) HYPERTHERMIA - Monitor body temperature every 2 to 4 hours. Treat hyperthermia by employing whole-body cooling, including alcohol on the foot-pads, packing the animal in ice, and training fans on the animal.
    G) URINARY ACIDIFICATION may speed the excretion of amphetamine. This is CONTRAINDICATED if the animal is in renal failure, or if myoglobinuria or severe electrolyte or blood pH imbalances are present. Urine pH goal: 4.5 to 5.5 in dogs and cats. Ammonium chloride or ascorbic acid may be used to acidify urine (Beasley et al, 1989).

Range Of Toxicity

    11.3.2) MINIMAL TOXIC DOSE
    A) SPECIFIC TOXIN
    1) LD50 is estimated at 10 to 30 mg/kg (Beasley et al, 1989).

Continuing Care

    11.4.1) SUMMARY
    11.4.1.2) DECONTAMINATION/TREATMENT
    A) GENERAL TREATMENT
    1) Begin treatment immediately.
    2) Keep animal warm and do not handle unnecessarily.
    3) Sample vomitus, blood, urine, and feces for analysis.
    4) ANIMAL POISON CONTROL CENTERS
    a) ASPCA Animal Poison Control Center, An Allied Agency of the University of Illinois, 1717 S. Philo Rd, Suite 36, Urbana, IL 61802, website www.aspca.org/apcc
    b) It is an emergency telephone service which provides toxicology information to veterinarians, animal owners, universities, extension personnel and poison center staff for a fee. A veterinary toxicologist is available for consultation.
    c) The following 24-hour phone number is available: (888) 426-4435. A fee may apply. Please inquire with the poison center. The agency will make follow-up calls as needed in critical cases at no extra charge.
    5) Due to lack of reports of large animal intoxication with this substance, the following sections address small animals (dogs and cats) only. In the case of a poisoning involving large animals, consult a veterinary poison control center.
    11.4.2) DECONTAMINATION
    11.4.2.2) GASTRIC DECONTAMINATION
    A) GASTRIC DECONTAMINATION
    1) GENERAL TREATMENT
    a) EMESIS/GASTRIC LAVAGE - Emesis may be initiated only if in the early period after ingestion, or if the animal is not overly stimulated. Lavage is preferred if the animal is stimulated (administer appropriate sedation) or if emesis is ineffective.
    1) If within 2 hours of exposure, induce emesis with 1 to 2 milliliters/kilogram syrup of ipecac per os. Dogs may vomit more readily with 1 tablet (6 milligrams) apomorphine diluted in 3 to 5 milliliters water and instilled into the conjunctival sac or per os.
    2) Dogs may also be given apomorphine intravenously at 40 micrograms/kilogram. Do not use an emetic if the animal is hypoxic. In the absence of a gag reflex or if vomiting cannot be induced, place a cuffed endotracheal tube and begin gastric lavage.
    3) Pass large bore stomach tube and instill 5 to 10 milliliters/kilogram water or lavage solution, then aspirate. Repeat 10 times (Kirk, 1986).
    b) ACTIVATED CHARCOAL - Administer activated charcoal, 2 grams/kilogram per os or via stomach tube. Avoid aspiration by proper restraint, careful technique, and if necessary tracheal intubation.
    c) CATHARTIC - Administer a dose of a saline cathartic such as magnesium or sodium sulfate (sodium sulfate dose is 1 gram/kilogram). If access to these agents is limited, give 5 to 15 milliliters magnesium oxide (Milk of Magnesia) per os for dilution.

Kinetics

    11.5.1) ABSORPTION
    A) SPECIFIC TOXIN
    1) Amphetamines are rapidly absorbed from the GI tract (Beasley et al, 1989).
    11.5.2) DISTRIBUTION
    A) SPECIFIC TOXIN
    1) Amphetamine also enters the CSF; CSF concentration is about 80% that of plasma (Beasley et al, 1989).
    11.5.4) ELIMINATION
    A) SPECIFIC TOXIN
    1) Elimination is via the kidney and is dependent on pH. Acidifying the urine will speed elimination (Beasley et al, 1989).

Pharmacology Toxicology

    A) SPECIFIC TOXIN
    1) Amphetamine stimulates catecholamine release centrally and from the adrenal glands; and stimulates cortical centers including the cerebral cortex, reticular activating system, and the medullary respiratory center.
    2) Amphetamine is also sympathomimetic and has a greater stimulant activity at sympathetic nerve endings than norepinephrine. It is a monoamine oxidase inhibitor (Beasley et al, 1989).

Other

    A) OTHER
    1) GENERAL
    a) DIFFERENTIAL DIAGNOSIS - Toxicoses: strychnine, organochlorine insecticides, methylxanthines, 4-aminopyridine, metaldehyde, and other illicit drugs. The animal's owner may hinder diagnosis by not admitting possession of illicit drugs (Beasley et al, 1989).

General Bibliography

    1) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    2) Ajaelo I, Koenig K, & Snoey E: Severe hyponatremia and inappropriate antidiuretic hormone secretion following ecstasy use (letter). Acad Emerg Med 1998; 5:839-840.
    3) Alansari M & Hamilton D: Nephrotoxicity of BZP-based herbal party pills: a New Zealand case report. N Z Med J 2006; 119(1233):U1959.
    4) Albertson TE, Walby WG, & Derlet RW: Stimulant-induced pulmonary toxicity. Chest 1995; 108:1140-49.
    5) Ambrose JB, Bennett HD, Lee HS, et al: Cerebral vasculopathy after 4-bromo-2,5-dimethoxyphenethylamine ingestion. Neurologist 2010; 16(3):199-202.
    6) American Heart Association: 2005 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2005; 112(24 Suppl):IV 1-203. Available from URL: http://circ.ahajournals.org/content/vol112/24_suppl/. As accessed 12/14/2005.
    7) Ames D & Wirshing WC: Ecstasy, the serotonin syndrome, and neuroleptic malignant syndrome - a possible link? (Letter). JAMA 1993; 269:869-870.
    8) Andreasen MF, Telving R, Birkler RI, et al: A fatal poisoning involving Bromo-Dragonfly. Forensic Sci Int 2009; 183(1-3):91-96.
    9) Anon: Long-term MDMA use may impair memory. Neurology 2001; 56:966-969.
    10) Appleton R: Lorazepam vs diazepam in the acute treatment of epileptic seizures and status epilepticus.. Dev Med Child Neuro 1995; 37:682-688.
    11) Austin H & Monasterio E: Acute psychosis following ingestion of "rapture". Australasian Psychiatry 2004; 12:406-408.
    12) Balikova M: Nonfatal and fatal DOB (2,5-dimethoxy-4-bromamphetamine) overdose. Forens Sci Int 2005; 153:85-91.
    13) Barrett PJ & Taylor GT: Ecstasy ingestion: a case report of severe complications. J Roy Soc Med 1993; 86:233-234.
    14) Barrett PJ: 'Ecstasy' misuse--overdose or normal dose (letter)?. Anaesthesia 1993; 48:83.
    15) Barrett PJ: Ecstasy and dantrolene (letter). Br Med J 1992; 305:1225.
