MOBILE VIEW  | 

PHENETHYLAMINE DESIGNER DRUGS-2C SERIES

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) 2C designer drugs are sold alone or in combination with other agents in many countries, including the United States. Their main phenethylamine-based structure is shared among amphetamines, catecholamines, cathinones, and many other drugs. Phenethylamines stimulate catecholamine, dopamine and serotonin activity. Naturally occurring phenethylamines include mescaline and other nonnative cactus species (See PLANTS-PEYOTE/MESCALINE).
    B) BODY PACKERS/BODY STUFFERS: Please refer to the appropriate management if body packing or body stuffing is known/suspected.
    C) 25I-NBOMe (25-I): A (n-benzyl) phenethylamine is discussed in the management: PHENETHYLAMINE DESIGNER DRUGS-N-BENZYL SERIES.

Specific Substances

    A) GENERAL TERMS
    1) 2C agents
    2) 2C family
    3) 2Cs
    4) 2C series
    5) 2,5-phenethylamines
    6) Designer drugs - 2C
    7) Designer drugs - 2C series
    8) Psychedelic phenethylamine
    2C-B (synonym)
    1) 2CB (synonym)
    2) 4-bromo-2,5-dimethoxy-beta-phenylethylamine (synonym)
    3) Afterburner (street name for 2C-B) (synonym)
    4) Bdmpea (street name for 2C-B) (synonym)
    5) Bees (street name for 2C-B) (synonym)
    6) Bromo (street name for 2C-B) (synonym)
    7) Cb2 (street name for 2C-B) (synonym)
    8) Erox (street name for 2C-B) (synonym)
    9) Eve (street name for 2C-B) (synonym)
    10) MFT (street name for 2C-B) (synonym)
    11) Nexus (street name for 2C-B) (synonym)
    12) Performax (street name for 2C-B) (synonym)
    13) Spectrum (street name for 2C-B) (synonym)
    14) Toonies (street name for 2C-B) (synonym)
    15) Venus (street name for 2C-B) (synonym)
    2C-B-Fly (synonym)
    1) 2CBFly (synonym)
    2) 1-(8-Bromo-2,3,6,7-tetrahydrobenzo[1,2-b:4,5-b'] difuran-4-yl)-2-aminoethane (synonym)
    3) 2C-Bromo-Fly (synonym)
    4) 2CBromoFly (synonym)
    2C-C (synonym)
    1) 2CC (synonym)
    2) 2,5-dimethoxy-4-chlorophenethylamine (synonym)
    3) 4-chloro-2,5-dimethoxyphenethylamine (synonym)
    2C-D (synonym)
    1) 2CD (synonym)
    2) 2,5-dimethoxy-4-methyl-beta-phenethylamine (synonym)
    2C-E (synonym)
    1) 2CE (synonym)
    2) 4-ethyl-2,5-dimethoxy-beta-phenethylamine (synonym)
    3) 2,5-dimethoxy-4-ethyl-phenethylamine (synonym)
    4) Europa (street name for 2C-E) (synonym)
    2C-F (synonym)
    1) 2CF (synonym)
    2) 4-fluoro-2,5-dimethoxyphenethylamine (synonym)
    2C-G (synonym)
    1) 2CG (synonym)
    2) 3,4-dimethyl-2,5-dimethoxyphenethylamine (synonym)
    2C-G-3 (synonym)
    1) 2CG3 (synonym)
    2) 2,5-dimethyoxy-3,4-(trimethylene)phenethylamine (synonym)
    2C-G-5 (synonym)
    1) 2CG5 (synonym)
    2) 3,6-dimethoxy-4-(2-aminoethyl)benzonorbornane (synonym)
    2C-I (synonym)
    1) 2CI (synonym)
    2) 4-iodo-2,5-dimethoxy-beta-phenethylamine (synonym)
    3) 2,5-dimethoxy-4-iodophenethylamine (synonym)
    2C-N (synonym)
    1) 2CN (synonym)
    2) 2,5-dimethoxy-4-nitrophenethylamine (synonym)
    2C-P (synonym)
    1) 2CP (synonym)
    2) 2,5-dimethoxy-4-propylphenethylamine (synonym)
    3) 2-(2,5-dimethoxy-4-propylphenyl)ethanamine (synonym)
    2C-SE (synonym)
    1) 2CSE (synonym)
    2) 2,5-dimethoxy-4-methylselenophenethylamine (synonym)
    2C-T-2 (synonym)
    1) 2CT2 (synonym)
    2) 4-ethylthio-2,5-dmiethoxy-beta-phenethylamine (synonym)
    2C-T-4 (synonym)
    1) 2CT4 (synonym)
    2) 2-[4-(isopropylthio)-2,5-dimethoxyphenyl]ethanamine (synonym)
    3) Vanilla Aroma (street name for 2C-T-4) (synonym)
    2C-T-7 (synonym)
    1) 2CT7 (synonym)
    2) 2,5-dimethoxy-4-propylthio-beta-phenethylamine (synonym)
    3) 2,5-dimethoxy-4-n-propylthiophenethylamine (synonym)
    4) 7th heaven (street name for 2C-T-7) (synonym)
    5) 7-Up (street name for 2C-T-7) (synonym)
    6) Blue Mystic (street name for 2C-T-7) (synonym)
    7) Lucky 7 (street name for 2C-T-7) (synonym)
    8) PT-DM-PEA (street name for 2C-T-7) (synonym)
    9) Red raspberry (street name for 2C-T-7) (synonym)
    10) Tripstay (street name for 2C-T-7) (synonym)
    11) Tripstasy (street name for 2C-T-7) (synonym)
    12) Tweety-Bird Mescaline (street name for 2C-T-7) (synonym)
    13) T7 (synonym)
    2C-T-8 (synonym)
    1) 2CT8 (synonym)
    2) 4-cyclopropylmethylthio-2,5-dimethoxy-phenethylamine (synonym)
    2C-T-9 (synonym)
    1) 2CT9 (synonym)
    2) 4-(t)-butylthio-2,5-dimethoxy-phenethylamine (synonym)
    2C-T-13 (synonym)
    1) 2CT13 (synonym)
    2) 4-(2-Methoxyethylthio)-2,5-dimethoxy-phenethylamine (synonym)
    2C-T-15 (synonym)
    1) 2CT15 (synonym)
    2) 4-Cyclopropylthios-2,5-dimethoxy-phenethylamine (synonym)
    2C-T-17 (synonym)
    1) 2CT17 (synonym)
    2) 4-(s)-Butylthio-2,5-dimethoxy-phenethylamine (synonym)
    2C-TFM (synonym)
    1) 2CTFM (synonym)
    2) 4-trifluoromethyl-2,5-dimethoxyphenethylamine (synonym)
    3) 2-[2,5-dimethoxy-4-(trifluoromethyl)phenyl]ethanamine (synonym)
    2C-T-21 (synonym)
    1) 2CT21 (synonym)
    2) 4-(2-Fluoroethylthio)-2,5-dimethoxy-phenethylamine (synonym)