    16) Baselt RC & Cravey RH: Disposition of Toxic Drugs and Chemicals in Man, 3rd ed. Year Book Medical Publishers, Chicago, IL, 1989.
    17) Battaglia G, Yeh SY, & De Souza EB: MDMA-induced neurotoxicity: parameters of degeneration and recovery of brain serotonin neurons. Pharmacol Biochem Behav 1988; 29:269-274.
    18) Beasley VR, Dorman DC, & Fikes JD: A Systems Affected Approach to Veterinary Toxicology, University of Illinois, Urbana, IL, 1989.
    19) Becker J, Neis P, Rohrich J, et al: A fatal paramethoxymethamphetamine intoxication. Leg.Med (Tokyo) 2003; 5(Suppl 1):S138-S141.
    20) Ben-Abraham R, Szold O, Rudick V, et al: 'Ecstasy' intoxication: life-threatening manifestations and resuscitative measures in the intensive care setting. Eur J Emerg Med 2003; 10:309-313.
    21) Benazzi F & Mazzoli M: Psychiatric illness associated with "ecstasy". Lancet 1991; 338:1520.
    22) Boja JW & Schechter MD: Behavioral effects of n-ethyl-3,4-methylenedioxyamphetamine (MDE; "EVE"). Pharmacol Biochem Behav 1987; 28:153-156.
    23) Bost RO: 3,4-methylenedioxymethamphetamine (MDMA) and other amphetamine derivatives. J Forensic Sci 1988; 33:576-587.
    24) Bowen JS, Davis GB, & Kearney TE: Diffuse vascular spasm associated with 4-bromo-2,5-dimethoxyamphetamine ingestion. JAMA 1983; 249:1477-1479.
    25) Boyer EW & Shannon M: The serotonin syndrome. N Eng J Med 2005; 352(11):1112-1120.
    26) Brncic N, Kraus I, Viskovic I, et al: 3,4-methylenedioxymethamphetamine (MDMA): an important cause of acute hepatitis. Med Sci Monit 2006; 12(11):CS107-CS109.
    27) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    28) Brown C & Osterloh J: Multiple severe complications from recreational ingestion of MDMA ('Ecstasy') (letter). JAMA 1987; 258:780-781.
    29) Brown CR, McKinney H, & Osterloh JD: Severe adverse reaction to 3,4-methylenedioxymethamphetamine (MDMA) (abstract). Vet Hum Toxicol 1986; 28:490.
    30) Brown CV, Rhee P, Chan L, et al: Preventing renal failure in patients with rhabdomyolysis: do bicarbonate and mannitol make a difference?. J Trauma 2004; 56(6):1191-1196.
    31) Bryden AA, Rothwell PJN, & O'Reilly PH: Urinary retention with misuse of "ecstasy" (letter). Br Med J 1995; 310:504.
    32) Buchanan JF & Brown CR: "Designer drugs": a problem in clinical toxicology. Med Toxicol 1988; 3:1-17.
    33) Buhrich N, Morris G, & Cook G: Bromo-DMA: the Australiasian hallucinogen. Aust NZ J Psychiatr 1983; 17:275-279.
    34) Bunchman TE, Lynch RE, & Wood EG: Intravenously administered labetalol for treatment of hypertension in children. J Pediatr 1992; 120(1):140-144.
    35) Byard RW, James RA, & Gilbert JD: Another PMA-related fatality in Adelaide (letter). Med J Austral 1999; 170:139-140.
    36) Caldicott DG, Edwards NA, Kruys A, et al: Dancing with "death": p-methoxyamphetamine overdose and its acute management. J Toxicol Clin Toxicol 2003; 41(2):143-154.
    37) Camp NE: Drug- and toxin-induced Rhabdomyolysis. J Emerg Nurs 2009; 35(5):481-482.
    38) Campkin NTA & Davies UM: Another death from ecstasy. J R Soc Med 1992; 85:61.
    39) Campkin NTA & Davies UM: Treatment of 'ecstasy' overdose with dantrolene (letter). Anaesthesia 1993; 48:82-83.
    40) Carvalho M, Milhazes N, Remiao F, et al: Hepatotoxicity of 3,4-methylenedioxyamphetamine and alpha-methyldopamine in isolated rat hepatocytes: formation of glutathione conjugates. Arch Toxicol 2004; 78:16-24.
    41) Chadwick IS, Linsley A, & Freemont AJ: Ecstasy, 3-4 methylenedioxymethamphetamine (MDMA), a fatality associated with coagulopathy and hyperthermia. J Royal Soc Med 1991; 84:371.
    42) Chamberlain JM, Altieri MA, & Futterman C: A prospective, randomized study comparing intramuscular midazolam with intravenous diazepam for the treatment of seizures in children. Ped Emerg Care 1997; 13:92-94.
    43) Chang S, Lai T, Chen W, et al: MDMA-induced acute pulmonary edema in a patient without other organ dysfunction. Am J Emerg Med 2006; 24(6):734-736.
    44) Chang Y, Lai M, Kong M, et al: Accidental ingestion of Ecstasy in a toddler. J Formos Med Assoc 2005; 104(12):946-947.
    45) Chin RF , Neville BG , Peckham C , et al: Treatment of community-onset, childhood convulsive status epilepticus: a prospective, population-based study. Lancet Neurol 2008; 7(8):696-703.
    46) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    47) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    48) Claassen JAHR & Gelissen HPMM: The serotonin syndrome (letter). N Eng J Med 2005; 352(23):2455.
    49) Climko RP, Roehrich H, & Sweeney DR: Ecstasy: a review of MDMA and MDA. Intl J Psychiatr Med 1986-7; 16:359-372.
    50) Cohen BM & Butler R: BZP-party pills: a review of research on benzylpiperazine as a recreational drug. Int J Drug Policy 2011; 22(2):95-101.
    51) Colak Y, Tuncer I, Enc FY, et al: Ecstasy induced fatal hepatic failure. J Gastrointestin Liver Dis 2011; 20(2):215-216.
    52) Commins DL, Vosmer G, & Virus RM: Biochemical and histological evidence that methylenedioxymethylamphetamine (MDMA) is toxic to neurons in the rat brain. J Pharmacol Exp Ther 1987; 241:338-345.
    53) Creighton FJ, Black DL, & Hyde CE: Ecstasy psychosis and flashbacks. Br J Psychiatry 1991; 159:713-715.
    54) Criddle LM: Rhabdomyolysis. Pathophysiology, recognition, and management. Crit Care Nurse 2003; 23(6):14-22, 24-26, 28.
    55) Curtis B, Kemp P, Harty L, et al: Postmortem identification and quantitation of 2,5-dimethoxy-4-n-propylthiophenethylamine using GC-MSD and GC-NPD. J Analy Toxicol 2003; 27:493-498.
    56) Dams R, De Letter EA, Mortier KA, et al: Fatality due to combined use of the designer drugs MDMA and PMA: a distribution study. J Analyt Toxicol 2003; 27:318-322.
    57) Daumann J, Schnitker R, Weidemann J, et al: Neural correlates of working memory in pure and polyvalent ecstasy (MDMA) users. NeuroReport 2003; 14:1983-1987.
    58) Davies O, Batajoo-Shrestha B, Sosa-Popoteur J, et al: Full recovery after severe serotonin syndrome, severe rhabdomyolysis, multi-organ failure and disseminated intravascular coagulopathy from MDMA. Heart Lung 2014; 43(2):117-119.
    59) Davis FT & Brewster ME: A fatality involving U4Euh, a cyclic derivative of phenylpropanolamine. J Forensic Sci 1988; 33:549-553.
    60) Davis WM & Borne RF: Pharmacologic investigation of compounds related to 3,4 methylenedioxyamphetamine. Subst Alcohol Actions Misuse 1984; 5:105-110.
    61) Davis WM, Catravas JD, & Waters IW: Effects of an IV lethal dose of 3,4-methylenedioxyamphetamine (MDA) in the dog and antagonism by chlorpromazine. Gen Pharmacol 1986; 17:179-183.
    62) De Letter EA, Bouche MPLA, Van Bocxlaer JF, et al: Interpretation of a 3,4-methylenedioxymethamphetamine (MDMA) blood level: discussion by means of a distribution study in two fatalities. Forens Sci Intl 2004; 141:85-90.
    63) De Negri M & Baglietto MG: Treatment of status epilepticus in children. Paediatr Drugs 2001; 3(6):411-420.
    64) DeSilva DA, Wong MC, Lee MP, et al: Amphetamine-associated ischemic stroke: clinical presentation and proposed pathogenesis. J Stroke Cerebrovasc Dis 2007; 16(4):185-186.
    65) Deal JE , Barratt TM , & Dillon MJ : Management of hypertensive emergencies. Arch Dis Child 1992; 67(9):1089-1092.
    66) Denborough MA & Hopkinson KC: Dantrolene and "ecstasy" (letter). Med J Austral 1997; 166:165-166.
    67) Dinis-Oliveira RJ , Caldas I , Carvalho F , et al: Bruxism after 3,4-methylenedioxymethamphetamine (ecstasy) abuse. Clin Toxicol (Phila) 2010; 48(8):863-864.
    68) Dobry Y, Rice T, & Sher L: Ecstasy use and serotonin syndrome: a neglected danger to adolescents and young adults prescribed selective serotonin reuptake inhibitors. Int J Adolesc Med Health 2013; 25(3):193-199.
    69) Dowling GP, McDonough ET III, & Bost RO: "Eve" and "Ecstasy". A report of five deaths associated with the use of MDEA and MDMA. JAMA 1987; 257:1615-1617.
    70) Doyon S: The many faces of ecstasy. Current Opin Pediatr 2001; 13:170-176.
    71) Drees JC , Stone JA , & Wu AH : Morbidity involving the hallucinogenic designer amines MDA and 2C-I. J Forensic Sci 2009; 54(6):1485-1487.
    72) Duflou J & Mark A: Aortic dissection after ingestion of "ecstasy" (MDMA). Am J Forensic Med Pathol 2000; 21:261-263.
    73) Dumont GJ, Kramers C, Sweep FC, et al: Cannabis coadministration potentiates the effects of "ecstasy" on heart rate and temperature in humans. Clin Pharmacol Ther 2009; 86(2):160-166.
    74) Dumont GJ, Sweep FC, van der Steen R, et al: Increased oxytocin concentrations and prosocial feelings in humans after ecstasy (3,4-methylenedioxymethamphetamine) administration. Soc Neurosci 2009a; 4(4):359-366.
    75) Duxbury AJ: Ecstasy - dental implications. Br Dent J 1993; 175:410.
    76) Dykhuizen RS, Brunt PW, & Arkinson P: Ecstasy induced hepatitis mimicking viral hepatitis. Gut 1995; 36:939-941.
    77) Edwards G: Blasted with ennui. Br Med J 1989; 298:136.
    78) Eifinger F, Roth B, Kroner L, et al: Severe Ecstasy poisoning in an 8-month-old infant. Eur J Pediatr 2008; 167(9):1067-1070.
    79) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    80) Elliott S: Current awareness of piperazines: pharmacology and toxicology. Drug Test Anal 2011; 3(7-8):430-438.
    81) Elliott SP: MDMA and MDA concentrations in antemortem and postmortem specimens in fatalities following hospital admission. J Anal Toxicol 2005; 29(5):296-300.
    82) Engebretsen KM & Harris CR: Severe hyponatremia with resultant noncardiogenic pulmonary edema and cerebral edema secondary to ecstasy intoxication (abstract). J Toxicol-Clin Toxicol 2001; 39:290-291.
    83) Erdman AR & Dart RC: Rhabdomyolysis. In: Dart RC, Caravati EM, McGuigan MA, et al, eds. Medical Toxicology, 3rd ed. Lippincott Williams & Wilkins, Philadelphia, PA, 2004, pp 123-127.
    84) Escalante FA, Del Bano F, & Supervia A: MDMA-induced angioedema treated with icatibant. Clin Toxicol (Phila) 2015; 53(10):1148-1149.
    85) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    86) Fahal IH, Sallomi DF, & Yaqoob M: Acute renal failure after ecstasy (letter). Br Med J 1992; 305:29.
    87) Farah R & Farah R: Ecstasy (3,4-methylenedioxymethamphetamine)-induced inappropriate antidiuretic hormone secretion. Pediatr Emerg Care 2008; 24(9):615-617.
    88) Feldman KW & Mazor S: Ecstasy ingestion causing heatstroke-like, multiorgan injury in a toddler. Pediatr Emerg Care 2007; 23(10):725-726.
    89) Felgate HE, Felgate PD, & James RA: Recent paramethoxyamphetamine deaths. J Analyt Toxicol 1998; 22:169-172.
    90) Fellows EJ & Bernheim F: The effect of a number of aralkylamines on the oxidation of tyramine by amine oxidase. J Pharmacol Exp Ther 1950; 100:94-99.
    91) Ferigolo M , Machado AG , Oliveira NB , et al: Ecstasy intoxication: the toxicological basis for treatment. Rev Hosp Clin FacMed Sao Paulo 2003; 58(6):332-341.
    92) Fineschi V & Masti A: Fatal poisoning by MDMA (ecstasy) and MDEA: a case report. Int J Legal Med 1996; 108:272-275.
    93) Finnegan KT, Ricaurte GA, & Ritchie LD: Orally administered MDMA causes a long-term depletion of serotonin in rat brain. Brain Res 1988; 447:141-144.
    94) Fivush B , Neu A , & Furth S : Acute hypertensive crises in children: emergencies and urgencies. Curr Opin Pediatr 1997; 9(3):233-236.
    95) Flynn JT & Tullus K: Severe hypertension in children and adolescents: pathophysiology and treatment. Pediatr Nephrol 2009; 24(6):1101-1112.
    96) Foex BA, Subramanian G, & Butler JM: Ecstasy overdose and memory loss. Emerg Med J 2010; 27(10):733.
    97) Friedman R: Ecstasy, the serotonin syndrome, and neuroleptic malignant syndrome - a possible link? (Reply). JAMA 1993; 269:869-870.