Available Forms Sources

    A) FORMS
    1) The 2C compounds are usually ingested or insufflated. They are available in tablet, capsule, powder, and liquid forms (Dean et al, 2013).
    2) 2C-B: 2C-B tablets usually contain 5 mg of 2C-B (Cole et al, 2002; de Boer & Bosman, 2004). 2C-B may be found in some Ecstasy tablets (de Boer et al, 1999). It may also be taken in combination with ketamine (Giroud et al, 1998).
    3) 2C-T-2: In one study, some tablets from Netherlands contained 8 mg of 2C-T-2 (de Boer & Bosman, 2004).
    4) 2C-T-4: This agent is a phenethylethanamine analog and structurally similar to 2C-T-7 (Miyajima et al, 2008).
    5) 2C-T-7: In one study, Blue Mystics and 2C-T-7 tablets from Netherland contained up to 10 mg of 2C-T-7 (de Boer & Bosman, 2004).
    B) USES
    1) 2C designer drugs are sold alone or in combination with other agents in many countries, including the United States. These agents are primarily used as drugs of abuse (Meyer & Maurer, 2010).
    2) 2C-T-4: found in a sexual enhancing product called "Vanilla Aroma" (Miyajima et al, 2008).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: 2C agents are used primarily as drugs of abuse.
    B) TOXICOLOGY: Phenethylamines cause clinical effects by a complex stimulation of catecholamine, dopamine and serotonin activity. Although the mechanism of action of phenethylamines is not completely understood, it is thought that all hallucinogens share a common site of action on central 5-HT2 receptors. The dopaminergic system is also involved. Following 2C intoxication, patients may present with either a sympathomimetic syndrome, serotonin toxicity, hallucinogenic effects, or a combination of these effects. Lower doses (eg, less than 10 mg for 2C-B) primarily produce stimulant effects and increased visual, auditory, and tactile sensations, 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 and sympathomimetics signs, such as tachycardia, hypertension, and hyperthermia.
    C) EPIDEMIOLOGY: Poisoning is not common, but may be severe. These drugs are most commonly ingested or nasally insufflated, but inhalational or parenteral abuse is possible depending on the substance.
    D) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: 2C agents have the potential to cause either pleasant or frightening hallucinations. Nausea, vomiting, diarrhea, abdominal pain, mydriasis, dizziness, sweating, headache, paresthesia, dizziness, tachycardia and hypertension may also occur.
    2) SEVERE TOXICITY: Severe effects may include severe agitation, tachypnea, hyperthermia, delirium, psychosis, seizures, disseminated intravascular coagulation (DIC), pulmonary edema, respiratory depression, 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. Excited delirium has been reported with 2C intoxication. It is characterized by delirium with agitation, violence, hyperactivity, and in many cases hyperthermia and cardiopulmonary arrest. One patient developed serotonin syndrome after insufflating liquid 2C-I.
    3) BODY PACKERS/BODY STUFFERS: Please refer to the appropriate management if body packing or body stuffing is known/suspected.

Laboratory Monitoring

    A) Plasma concentrations are not clinically useful or readily available.
    B) Standard urine toxicology screen may not produce a positive result for many 2C agents.
    C) Monitor vital signs and mental status.
    D) Monitor serum electrolytes and renal function.
    E) Obtain ECG and institute continuous cardiac monitoring in patients with moderate to severe toxicity (ie, agitation, delirium, seizures, coma, hypotension).
    F) Monitor creatinine phosphokinase in patients with prolonged agitation, seizures or coma; monitor renal function and urine output in patients with rhabdomyolysis.