    98) Garbino J, Henry JA, & Mentha G: Ecstasy ingestion and fulminant hepatic failure: liver transplantation to be considered as a last therapeutic option. Vet Human Toxicol 2001; 43:99-102.
    99) Garcia-Algar O, Lopez N, Bonet M, et al: 3,4-methylenedioxymethamphetamine (MDMA) intoxication in an infant chronically exposed to cocaine. Ther Drug Monit 2005; 27(4):409-411.
    100) Gardner H, Lawn N, Fatovich DM, et al: Acute hippocampal sclerosis following ecstasy ingestion. Neurology 2009; 73(7):567-569.
    101) Gee P, Gilbert M, Richardson S, et al: Toxicity from the recreational use of 1-benzylpiperazine. Clin Toxicol (Phila) 2008; 46(9):802-807.
    102) Gee P, Jackson S, & Easton J: Another bitter pill: a case of toxicity from DMAA party pills. N Z Med J 2010; 123(1327):124-127.
    103) Gee P, Richardson S, Woltersdorf W, et al: Toxic effects of BZP-based herbal party pills in humans: a prospective study in Christchurch, New Zealand. N Z Med J 2005; 118(1227):U1784-.
    104) Ghatol A & Kazory A: Ecstasy-associated acute severe hyponatremia and cerebral edema: a role for osmotic diuresis?. J Emerg Med 2012; 42(6):e137-e140.
    105) Giang DW & McBride MC : Lorazepam versus diazepam for the treatment of status epilepticus. Pediatr Neurol 1988; 4(6):358-361.
    106) Gillman PK: Ecstasy, serotonin syndrome and the treatment of hyperpyrexia (letter). MJA 1997; 167:109-111.
    107) Gillman PK: Successful treatment of serotonin syndrome with chlorpromazine (letter). MJA 1996; 165:345.
    108) Ginat DT: MRI of toxic leukoencephalopathy syndrome associated with methylenedioxymethamphetamine. Neurology 2015; 84(7):757.
    109) Ginsberg MD, Hertzman M, & Schmidt-Nowara WW: Amphetamine intoxication with coagulopathy, hyperthermia, and reversible renal failure, a syndrome resembling heatstroke. Ann Intern Med 1970; 73:81-85.
    110) Gledhill JA, Moore DF, & Bell D: Subarachnoid haemorrhage associated with MDMA abuse. J Neurol, Neurosurg & Psych 1993; 56:1036-1037.
    111) Gold LH, Koob GF, & Geyer MA: Stimulant and hallucinogenic behavioral profiles of 3,4-methylenedioxymethamphetamine and n-ethyl-3,4-methylenedioxyamphetamine in rats. J Pharmacol Exp Ther 1988; 247:547-555.
    112) Goldberg RJ & Huk M: Serotonin syndrome from trazodone and buspirone (letter). Psychosomatics 1992; 33:235-236.
    113) Goldstein LH, Mordish Y, Abu-Kishak I, et al: Acute paralysis following recreational MDMA (Ecstasy) use. Clin Toxicol (Phila) 2006; 44(3):339-341.
    114) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    115) Gorard DA, Davies SE, & Clark MI: Misuse of ecstasy (letter). Br Med J 1992; 305:309.
    116) Gosselin RE, Smith RP, & Hodge HC: Clinical Toxicology of Commercial Products, 5th ed, Williams & Wilkins, Baltimore, MD, 1984.
    117) Gouzoulis E, Borchardt D, & Hermle L: A case of toxic psychosis induced by EVE (3,4-methylene-dioxyethylam-phetamine) (letter). Arch Gen Psychiatry 1993; 50:75.
    118) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    119) Graham PM: Successful treatment of the toxic serotonin syndrome with chlorpromazine (letter). Med J Australia 1997; 166:166-167.
    120) Grob C, Bravo G, & Walsh R: Second thoughts on 3,4-methylenedioxymethamphetamine (MDMA) neurotoxicity. Arch Gen Psychiatry 1990; 47:288-299.
    121) Grunau BE, Wiens MO, & Brubacher JR: Dantrolene in the treatment of MDMA-related hyperpyrexia: a systematic review. CJEM 2010; 12(5):435-442.
    122) Hanyu S, Ikeguchi K, & Imai H: Cerebral infarction associated with 3,4-methylenedioxymethamphetamine ('Ecstasy') abuse. Eur Neurol 1995; 35:173.
    123) Hari P & Sinha A: Hypertensive emergencies in children. Indian J Pediatr 2011; 78(5):569-575.
    124) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    125) Hayner GN & McKinney H: MDMA the dark side of ecstasy. J Psychoactive Drugs 1986; 18:341-347.
    126) Hegadoren KM, Baker GB, & Coutts RT: Analysis of the enantiomers of 3,4- methylenedioxy-N-ethylamphetamine (MDE, "eve") and its metabolite 3,4- methylenedioxyamphetamine (MDA) in rat brain. J Pharm Toxicol Method 1995; 34:117-123.
    127) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    128) Henry JA & Hill IR: Fatal interaction between ritonavir and MDMA (letter). Lancet 1998; 352:1751-1752.
    129) Henry JA, Jeffreys KJ, & Dawling S: Toxicity and deaths from 3,4-methylenedioxymethamphetamine ("ecstasy"). Lancet 1992; 340:384-387.
    130) Hill SL & Thomas SH: Clinical toxicology of newer recreational drugs. Clin Toxicol (Phila) 2011; 49(8):705-719.
    131) Hiramatsu M, Kumagai Y, & Unger SE: Metabolism of methylenedioxymethamphetamine: formation of dihydroxymethamphetamine and a quinone identified as its glutathione adduct. J Pharmacol Exp Ther 1990; 254:521-527.
    132) Hoggett K, McCoubrie D, & Fatovich DM: Ecstasy-induced acute coronary syndrome: something to rave about. Emerg Med Australas 2012; 24(3):339-342.
    133) Holden R & Jackson MA: Near-fatal hyponatraemic coma due to vasopressin over-secretion after "ecstasy" (3,4-MDMA). Lancet 1996; 347:1052.
    134) Homsi E, Barreiro MF, Orlando JM, et al: Prophylaxis of acute renal failure in patients with rhabdomyolysis. Ren Fail 1997; 19(2):283-288.
    135) Hsu J, Liu C, Hsu CP, et al: Performance characteristics of selected immunoassays for preliminary test of 3,4-methylenedioxymethamphetamine, methamphetamine, and related drugs in urine specimens. J Anal Toxicol 2003; 27:471-478.
    136) Huang HH & Bai YM: Persistent psychosis after ingestion of a single tablet of '2C-B'. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35(1):293-294.
    137) Huerta-Alardin AL, Varon J, & Marik PE: Bench-to-bedside review: Rhabdomyolysis -- an overview for clinicians. Crit Care 2005; 9(2):158-169.
    138) Humphreys A & Tanner AR: Acute dystonic drug reaction or tetanus? an unusual consequence of a 'Whizz' overdose. Human & Experiment Toxicol 1994; 13:311-312.
    139) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    140) Hysek CM, Vollenweider FX, & Liechti ME: Effects of a beta-blocker on the cardiovascular response to MDMA (Ecstasy). Emerg Med J 2010; 27(8):586-589.