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. Seizures may require aggressive use of benzodiazepines, propofol and/or barbiturates. Monitor and treat for dysrhythmias. Monitor core temperature and treat hyperthermia with aggressive benzodiazepine sedation to control agitation, external cooling. Clinical manifestations may be prolonged due to the long half-life of many hallucinogenic amphetamines.
    C) DECONTAMINATION
    1) PREHOSPITAL: Prehospital gastrointestinal decontamination is not recommended because of the potential for agitation and seizures and subsequent aspiration.
    2) HOSPITAL: Gastrointestinal decontamination is often NOT needed because the patient presents several hours after ingestion when absorption is complete. Administer activated charcoal if recent ingestion or possibility of coingestants, and only if the patient is cooperative and can protect their airway or is intubated. Administer to body stuffers that are not at risk for aspiration.
    D) AIRWAY MANAGEMENT
    1) Perform early in a patient with severe intoxication (ie, seizures, dysrhythmias, severe delirium or hyperthermia).
    E) ANTIDOTE
    1) None.
    F) SEIZURES
    1) Treat seizures with IV benzodiazepines, propofol or barbiturates if seizures persist or recur.
    G) HYPERTHERMIA
    1) Control agitation with benzodiazepines; initiate aggressive external cooling measures. Remove the patient's clothing, keep skin damp and use fans to enhance evaporative cooling. A cooling blanket and ice packs to the groin and axilla may be useful. For severe cases, ice water immersion may be necessary, but it can make access to the patient for resuscitation more cumbersome. If needed, intubate, sedate and paralyze.
    H) DELIRIUM
    1) Sedate patient with benzodiazepines as necessary; large doses may be required. Consider antipsychotics in patients with persistent delirium, despite high-dose benzodiazepines. Minimize external stimuli; place in quiet, dark room.
    I) SEROTONIN SYNDROME
    1) Treat aggressively with benzodiazepines, external cooling measures, if needed. Severe cases may require orotracheal intubation and neuromuscular paralysis. CYPROHEPTADINE is an oral antihistamine with serotonin antagonism at 5-hydroxytryptamine (HT)-1A and 5-HT-2A receptors. It is used to improve mild to moderate symptoms of serotonin syndrome (eg, core temperature greater than 38.5 degrees C or severe muscle rigidity requiring neuromuscular paralysis). DOSE: ADULT: Administer an initial dose of 12 mg, then 2 mg every 2 hours if the patient remains symptomatic. Maintenance: 8 mg every 6 hours. Maximum dose: 32 mg in 24 hours. CHILD: 0.25 mg/kg/day divided every 6 hours, maximum 12 mg/day. It may be crushed and administered via a nasogastric tube.
    J) ENHANCED ELIMINATION
    1) Hemodialysis and hemoperfusion are of no value because of the large volume of distribution.
    K) PATIENT DISPOSITION
    1) HOME CRITERIA: Asymptomatic adults may be monitored at home, if a responsible adult is present.
    2) OBSERVATION CRITERIA: Patients with deliberate self-harm ingestions, children with any ingestion, and symptomatic patients should be sent to a healthcare facility for evaluation, treatment and observation for 6 to 8 hours.
    3) ADMISSION CRITERIA: Patients with significant persistent central nervous system toxicity (ie, hallucinations, somnolence, delirium, coma), 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 (ie, seizures, dysrhythmias, severe delirium, coma), or in whom the diagnosis is not clear. Refer patients for substance abuse counseling as appropriate.
    L) 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 evaluated for intracranial hemorrhage, infection, and metabolic disturbance.
    M) TOXICOKINETICS
    1) INSUFFLATION: ONSET: 5 to 7 minutes; DURATION: 2 to 4 hours. ORAL: ONSET: 1 to 2.5 hours; PEAK: 2 hours; DURATION: 5 to 7 hours. Vd: Although it has not been fully established, it is thought to be large (greater than 5 L/kg). Metabolism: The main method of 2C metabolism is by O-demethylation at positions 2 and 5. It is deaminated (major enzymes, MAO-A and MAO-B) and then oxidated to either acid or base. 2C-T-7 is excreted unchanged in the urine. 2C-B and its metabolites are excreted in urine for up to 3 hours postingestion. Abusers will often "stack" doses of the drug, prolonging the apparent half-life and clinical effects.
    N) 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) TOXICITY: Severe toxicity has developed following recreational doses in some individuals. A man developed agitation, hallucinations, tachycardia, and fever after insufflating about 0.1 g of 25I-NBOMe powder. He recovered following supportive care. FATALITIES: Two men died after insufflating 30 to 35 mg of 2C-T-7. Fatalities have also been reported following the use of 2C-E, 2C-T-21, and 2C-I-NBOMe (unknown amounts).
    B) TYPICAL DOSES: 2C-B: 12 to 24 mg; 2C-B doses up to 100 mg have also been used and tolerated in some cases. 2C-B powder form doses for insufflation are usually one third of the oral dose. 2C-C: 20 to 40 mg; 2C-D: 20 to 60 mg; 2C-E: 10 to 25 mg; 2C-G: 20 to 35 mg; 2C-G-3: 16 to 25 mg; 2C-G-5: 10 to 16 mg; 2C-I: 14 to 22 mg; 2C-N: 100 to 150 mg; 2C-P: 6 to 10 mg; 2C-SE: approximately 100 mg; 2C-T: 60 to 100 mg; 2C-T-2: 12 to 25 mg; 2C-T-4: 8 to 20 mg; 2C-T-7: 10 to 30 mg; 2C-T-8: 30 to 50 mg; 2C-T-9: 60 to 100 mg; 2C-T-13: 25 to 40 mg; 2C-T-15: greater than 30 mg; 2C-T-17: 60 to 100 mg; 2C-T-21: 8 to 12 mg.

Summary Of Exposure

    A) USES: 2C agents are used primarily as drugs of abuse.
    B) TOXICOLOGY: Phenethylamines cause clinical effects by a complex stimulation of catecholamine, dopamine and serotonin activity. Although the mechanism of action of phenethylamines is not completely understood, it is thought that all hallucinogens share a common site of action on central 5-HT2 receptors. The dopaminergic system is also involved. Following 2C intoxication, patients may present with either a sympathomimetic syndrome, serotonin toxicity, hallucinogenic effects, or a combination of these effects. Lower doses (eg, less than 10 mg for 2C-B) primarily produce stimulant effects and increased visual, auditory, and tactile sensations, 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 and sympathomimetics signs, such as tachycardia, hypertension, and hyperthermia.
    C) EPIDEMIOLOGY: Poisoning is not common, but may be severe. These drugs are most commonly ingested or nasally insufflated, but inhalational or parenteral abuse is possible depending on the substance.
    D) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: 2C agents have the potential to cause either pleasant or frightening hallucinations. Nausea, vomiting, diarrhea, abdominal pain, mydriasis, dizziness, sweating, headache, paresthesia, dizziness, tachycardia and hypertension may also occur.
    2) SEVERE TOXICITY: Severe effects may include severe agitation, tachypnea, hyperthermia, delirium, psychosis, seizures, disseminated intravascular coagulation (DIC), pulmonary edema, respiratory depression, 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. Excited delirium has been reported with 2C intoxication. It is characterized by delirium with agitation, violence, hyperactivity, and in many cases hyperthermia and cardiopulmonary arrest. One patient developed serotonin syndrome after insufflating liquid 2C-I.
    3) BODY PACKERS/BODY STUFFERS: Please refer to the appropriate management if body packing or body stuffing is known/suspected.

Vital Signs

    3.3.3) TEMPERATURE
    A) WITH POISONING/EXPOSURE
    1) Hyperthermia occurs in severe cases (Dean et al, 2013; Drees et al, 2009). It may be life threatening, and has been contributory to death (Dean et al, 2013).
    2) CASE SERIES (25I-NBOMe): Hyperpyrexia developed in 3 of 7 patients after using 25I-NBOMe (Hill et al, 2013).
    a) A 21-year-old man developed agitation, hallucinations, tachycardia, and fever after insufflating about 0.1 g of 25I-NBOMe powder. He recovered gradually following supportive care (Hill et al, 2013).
    3) CASE REPORT 2C-T-7: A 17-year-old man developed vomiting, agitation, violence/aggression, possible hyperthermia, rigidity and cardiopulmonary arrest, and died after insufflating 30 to 35 mg of 2C-T-7 (Dean et al, 2013).
    4) CASE REPORT (2C-T-21): A 22-year-old man developed hyperthermia (108 degrees C), seizures and coma, and died after ingesting an unknown amount (dipping his tip of tongue into the powder) of 2C-T-21 (Dean et al, 2013).
    5) CASE REPORT (2C-E): A 19-year-old man developed agitation, aggression, hyperthermia, disseminated intravascular coagulation (DIC) and multi-organ failure, and died after insufflating an unknown amount of 2C-E (Dean et al, 2013).