    141) Insel TR, Battaglia G, & Johannessen JN: 3,4-methylenedioxymethamphetamine ("Ecstasy") selectively destroys brain serotonin terminals in Rhesus monkeys. J Pharmacol Exp Ther 1989; 249:713-720.
    142) Iwersen S & Schmoldt A: Two very different fatal cases associated with the use of methylenedioxyethylamphetamine (MDEA): eve as deadly as adam. Clin Toxicol 1996; 34:241-244.
    143) Jackson WL Jr: Toward directed therapy for amphetamine-mediated hyperthermia: is carvedilol worth raving about?. Crit Care Med 2005; 33(6):1443-1445.
    144) Johansen SS, Hansen AC, Muller IB, et al: Three fatal cases of PMA and PMMA poisoning in Denmark. J Analyt Toxicol 2003; 27:253-256.
    145) Jones AL & Simpson KJ: Review article: mechanisms and management of hepatotoxicity in ecstasy (MDMA) and amphetamine intoxications. Aliment Pharmacol Ther 1999; 13:129-133.
    146) Jones AL, Jarvie DR, & McDermid G: Hepatocellular damage following amphetamine intoxication. Clin Toxicol 1994; 32:435-444.
    147) Kirk RW: Current Veterinary Therapy IX, Saunders, Philadelphia, PA, 1986.
    148) Kleinman ME, Chameides L, Schexnayder SM, et al: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care. Part 14: pediatric advanced life support. Circulation 2010; 122(18 Suppl.3):S876-S908.
    149) Kline SS, Mauro LS, & Scala-Barnett DM: Serotonin syndrome versus neuroleptic malignant syndrome as a cause of death. Clin Pharmac 1989; 8:510-514.
    150) Koch-Weser J: Hypertensive emergencies. N Engl J Med 1974; 290:211.
    151) Kramer L, Bauer E, Schenk P, et al: Successful treatment of refractory cerebral oedema in ecstasy/cocaine-induced fulminant hepatic failure using a new high-efficiency liver detoxification device (FPSA-Prometheus). Wien Klin Wochenschr 2003; 115:599-603.
    152) Krystal JH, Price LH, & Opsahl C: Chronic 3,4-methylenedioxymethamphetamine (MDMA) use: effects on mood and neuropsychological function?. Am J Drug Alcohol Abuse 1992; 18:331-341.
    153) Lai TI, Hwang JJ, Fang CC, et al: Methylene 3,4 Dioxymethamphetamine-induced acute myocardial infarction. Ann Emerg Med 2003; 42:759-762.
    154) Lamberth PG, Ding GK, & Nurmi LA: Fatal paramethoxy-amphetamine (PMA) poisoning in the Australian Capital Territory. Med J Aust 2008; 188(7):426.
    155) Lehmann E, Thom CH, & Croft DN: Delayed severe rhabdomyolysis after taking 'ecstasy' (letter). Postgrad Med J 1995; 71:186-188.
    156) Lester SJ, Baggott M, & Welm S: Cardiovascular effects of 3,4-methylenedioxymethamphetamine: a double-blind, placebo-controlled trial. Ann Intern Med 2000; 133:969-973.
    157) Levine AJ, Rees GM, & Drew S: Ecstasy induced pneumomediastinum. J Roy Soc Med 1993; 86:232-233.
    158) Lidder S, Dargan P, Sexton M, et al: Cardiovascular toxicity associated with recreational use of diphenylprolinol (diphenyl-2-pyrrolidinemethanol [D2PM]). J Med Toxicol 2008; 4(3):167-169.
    159) Lin DL, Liu HC, & Yin HL: Recent paramethoxymethamphetamine (PMMA) deaths in Taiwan. J Anal Toxicol 2007; 31(2):109-113.
    160) Link MS, Berkow LC, Kudenchuk PJ, et al: Part 7: Adult Advanced Cardiovascular Life Support: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S444-S464.
    161) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    162) Logan ASC, Stickle B, & O'Keefe N: Survival following 'Ecstasy' ingestion with a peak temperature of 42 degrees C. Anaesthesia 1993; 48:507-513.
    163) Manchanda S & Connolly MJ: Cerebral infarction in association with Ecstasy abuse. Postgrad Med J 1993; 69:874-889.
    164) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    165) Marsh JCW, Abboudi ZH, & Gibson FM: Aplastic anaemia following exposure to 3,4-methylenedioxymethamphetamine ('Ecstasy'). Br J Haematol 1994; 88:281-285.
    166) Martin TL, Chiasson DA, & Kish SJ: Does hyperthyroidism increase risk of death due to the ingestion of ecstasy?. J Forensic Sci 2007; 52(4):951-953.
    167) Maurer HH, Moeller MR, & Roesler M: On the metabolism of 3,4-methylenedioxymethamphetamine (MDMA) in man (abstract). Ther Drug Monit 1993; 15:148.
    168) Maxwell DL, Polkey MI, & Henry JA: Hyponatraemia and catatonic stupor after taking "ecstasy". Br Med J 1993; 27:307.
    169) McCann UD & Ricaurte: MDMA ("Ecstasy") and panic disorder: induction by a single dose. Biol Psych 1992; 32:950-953.
    170) McCann UD, Slate SO, & Ricaurte GA: Adverse reactions with 3,4-methylenedioxymethamphetamine (MDMA; 'ecstasy'). Drug Safety 1996; 15:107-115.
    171) McCann UD, Szabo Z, & Scheffel U: Positron emission tomographic evidence of toxic effect of MDMA ("ecstasy") on brain serotonin neurons in human beings. Lancet 1998; 352:1433-1437.
    172) McElhatton PR, Bateman DN, & Evans C: Congenital anomalies after prenatal ecstasy exposure. Lancet 1999; 354:1441-1442.
    173) McGuire P & Fahy T: Chronic paranoid psychosis after misuse of MDMA ("Ecstasy"). BMJ 1991; 302:697.
    174) McGuire P & Fahy T: Flashbacks following MDMA (letter). Br J Psychiatry 1992; 160:276-285.
    175) McGuire PK, Cope H, & Fahy TA: Diversity of Psychopathology associated with use of 3,4-methylenedioxymethamphetamine ('Ecstasy'). Br J Psych 1994; 165:391-395.
    176) McMillian WD, Trombley BJ, Charash WE, et al: Phentolamine continuous infusion in a patient with pheochromocytoma. Am J Health Syst Pharm 2011; 68(2):130-134.
    177) Melian AM, Burillo-Putze G, Campo CG, et al: Accidental ecstasy poisoning in a toddler. Pediatr Emerg Care 2004; 20(8):534-535.
    178) Meyer MR & Maurer HH: Metabolism of designer drugs of abuse: an updated review. Curr Drug Metab 2010; 11(5):468-482.
    179) Midha KK, Hubbard JW, & Bailey K: Alpha-methyldopamine, a key intermediate in the metabolic disposition of 3,4-methylenedioxyamphetamine in vivo in dog and monkey. Drug Metab Dispos 1978; 6:623-630.