Heent

    3.4.2) HEAD
    A) WITH POISONING/EXPOSURE
    1) JAW CLENCHING (2C-B): In a study of 2C-B use in Spain, jaw clenching was reported in 4 (11%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    2) STIFF NECK (2C-B): In a study of 2C-B use in Spain, stiff neck was reported in 1 (3%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    3.4.3) EYES
    A) WITH POISONING/EXPOSURE
    1) MYDRIASIS (25I-NBOMe): Mydriasis has been reported with 25I-NBOMe (a (n-benzyl) phenethylamine in the 2C family) use (Rose et al, 2013).
    2) DIFFICULTY FOCUSING GAZE (2C-B): In a study of 2C-B use in Spain, difficulty focusing gaze was reported in 13 (37%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    3.4.5) NOSE
    A) WITH POISONING/EXPOSURE
    1) CASE REPORT (2C-T-7): A 20-year-old man developed vomiting, hallucinations, agitation, violence/aggression, nasal bleeding, seizures, pulmonary edema and cardiopulmonary arrest, and died after insufflating 35 mg of 2C-T-7 (Curtis et al, 2003; Dean et al, 2013).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) HYPERTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) Hypertension may occur (Rose et al, 2013; Drees et al, 2009).
    b) CASE SERIES (25I-NBOMe): Hypertension developed in 4 of 7 patients after using 25I-NBOMe (Hill et al, 2013).
    1) A 29-year-old man presented with agitation, aggression, seizures, tachycardia, hypertension, tachypnea, and fever after the intravenous injection of 3 mL of 25I-NBOMe (unknown concentration). Laboratory results revealed respiratory and metabolic acidosis, elevation of creatine kinase, impaired renal function, and elevated alanine transaminase. A CT scan of the head showed mild cerebral edema. Despite supportive care, he developed anuria with acute kidney injury within the next 2 days and he underwent a continuous veno-venous hemofiltration (CVVH). His condition deteriorated later with fever, high-oxygen and vasopressor requirements and worsening renal impairment. Acute lung injury and pulmonary abscesses were observed in chest radiographs. Following a complicated hospital course and aggressive supportive care, his condition gradually normalized and he was discharged home 43 days after presentation (Hill et al, 2013).
    c) CASE REPORT (25I-NBOMe): An 18-year-old man presented with severe agitation and hallucinations after using 25I-NBOMe (a (n-benzyl) phenethylamine in the 2C family) and then jumping out of a moving car. On admission, he had tachycardia (heart rate 150 to 160 beats/min) and hypertension (BP 150-170 mmHg systolic and 110 mm Hg diastolic). Laboratory results revealed hypokalemia (potassium 2.8 mEq/L; 3.6 to 5.2 mEq/L). Following supportive care, his symptoms gradually improved; however, he continued to have episodes of aggressiveness for the next 24 hours (Rose et al, 2013).
    B) TACHYCARDIA
    1) WITH POISONING/EXPOSURE
    a) Tachycardia is common (Rose et al, 2013; Caudevilla-Galligo et al, 2012).
    b) 2C-B: In a study of 2C-B use in Spain, tachycardia was reported in 4 (11%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    c) CASE SERIES (25I-NBOMe): Tachycardia developed in all 7 patients after using 25I-NBOMe (Hill et al, 2013).
    1) A 29-year-old man presented with agitation, aggression, seizures, tachycardia, hypertension, tachypnea, and fever after the intravenous injection of 3 mL of 25I-NBOMe (unknown concentration). Laboratory results revealed respiratory and metabolic acidosis, elevation of creatine kinase, impaired renal function, and elevated alanine transaminase. A CT scan of the head showed mild cerebral edema. Despite supportive care, he developed anuria with acute kidney injury within the next 2 days and he underwent a continuous veno-venous hemofiltration (CVVH). His condition deteriorated later with fever, high-oxygen and vasopressor requirements and worsening renal impairment. Acute lung injury and pulmonary abscesses were observed in chest radiographs. Following a complicated hospital course and aggressive supportive care, his condition gradually normalized and he was discharged home 43 days after presentation (Hill et al, 2013).
    2) A 21-year-old man developed agitation, hallucinations, tachycardia, and fever after insufflating about 0.1 g of 25I-NBOMe powder. He recovered gradually following supportive care (Hill et al, 2013).
    d) CASE REPORT (25I-NBOMe): An 18-year-old man presented with severe agitation and hallucinations after using 25I-NBOMe (a (n-benzyl) phenethylamine in the 2C family) and then jumping out of a moving car. On admission, he had tachycardia (heart rate 150 to 160 beats/min) and hypertension (BP 150-170 mmHg systolic and 110 mm Hg diastolic). Laboratory results revealed hypokalemia (potassium 2.8 mEq/L; 3.6 to 5.2 mEq/L). Following supportive care, his symptoms gradually improved; however, he continued to have episodes of aggressiveness for the next 24 hours (Rose et al, 2013).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) HYPERVENTILATION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT (2C-I-NBOMe): A 17-year-old man developed hyperventilation and foaming at the mouth, and died after ingesting an unknown amount of 2C-I-NBOMe mixed with chocolate (Dean et al, 2013).
    B) ACUTE LUNG INJURY
    1) WITH POISONING/EXPOSURE
    a) Pulmonary edema may occur in severe intoxications (Curtis et al, 2003).
    b) CASE REPORT (2C-T-7): A 20-year-old man developed vomiting, hallucinations, agitation, violence/aggression, nasal bleeding, seizures, pulmonary edema and cardiopulmonary arrest, and died after insufflating 35 mg of 2C-T-7 (Curtis et al, 2003; Dean et al, 2013).
    C) COUGH
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, cough was reported in 3 (9%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    D) DYSPNEA
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, difficulty breathing was reported in 3 (9%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) PSYCHOMOTOR AGITATION
    1) WITH POISONING/EXPOSURE
    a) Agitation has been reported following the use of 2C-T-7 (Curtis et al, 2003).
    b) CASE SERIES (25I-NBOMe): Agitation developed in 6 of 7 patients after using 25I-NBOMe (Hill et al, 2013).
    1) A 21-year-old man developed agitation, hallucinations, tachycardia, and fever after insufflating about 0.1 g of 25I-NBOMe powder. He recovered gradually following supportive care (Hill et al, 2013).
    c) CASE REPORT (2C-T-7): A 20-year-old man developed vomiting, hallucinations, agitation, violence/aggression, nasal bleeding, seizures, pulmonary edema and cardiopulmonary arrest, and died after insufflating 35 mg of 2C-T-7 (Curtis et al, 2003; Dean et al, 2013).
    d) CASE REPORT 2C-T-7: A 17-year-old man developed vomiting, agitation, violence/aggression, possible hyperthermia, rigidity and cardiopulmonary arrest, and died after insufflating 30 to 35 mg of 2C-T-7 (Dean et al, 2013).
    e) CASE REPORT (2C-T-7): A man developed agitation, aggression, seizures, hallucinations, cardiopulmonary arrest and cerebral hemorrhage, and died after using an unknown amount of 2C-T-7 and 200 mg of MDMA (Dean et al, 2013).
    f) CASE REPORT (2C-E): A 19-year-old man developed agitation, aggression, hyperthermia, disseminated intravascular coagulation (DIC) and multi-organ failure, and died after insufflating an unknown amount of 2C-E (Dean et al, 2013).
    B) COMA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT (2C-T-21): A 22-year-old man developed hyperthermia (108 degrees C), seizures and coma, and died after ingesting an unknown amount (dipping his tip of tongue into the powder) of 2C-T-21 (Dean et al, 2013).
    C) CEREBRAL HEMORRHAGE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT (2C-T-7): A man developed agitation, aggression, seizures, hallucinations, cardiopulmonary arrest and cerebral hemorrhage, and died after using an unknown amount of 2C-T-7 and 200 mg of MDMA (Dean et al, 2013).
    b) COMBINED INGESTION
    1) CASE REPORT (MDA and 2C-I): A 39-year-old woman presented with rapidly diminishing mental status, agitation, hypothermia, vomiting, urinary incontinence, severe hypertension, vasoconstriction and extensor posturing after ingesting MDA and 2C-I. Urinary drug concentrations were MDA 5.56 mg/L and 2C-I 0.31 mg/L. A head CT scan 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).
    D) CEREBRAL INFARCTION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT (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).
    E) SEIZURE
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT (2C-T-7): A 20-year-old man developed vomiting, hallucinations, agitation, violence/aggression, nasal bleeding, seizures, pulmonary edema and cardiopulmonary arrest, and died after insufflating 35 mg of 2C-T-7 (Curtis et al, 2003; Dean et al, 2013).
    b) CASE REPORT (2C-T-7): A man developed agitation, aggression, seizures, hallucinations, cardiopulmonary arrest and cerebral hemorrhage, and died after using an unknown amount of 2C-T-7 and 200 mg of MDMA (Dean et al, 2013).
    c) CASE REPORT (2C-T-21): A 22-year-old man developed hyperthermia (108 degrees C), seizures and coma, and died after ingesting an unknown amount (dipping his tip of tongue into the powder) of 2C-T-21 (Dean et al, 2013).
    d) CASE REPORT (2C-I): A 19-year-old man developed recurrent generalized tonic-clonic seizures, serotonin syndrome, and prolonged respiratory failure after insufflating a liquid called "liquid acid". Following supportive care, he recovered and was discharged home on day 6. A comprehensive urine test from the ICU revealed the presence of 2C-I (Bosak et al, 2013)
    e) CASE SERIES (25I-NBOMe): Seizures developed in 3 of 7 patients after using 25I-NBOMe (Hill et al, 2013).
    F) HEADACHE
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, headache was reported in 1 (3%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    G) PARESTHESIA
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, paresthesia was reported in 1 (3%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    H) DIZZINESS
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, dizziness was reported in 2 (6%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    I) TREMOR
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, trembling was reported in 9 (26%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    J) LEUKOENCEPHALOPATHY
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT (2C-E): A 26-year-old man was found unconscious with an empty bottle of zolpidem. Despite naloxone therapy, his condition did not improve. Physical examination revealed no focal neurological deficits and intact brainstem reflexes. The lumbar puncture results and a brain CT scan were normal. An initial urine toxicology screen was positive for benzodiazepines and tetrahydrocannabinol. It was found later that he also used 2C-E. Another urine test revealed the presence of 2C-E. A brain MRI revealed large symmetric areas of restricted diffusion within the subcortical white matter, genu, splenium of the corpus callosum, and minimal restricted diffusion in the bilateral globus pallidi, but sparing of the striatum, cortices, and hippocampus, suggesting toxic leukoencephalopathy. Despite supportive care, his condition did not improve and he died after withdrawal of life support (Sacks et al, 2012).
    K) CLONUS
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES (25I-NBOMe): Clonus developed in 2 of 7 patients after using 25I-NBOMe (Hill et al, 2013).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) NAUSEA AND VOMITING
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, nausea was reported in 5 (14%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    b) CASE REPORT (2C-T-7): A 20-year-old man developed vomiting, hallucinations, agitation, violence/aggression, nasal bleeding, seizures, pulmonary edema and cardiopulmonary arrest, and died after insufflating 35 mg of 2C-T-7 (Curtis et al, 2003; Dean et al, 2013).
    c) CASE REPORT 2C-T-7: A 17-year-old man developed vomiting, agitation, violence/aggression, possible hyperthermia, rigidity and cardiopulmonary arrest, and died after insufflating 30 to 35 mg of 2C-T-7 (Dean et al, 2013).
    B) DIARRHEA
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, diarrhea was reported in 3 (9%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    C) ABDOMINAL PAIN
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, abdominal pain was reported in 5 (14%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) ACUTE RENAL FAILURE SYNDROME
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES (25I-NBOMe): Acute renal injury developed in 1 of 7 patients after using 25I-NBOMe (Hill et al, 2013).
    b) A 29-year-old man presented with agitation, aggression, seizures, tachycardia, hypertension, tachypnea, and fever after the intravenous injection of 3 mL of 25I-NBOMe (unknown concentration). Laboratory results revealed respiratory and metabolic acidosis, elevation of creatine kinase, impaired renal function, and elevated alanine transaminase. A CT scan of the head showed mild cerebral edema. Despite supportive care, he developed anuria with acute kidney injury within the next 2 days and he underwent continuous veno-venous hemofiltration (CVVH). His condition deteriorated later with fever, high-oxygen and vasopressor requirements and worsening renal impairment. Acute lung injury and pulmonary abscesses were observed in chest radiographs. He began producing urine 27 days after admission. Following a complicated hospital course and aggressive supportive care, his condition gradually normalized and he was discharged home 43 days after presentation (Hill et al, 2013).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES (25I-NBOMe): Metabolic acidosis developed in 3 of 7 patients after using 25I-NBOMe (Hill et al, 2013).
    1) A 29-year-old man presented with agitation, aggression, seizures, tachycardia, hypertension, tachypnea, and fever after the intravenous injection of 3 mL of 25I-NBOMe (unknown concentration). Laboratory results revealed respiratory and metabolic acidosis, elevation of creatine kinase, impaired renal function, and elevated alanine transaminase. A CT scan of the head showed mild cerebral edema. Despite supportive care, he developed anuria with acute kidney injury within the next 2 days and he underwent a continuous veno-venous hemofiltration (CVVH). His condition deteriorated later with fever, high-oxygen and vasopressor requirements and worsening renal impairment. Acute lung injury and pulmonary abscesses were observed in chest radiographs. Following a complicated hospital course and aggressive supportive care, his condition gradually normalized and he was discharged home 43 days after presentation (Hill et al, 2013).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) DISSEMINATED INTRAVASCULAR COAGULATION
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT (2C-E): A 19-year-old man developed agitation, aggression, hyperthermia, disseminated intravascular coagulation (DIC) and multi-organ failure, and died after insufflating an unknown amount of 2C-E (Dean et al, 2013).
    B) LEUKOCYTOSIS
    1) WITH POISONING/EXPOSURE
    a) CASE SERIES (25I-NBOMe): Elevated WBC developed in 2 of 7 patients after using 25I-NBOMe (Hill et al, 2013).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) EXCESSIVE SWEATING
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, sweating was reported in 8 (23%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    b) Pallor, diaphoresis, and piloerection may be present (Watson et al, 1993).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) PAIN
    1) WITH POISONING/EXPOSURE
    a) 2C-B: In a study of 2C-B use in Spain, muscle or joint pain was reported in 2 (6%) of 35 2C-B users (doses: 20 to 60 mg); however, 29 (83%) patients used 2C-B in combination with other drugs (eg, MDMA, ethanol, cannabis) (Caudevilla-Galligo et al, 2012).
    B) RHABDOMYOLYSIS
    1) WITH POISONING/EXPOSURE
    a) Rhabdomyolysis is a common complication in patients who develop hyperthermia, or prolonged seizures, coma, or muscular hyperactivity (Watson et al, 1993).
    b) CASE SERIES (25I-NBOMe): Elevated creatine kinase developed in 7 patients after using 25I-NBOMe (Hill et al, 2013).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Plasma concentrations are not clinically useful or readily available.
    B) Standard urine toxicology screen may not produce a positive result for many 2C agents.
    C) Monitor vital signs and mental status.
    D) Monitor serum electrolytes and renal function.
    E) Obtain ECG and institute continuous cardiac monitoring in patients with moderate to severe toxicity (ie, agitation, delirium, seizures, coma, hypotension).
    F) Monitor creatinine phosphokinase in patients with prolonged agitation, seizures or coma; monitor renal function and urine output in patients with rhabdomyolysis.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) Plasma concentrations are not clinically useful or readily available.
    2) Monitor serum electrolytes and renal function.
    3) Monitor creatinine phosphokinase in patients with prolonged agitation, seizures or coma; monitor renal function and urine output in patients with rhabdomyolysis.
    4.1.3) URINE
    A) TOXICOLOGY SCREEN
    1) Standard urine toxicology screen may not produce a positive result for many 2C agents.
    2) 2C-B
    a) It is difficult to detect 2C-B in urine with commercially available immunoassays because of the low cross-reactivity of 2C-B (Giroud et al, 1998).
    3) 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. However, the substance can be detected by gas-chromatography-mass spectrometry (Miyajima et al, 2008).
    4.1.4) OTHER
    A) OTHER
    1) Obtain ECG and institute continuous cardiac monitoring in patients with moderate to severe toxicity (ie, agitation, delirium, seizures, coma, hypotension).