    180) Miller K: Pharmacological management of hypertension in paediatric patients. A comprehensive review of the efficacy, safety and dosage guidelines of the available agents. Drugs 1994; 48(6):868-887.
    181) Mills KC: Serotonin syndrome: a clinical update. Med Toxicol 1997; 13:763-783.
    182) Milroy CM: Ten years of 'ecstasy'. J Roy Soc Med 1999; 92:68-72.
    183) Mintzer S, Hickenbottom S, & Gilman S: Parkinsonism after taking ecstasy (letter). New Engl J Med 1999; 340:1443.
    184) Mitchell WG: Status epilepticus and acute repetitive seizures in children, adolescents, and young adults: etiology, outcome, and treatment. Epilepsia 1996; 37(S1):S74-S80.
    185) Miyajima M, Matsumoto T, & Ito S: 2C-T-4 intoxication: acute psychosis caused by a designer drug. Psychiatry Clin Neurosci 2008; 62(2):243-.
    186) Moeller KE, Lee KC, & Kissack JC: Urine drug screening: practical guide for clinicians. Mayo Clin Proc 2008; 83(1):66-76.
    187) Montastruc F, Montastruc G, Vigreux P, et al: Valvular heart disease in a patient taking 3,4-methylenedioxymethamphetamine (MDMA, 'Ecstasy'). Br J Clin Pharmacol 2012; 74(3):547-548.
    188) Mortelmans LJM, Bogaerts PJM, Hellemans S, et al: Spontaneous penumomediastinum and myocarditis following Ecstasy use: a case report. Eur J Emerg Med 2005; 12:36-38.
    189) Mueller PD & Korey WS: Death by "ecstasy": - The serotonin syndrome?. Ann Emerg Med 1998; 32:377-380.
    190) Murthy BVS, Wilkes RG, & Roberts NB: Creatine kinase isoform changes following ecstasy overdose. Anaesth Intens Care 1997; 25:156-159.
    191) Nash JF, Meltzer HY, & Gudelsky GA: Elevation of serum prolactin and corticosterone concentrations in the rat after the administration of 3,4-methylenedioxymethamphetamine. J Pharmacol Exp Ther 1988; 245:873-879.
    192) Neumar RW , Otto CW , Link MS , et al: Part 8: adult advanced cardiovascular life support: 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2010; 122(18 Suppl 3):S729-S767.
    193) Nichols DE, Ilhan M, & Long JP: Comparison of cardiovascular, hyperthermic, and toxic effects of para-methoxyamphetamine (PMA) and 3,4-methylenedioxyamphetamine (MDA). Arch Int Pharmacodyn 1975; 214:133-140.
    194) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    195) O'Neill D & Dart JK: Methylenedioxyamphetamine ('Ecstasy') associated keratopathy (letter). Eye 1993; 7:805-809.
    196) Obrocki J, Buchert R, & Vaterlein O: Ecstasy - long-term effects on the human central nervous system revealed by positron emission tomography. Br J Psy 1999; 175:186-188.
    197) Ovaska H, Viljoen A, Puchnarewicz M, et al: First case report of recreational use of 2,5-dimethoxy-4-chloroamphetamine confirmed by toxicological screening. Eur J Emerg Med 2008; 15(6):354-356.
    198) Pallanti S & Mazzi D: MDMA (ecstasy) precipitation of panic disorder. Biol Psychiatr 1992; 32:91-95.
    199) Parr MJ, Low HM, & Botterill P: Hyponatremia and death after "ecstasy" ingestion. Med J Austral 1997; 166:136-137.
    200) Patel MM, Belson MG, Longwater AB, et al: Methylenedioxymethamphetamine (Ecstasy)-related hyperthermia. J Emerg Med 2005; 29(4):451-454.
    201) Patel MM, Belson MG, Wright D, et al: Methylenedioxymethamphetamine (ecstasy)-related myocardial hyperthrophy: an autopsy study. Resuscitation 2005a; 66:197-202.
    202) Paton DM, Bell JI, & Yee R: Pharmacology and toxicity of 3,4-methylenedioxyamphetamine, para-methoxyamphetamine, and related dimethoxyamphetamines. Proc West Pharmacol Soc 1975; 18:229-231.
    203) Pearce CJ & Wallin JD: Labetalol and other agents that block both alpha- and beta-adrenergic receptors. Cleve Clin J Med 1994; 61(1):59-69.
    204) Peberdy MA , Callaway CW , Neumar RW , et al: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care science. Part 9: post–cardiac arrest care. Circulation 2010; 122(18 Suppl 3):S768-S786.
    205) Peroutka SJ: `Ecstasy': a human neurotoxin?. Arch Gen Psychiatry 1989; 46:191.
    206) Pham JV & Puzantian T: Ecstasy: Dangers and controversies. Pharmacother 2001; 21:1561-1565.
    207) Polderman KH: Acute renal failure and rhabdomyolysis. Int J Artif Organs 2004; 27(12):1030-1033.
    208) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    209) Poon WT, Lai CF, Lui MC, et al: Piperazines: a new class of drug of abuse has landed in Hong Kong. Hong Kong Med J 2010; 16(1):76-77.
    210) Price LH, Ricaurte GA, & Krystal JH: Neuroendocrine and mood responses to intravenous L-tryptophan in 3,4-methylenedioxymethamphetamine (MDMA) users. Arch Gen Psychiatry 1989; 46:20-22.
    211) Priori A, Bertolasi L, & Berardelli A: Acute dystonic reaction to Ecstasy (letter). Movement Dis 1995; 10:353.
    212) Product Information: ATIVAN(R) injection, lorazepam injection. Baxter Healthcare Corporation, Deerfield, IL, 2003.
    213) Product Information: BREVIBLOC(TM) intravenous injection, esmolol HCl intravenous injection. Baxter Healthcare Corporation (per FDA), Deerfield, IL, 2012.
    214) Product Information: Cordarone(R) oral tablets, amiodarone HCl oral tablets. Wyeth Pharmaceuticals Inc (per FDA), Philadelphia, PA, 2015.
    215) Product Information: Lidocaine HCl intravenous injection solution, lidocaine HCl intravenous injection solution. Hospira (per manufacturer), Lake Forest, IL, 2006.
    216) Product Information: NITROPRESS(R) injection for IV infusion, Sodium Nitroprusside injection for IV infusion. Hospira, Inc., Lake Forest, IL, 2007.
    217) Product Information: NITROPRESS(R) injection, sodium nitroprusside injection. Hospira,Inc, Lake Forest, IL, 2004.
    218) Product Information: Phentolamine Mesylate IM, IV injection Sandoz Standard, phentolamine mesylate IM, IV injection Sandoz Standard. Sandoz Canada (per manufacturer), Boucherville, QC, 2005.
    219) Product Information: Trandate(R) IV injection, labetalol hydrochloride IV injection. Prometheus Laboratories Inc., San Diego, CA, 2010.
    220) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    221) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    222) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    223) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    224) Quang LS, Boyer EW, & Chumpa AL: Hyponatremia and altered mental status from water intoxication and ecstasy-induced syndrome of inappropriate antidiuretic hormone resulting in a motor vehicle accident (abstract). J Toxicol-Clin Toxicol 2001; 39:290.