Radiographic Studies

    A) MRI
    1) MRI can be used to diagnose leukoencephalopathy and should be considered in patients with prolonged depressed mental status after 2C series drug overdose.
    2) CASE REPORT (2C-E): A 26-year-old man was found unconscious with an empty bottle of zolpidem. Despite naloxone therapy, his condition did not improve. Physical examination revealed no focal neurological deficits and intact brainstem reflexes. The lumbar puncture results and a brain CT scan were normal. An initial urine toxicology screen was positive for benzodiazepines and tetrahydrocannabinol. It was found later that he also used 2C-E. Another urine test revealed the presence of 2C-E. A brain MRI revealed large symmetric areas of restricted diffusion within the subcortical white matter, genu, splenium of the corpus callosum, and minimal restricted diffusion in the bilateral globus pallidi, but sparing of the striatum, cortices, and hippocampus, suggesting toxic leukoencephalopathy. Despite supportive care, his condition did not improve and he died after withdrawal of life support (Sacks et al, 2012).

Methods

    A) MULTIPLE ANALYTICAL METHODS
    1) STANDARD TOXICOLOGY SCREENING
    a) Standard urine toxicology screen may not produce a positive result for many 2C agents.
    b) 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.
    c) Several analytical techniques (gas chromatography-mass spectrometry [GC-MS], high-performance liquid chromatography-diode-array detection [HPLC-DAD], capillary electrophoresis-diode-array detection [CE-DAD], Fourier-transform infrared spectroscopy [FTIR], and nuclear magnetic resonance spectroscopy [NMR]) identified 2C-B in illicit tablets. A combination of MS and NMR analysis was the only method that achieved the unequivocal identification of 2C-B. HPLC-DAD and CE-DAD were used to quantitate 2C-B in tablets. It was determined that 3 to 8 mg of 2C-B were the minimum quantity required to induce the effects characteristic of 2C-B (Giroud et al, 1998).
    2) PRESUMPTIVE TESTS
    a) 2C-B produced a positive color reaction (yellow turning green) with the Marquis reagent. Since the same color is produced for DOB (2,5-dimethoxy-4-bromoamphetamine), further analysis should be performed to identify 2C-B (Cole et al, 2002).
    3) IMMUNOASSAYS
    a) It is difficult to detect 2C-B in urine with commercially available immunoassays because of the low cross-reactivity of 2C-B (Giroud et al, 1998).
    b) In one study, the cross-reactivity of 9 commercial enzyme-linked immunosorbent assays (ELISAs) was evaluated using 11 designer drugs (2C-B; 2C-H, 2C-I, 2C-T-2, 2C-T-4, 2C-T-7, DOB, DOET, DOI, DOM, and 4-MTA). 4-MTA was the only substance with any measurable cross-reactivity. Using commercial amphetamine, methamphetamine, or MDMA assays, cross-reactivities were less than 0.4% for 2C, 2C-T, and DO series. Concentrations up to 50,000 ng/mL (generally higher than case samples) were not adequate to produce a positive result. Although these assays are very effective for methamphetamine, MDMA, and amphetamine, they cannot reliably identify other designer drugs such as 2C series, even after very high concentrations are used (Kerrigan et al, 2011).
    4) GAS CHROMATOGRAPHY MASS SPECTROMETRY
    a) 2C-B
    1) GC-MS was used to determine phenethylamine brominated in the meta or para position with respect to the ethylamine chain; however, it was not useful in discriminating between the two. Therefore, it could not accurately identify 2C-B (Cole et al, 2002).
    2) Mass spectroscopy was used to identify 2C-B in street tables sold in Switzerland as Ecstasy (Giroud et al, 1998).
    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) 2C-T-7 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 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).
    5) HIGH PERFORMANCE LIQUID CHROMATOGRAPHY
    a) HPLC determined 3 to 8 mg of 2C-B per tablet obtained from various sources (Cole et al, 2002).
    b) 25I-NBOMe: In a case report of a man who used 25I-NBOMe (a (n-benzyl) phenethylamine in the 2C family), high performance liquid chromatography with tandem mass spectrometry (HPLC/MS/MS) analysis was used to confirm the presence of 25I-NBOMe in serum (Rose et al, 2013).
    6) INFRARED AND NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY
    a) 2C-B was identified in the infrared (IR) spectrum, using the standard 2C-B and street sample. However, another method should also be used because of the differences in the degree of noise in the spectra. Nuclear magnetic resonance (NMR) spectra, mainly 2D method was used to identify 2C-B, including positional and stereo isomers. It showed the two aromatic protons in the para position and other functional groups of 2C-B (Cole et al, 2002).
    b) IR and NMR spectroscopy methods were used to identify 2C-B in street tablets sold in Switzerland as Ecstasy (Giroud et al, 1998).
    7) LIQUID CHROMATOGRAPHY TIME OF FLIGHT MASS SPECTROMETRY
    a) CASE REPORT: In a case report of a man who insufflated a liquid called "liquid acid", a comprehensive urine test using high-performance liquid chromatography/time of flight mass-spectrometry (LC/TOF-MS) revealed the presence of 2C-I (Bosak et al, 2013)
    8) LIQUID CHROMATOGRAPHY TANDEM MASS SPECTROMETRY
    a) The following agents can be detected using LC-MS/MS (liquid chromatography tandem mass spectrometry) (Wohlfarth et al, 2010; Hill et al, 2013):
    1) 2C-B: (2-(4-bromo-2,5-dimethoxyphenyl)ethanamine)
    2) 2C-D: (2-(2,5 dimethoxy-4-methylphenyl)ethanamine)
    3) 2C-H: (2-(2,5-dimethoxyphenyl)ethanamine)
    4) 2C-I: (2-(4-iodo-2,5-dimethoxyphenyl)ethanamine)
    5) 2C-P: (2-2,5-diemethoxy-4-prophylphenyl)ethanamine)
    6) 2C-T-2: (2-[4-(ethylthio)-2,5-dimethoxyphenyl]ethanamine)
    7) 2C-T-4:(2-[4-(isopropylthio)-2,5-dimethoxyphenyl]ethanamine)
    8) 2C-T-7: (2-[2,5-dimethoxy-40(propylthio)phenyl]ethanamine)
    9) 25I-NBOMe: 2-(4-iodo-2,5-dimethoxyphenyl)-N-[(2-methoxyphenyl)methyl]ethanamine
    b) A 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) LC-MS/MS was successfully used to identify 25I-NBOMe in the plasma of 7 patients after using 25I-NBOMe (Hill et al, 2013).
    d) In one study, liquid chromatography-atomospheric pressure ionization electrospray mass spectrometry (LC-MS) was used to determine MDMA, 2C-D, 2C-B, 2C-B-Fly, 2C-T-2, 2C-I, 2C-E, m-CPP, 4-OH-DIPT, and 4-acetoxy-DIPT in urine of 32 consumers using MDPA as internal standard. Absolute analytical mean recoveries after extraction procedure for the 3 different quality control concentration samples (3000 ng/mL, 500 ng/mL, and 75 ng/mL) were as follows: 2C-D (83.6%, 85.3%, 82.6%, respectively), 2C-B (85.2%, 80.9%, 82.8%, respectively), 2C-B-Fly (84.9%, 83%, 82.9%, respectively), 2C-T-2 (72.7%, 71.8%, 73.5%, respectively), 2C-I (76.7%, 74.5%, 76.2%, respectively), 2C-E (92.6%, 90.5%, 91.2%, respectively), MDMA (73.8%, 79.7%, 75.8%, respectively), m-CPP (95.6%, 91.8%, 93.5%, respectively), 4-OH-DIPT (59.3%, 55.4%, 56.1%, respectively), and 4-acetoxy-DIPT (75.3%, 72.7%, 73.9%, respectively). Limits of quantification were between 20 and 60 ng/mL (Pichini et al, 2008).
    e) 2C-E: A 26-year-old man developed toxic leukoencephalopathy after using 2C-E. A urine toxicology screen using LC-MS/MS (liquid chromatography tandem mass spectrometry) was positive for 2C-E. Despite supportive care, his condition did not improve and he died after withdrawal of life support (Van Vrancken et al, 2013; Sacks et al, 2012).
    9) ULTRAVIOLET SPECTROSCOPY
    a) Ultraviolet (UV) spectroscopy method was used to identify 2C-B in street tables sold in Switzerland as Ecstasy (Giroud et al, 1998).