    225) Quin GI, McCarthy GM, & Harries DK: Spontaneous pneumomediastinum and ecstasy abuse (letter). J Accid Emerg Med 1999; 16:382.
    226) Qureshi A, Wassmer E, Davies P, et al: Comparative audit of intravenous lorazepam and diazepam in the emergency treatment of convulsive status epilepticus in children. Seizure 2002; 11(3):141-144.
    227) Ramcharan S, Meenhorst PL, & Otten JM: Survival after massive ecstasy overdose. Clin Tox 1998; 36:727-731.
    228) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    229) Ravina P, Quiroga JM, & Ravina T: Hyperkalemia in fatal MDMA ('ecstasy') toxicity. Intl J Cardiol 2004; 93:307-308.
    230) Refstad S: Paramethoxyamphetamine (PMA) poisoning; a 'party drug' with lethal effects. Acta Anaesthesiol Scand 2003; 47(10):1298-1299.
    231) Rella JG, Nelson LS, & Hoffman RS: 7 years of 3,4-methylenedioxymethamphetamine (MDMA) toxicity. Int J Med Toxicol 2000; 3(5):28.
    232) Reneman L, Booij J, & de Bruin K: Effects of dose, sex, and long-term abstention from use on toxic effects of MDMA (ecstasy) on brain serotonin neurons. Lancet 2001; 358:1864-1869.
    233) Reneman L, deWin MM, vandenBrink W, et al: Neuroimaging findings with MDMA/ecstasy: technical aspects, conceptual issues and future prospects. J Psychopharmacol 2006; 20(2):164-175.
    234) Renfroe CL: MDMA on the street: Analysis Anonymous(R). J Psychoactive Drugs 1986; 18:363-369.
    235) Rhoney D & Peacock WF: Intravenous therapy for hypertensive emergencies, part 1. Am J Health Syst Pharm 2009; 66(15):1343-1352.
    236) Ricaurte G, Bryan G, & Strauss L: Hallucinogenic amphetamine selectively destroys brain serotonin nerve terminals. Science 1985; 229:986-988.
    237) Ricaurte GA, DeLanney LE, & Irwin I: Toxic effects of MDMA on central serotonergic neurons in the primate: importance of route and frequency of drug administration. Brain Res 1988; 446:165-168.
    238) Ricaurte GA, Forno LS, & Wilson Ma: (+/-)3,4-methylenedioxymethamphetamine selectively damages central serotonergic neurons in nonhuman primates. JAMA 1988a; 260:51-55.
    239) Rittoo D, Rittoo DB, & Rittoo D: Misuse of ecstasy (letter). Br Med J 1992; 309-310.
    240) Rohrig TP & Prouty RW: Tissue distribution of methylenedioxymethamphetamine. J Anal Toxicol 1992; 16:52-53.
    241) Rome ES: It's a rave new world: Rave culture and illicit drug use in the young. Cleveland Clin J Med 2001; 68:541-550.
    242) Rosenson J, Smollin C, Sporer KA, et al: Patterns of ecstasy-associated hyponatremia in California. Ann Emerg Med 2007; 49(2):164-171.
    243) Rothwell PM & Grant R: Cerebral venous sinus thrombosis induced by 'ecstasy' (letter). J Neurol, Neurosurg & Psych 1993; 5:1035.
    244) Russell ARB, Schwartz RH, & Dawling S: Accidental ingestion of 'ecstasy' (3,4-methylenedioxymethylamphetamine). Arch Dis Child 1992; 67:1114-1115.
    245) Sano R , Hasuike T , Nakano M , et al: A fatal case of myocardial damage due to misuse of the "designer drug" MDMA. LegMed (Tokyo) 2009; 11(6):294-297.
    246) Sawyer J & Stephens WP: Misuse of ecstasy (letter). Br Med J 1992; 305:310.
    247) Schifano F, Deluca P, Agosti L, et al: New trends in the cyber and street market of recreational drugs? The case of 2C-T-7 ('Blue Mystic'). J Psychopharmacol 2005; 19(6):675-679.
    248) Schifano F: Chronic atypical psychosis associated with MDMA ("ecstasy") abuse. Lancet 1991; 338:1335.
    249) Schmidt CJ, Wu L, & Lovenberg W: Methylenedioxymethamphetamine: a potentially neurotoxic amphetamine analogue. Eur J Pharmacol 1986; 124:175-178.
    250) Schmidt CJ: Acute administration of methylenedioxymethamphetamine: comparison with the neurochemical effects of its N-desmethyl and N-ethyl analogs. Eur J Pharmacol 1987a; 81-88.
    251) Schmidt CJ: Neurotoxicity of the psychedelic amphetamine, methylenedioxymethamphetamine. J Pharmacol Exp Ther 1987; 240:1-7.
    252) Schwartz RH & Miller NS: MDMA (Ecstasy) and the Rave: A review. Pediatrics 1997; 100:705-708.
    253) Scott R, Besag FMC, & Neville BGR: Buccal midazolam and rectal diazepam for treatment of prolonged seizures in childhood and adolescence: a randomized trial. Lancet 1999; 353:623-626.
    254) Screaton GR, Singer M, & Cairns HS: Hyperpyrexia and rhabdomyolysis after MDMA ("ecstasy") abuse (letter). Lancet 1992; 339:677-678.
    255) Shannon M: Methylenedioxymethamphetamine (MDMA, "Ecstasy"). Ped Emerg Care 2000; 16:377-380.
    256) Sharma R & Nelson LS: Methylenedioxymethylamphetamine (MDMA) induced hyponatremia. Int J Med Toxicol 2000; 3:29.
    257) Shearman JD, Satsangi J, & Chapman RWG: Misuse of ecstasy (letter). Br Med J 1992; 305:309.
    258) Sherlock K, Wolff K, & Hay AWM: Analysis of illicit ecstasy tablets: implications for clinical management in the accident and emergency department. J Acad Emerg Med 1999; 16:194-197.
    259) Shulgin AT: Profiles of psychedelic drugs - DOB. J Psychoactive Drugs 1981; 13:99.
    260) Siegel RK: MDMA nonmedical use and intoxication. J Psychoactive Drugs 1986; 18:349-354.
    261) Simpson DL & Rumack BH: Methylenedioxyamphetamine: clinical description of overdose, death, and review of pharmacology. Arch Intern Med 1981; 141:1507-1509.
    262) Singarajah C & Lavies NG: An overdose of ecstasy: a role for dantrolene. Anaesthesia 1992; 47:686-687.
    263) Singer LT, Moore DG, Min MO, et al: Developmental outcomes of 3,4-methylenedioxymethamphetamine (ecstasy)-exposed infants in the UK. Hum Psychopharmacol 2015; 30(4):290-294.
    264) Singh D, Akingbola O, Yosypiv I, et al: Emergency management of hypertension in children. Int J Nephrol 2012; 2012:420247.
    265) Slikker W Jr, Holson RR, & Ali SF: Behavioral and neurochemical effects of orally administered MDMA in the rodent and nonhuman primate. NeuroToxicol 1989; 10:529-542.
    266) Smilkstein MJ, Smolinske SC, & Rumack BH: MAO inhibitor/MDMA interaction: agony after ecstasy. Clin Toxicol 1987; 25:149-159.
    267) Snyder SH, Richelson E, & Weingartner H: Psychotropic methoxyamphetamines: structure and activity in man. In: Amphetamines and Related Compounds; Proceedings of the Mario Negri Institute for Pharmacological Research, E Costa and S Garattini (eds), Raven Press, 1970, pp 905-927.
    268) Sprague JE, Moze P, Caden D, et al: Carvedilol reverses hyperthermia and attenuates rhabdomyolysis induced by 3,4-methylenedioxymethamphetamine (MDMA, Ecstasy) in an animal model. Crit Care Med 2005; 33(6):1311-1316.
    269) Springer D, Fritschi G, & Maurer HH: Metabolism of the new designer drug alpha-pyrrolidinopropiophenone (PPP) and the toxicological detection of PPP and 4'-methyl-alpha-pyrrolidinopropiophenone (MPPP) studies in rat urine using gas chromatography-mass spectrometry. J Chromatography B 2003; 796:253-266.
    270) Sreenath TG, Gupta P, Sharma KK, et al: Lorazepam versus diazepam-phenytoin combination in the treatment of convulsive status epilepticus in children: A randomized controlled trial. Eur J Paediatr Neurol 2010; 14(2):162-168.
    271) Steele TD, McCann UD, & Ricaurte GA: 3,4-methylenedioxymethamphetamine (MDMA, "ecstasy"): pharmacology and toxicology in animals and humans. Addiction 1994; 89:539-551.
    272) Sternbach H: The serotonin syndrome. Am J Psychiatr 1991; 148:705-713.
    273) Stone DM, Stahl DC, & Hanson GR: The effects of 3,4-methylenedioxymethamphetamine (MDMA) and 3,4-methylenedioxyamphetamine (MDA) on monoaminergic systems in the rat brain. Eur J Pharmacol 1986; 128:41-48.
    274) Suarez RV & Riemersma R: "Ecstasy" and sudden cardiac death. Am J Forensic Med Pathol 1988; 9:339-341.
    275) Swan MC, Lam D, & Giele HP: Intravascular ecstasy: an unusual cause of thigh compartment syndrome. J Trauma 2006; 60(5):1129-1131.
    276) Tanner-Smith EE: Pharmacological content of tablets sold as "ecstasy": results from an online testing service. Drug Alcohol Depend 2006; 83(3):247-254.
    277) Tehan B: Ecstasy and dantrolene (letter). Br Med J 1993; 306.
    278) Temple ME & Nahata MC: Treatment of pediatric hypertension. Pharmacotherapy 2000; 20(2):140-150.
    279) Thiessen PN & Cook DA: The properties of 3,4-methylenedioxyamphetamine (MDA). II. Studies of acute toxicity in the mouse and protection by various agents. Clin Toxicol 1973b; 6:193-199.
    280) Thomasius R, Petersen KU, Zapletalova P, et al: Mental disorders in current and former heavy ecstasy (MDMA) users. Addiction 2005; 100(9):1310-1319.
    281) Tillman HL, van Pelt F, & Martz W: Accidental intoxication with methylene dianiline p,p'-diaminodiphenylmethane: acute liver damage after presumed ecstasy consumption. Clin Toxicol 1997; 35:35-40.
    282) U.S. Department of Health and Human Services; National Institutes of Health; and National Heart, Lung, and Blood Institute: The seventh report of the Joint National Committee on Prevention, Detection, Evaluation and Treatment of High Blood Pressure. U.S. Department of Health and Human Services. Washington, DC. 2004. Available from URL: http://www.nhlbi.nih.gov/guidelines/hypertension/jnc7full.pdf. As accessed 2012-06-20.
    283) Vanden Eede H, Montenij LJ, Touw DJ, et al: Rhabdomyolysis in MDMA intoxication: a rapid and underestimated killer. "Clean" Ecstasy, a safe party drug?. J Emerg Med 2012; 42(6):655-658.
    284) Vanholder R, Sever MS, Erek E, et al: Rhabdomyolysis. J Am Soc Nephrol 2000; 11(8):1553-1561.
    285) Verebey K, Alrazi JJ, & Jaffee JH: The complications of 'Ecstasy'(MDMA). J Am Vet Med Assoc 1988; 259:1649-1650.
    286) Wake D: Ecstasy overdose: a case study. Intens Crit Care Nsg 1995; 11:6-9.
    287) Walter LA & Catenacci MH: Rhabdomyolysis. Hosp Physician 2008; 44(1):25-31.
    288) Walubo A & Seger D: Fatal multi-organ failure after suicidal overdose with MDMA, 'ecstasy': case report and review of the literature. Hum Experimen Toxicol 1999; 18:119-125.
    289) Watson JD, Ferguson C, & Hinds CJ: Exertional heat stroke induced by amphetamine analogues. Anaesthesia 1993; 48:1057-1060.
    290) Webb C & Williams V: Ecstasy intoxication: appreciation of complications and the role of dantrolene. Anaesthesia 1993; 18:542-543.
    291) Wheless JW : Treatment of status epilepticus in children. Pediatr Ann 2004; 33(6):376-383.
    292) Whitaker-Azmitia PM & Aronson TA: "Ecstasy" (MDMA)-induced panic. Am J Psychiatry 1989; 146:1.
    293) White JM, Bochner F, & Irvine RJ: The agony of "ecstasy". Med J Austral 1997; 166:117-118.
    294) Wohlfarth A, Weinmann W, & Dresen S: LC-MS/MS screening method for designer amphetamines, tryptamines, and piperazines in serum. Anal Bioanal Chem 2010; 396(7):2403-2414.
    295) Wood DM , Looker JJ , Shaikh L , et al: Delayed onset of seizures and toxicity associated with recreational use of Bromo-dragonFLY. J Med Toxicol 2009; 5(4):226-229.
    296) Wood DM, Button J, Lidder S, et al: Dissociative and sympathomimetic toxicity associated with recreational use of 1-(3-trifluoromethylphenyl) piperazine (TFMPP) and 1-benzylpiperzine (BZP). J Med Toxicol 2008; 4(4):254-257.
    297) Wood DM, Dargan PI, Button J, et al: Collapse, reported seizure--and an unexpected pill. Lancet 2007; 369(9571):1490.
    298) Woods JD & Henry JA: Hyperpyrexia induced by 3,4-methylenedioxyamphetamine ("Eve") (letter). Lancet 1992; 340:305.
    299) Zenenberg R & Goldfarb DS: Evaluation of hyponatremia associated with use of methylenedioxymethylamphetamine (MDMA). Int J Med Toxicol 2000; 3:30.
    300) de Caen AR, Berg MD, Chameides L, et al: Part 12: Pediatric Advanced Life Support: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S526-S542.
    301) de Fallois J, Regenthal R, & Petros S: Fulminant thrombotic thrombocytopenic purpura (TTP): association with amphetamine consumption?. Ann Hematol 2015; 94(2):337-338.
    302) de la Torre R, Ortuno J, & Mas M: Fatal MDMA intoxication (letter). Lancet 1999; 353:593.