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 (ie, hallucinations, somnolence, delirium, coma), 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, if a responsible adult is present.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Consult a poison center or medical toxicologist for assistance in managing patients with severe toxicity (ie, seizures, dysrhythmias, severe delirium, coma), or in whom the diagnosis is not clear. Refer for substance abuse counseling as appropriate.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Patients with deliberate self-harm ingestions, children with any ingestion, and symptomatic patients should be sent to a healthcare facility for evaluation, treatment and observation for 6 to 8 hours.

Monitoring

    A) Plasma concentrations are not clinically useful or readily available.
    B) Standard urine toxicology screen may not produce a positive result for many 2C agents.
    C) Monitor vital signs and mental status.
    D) Monitor serum electrolytes and renal function.
    E) Obtain ECG and institute continuous cardiac monitoring in patients with moderate to severe toxicity (ie, agitation, delirium, seizures, coma, hypotension).
    F) Monitor creatinine phosphokinase in patients with prolonged agitation, seizures or coma; monitor renal function and urine output in patients with rhabdomyolysis.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) Prehospital gastrointestinal decontamination is not recommended because of the potential for agitation and seizures and subsequent aspiration.
    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. Administer activated charcoal if recent ingestion or possibility of coingestants, and only if the patient is cooperative and can protect their airway or is intubated. Administer to body stuffers that are not at risk for aspiration.
    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) MONITORING OF PATIENT
    1) Plasma levels are not clinically useful or readily available.
    2) Standard urine toxicology screen may not produce a positive result for many 2C agents.
    3) Monitor vital signs and mental status.
    4) Monitor serum electrolytes and renal function.
    5) Obtain ECG and institute continuous cardiac monitoring in patients with moderate to severe toxicity (ie, agitation, delirium, seizures, coma, hypotension).
    6) Monitor creatinine phosphokinase in patients with prolonged agitation, seizures or coma; monitor renal function and urine output in patients with rhabdomyolysis.
    B) 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) (Grinspoon & Hedblom, 1975; Snyder et al, 1970; Solursh & Clement, 1968).
    6) NEUROLEPTICS
    a) ZIPRASIDONE: Oral ziprasidone was used to treat episodes of aggressiveness in an 18-year-old man who used 25I-NBOMe (Rose et al, 2013). However, neuroleptic agents can cause QT prolongation and increase the risk of developing ventricular dysrhythmias. Neuroleptics should be avoided in patients with known QT prolongation (Dean et al, 2013).
    b) HALOPERIDOL: 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). 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.
    c) CASE REPORT: A 40-year-old man without a history of a mental disorder, presented in a delusional state with incoherent speech after ingesting a sexual enhancing product containing 2C-T-4. Six hours after ingestion, the patient was overcome with extreme fear which prompted emergency treatment. The patient was treated with neuroleptic medication and recovered without neurological deficit approximately 17 hours after ingestion (Miyajima et al, 2008).
    7) KETAMINE
    a) Since benzodiazepines and neuroleptics have a slow onset of action via the IM route, ketamine has been suggested as an alternative chemical restraint in patients with excited delirium. Ketamine has a rapid onset, predictable ability to induce a dissociated state, and lack of cardiovascular or respiratory depression. However, airway compromise, including laryngospasm and hypoxia may occur following ketamine therapy. In addition, ketamine can cause stimulatory cardiovascular effects resulting in increased tachycardia and hypertension (Dean et al, 2013).
    8) PHENOBARBITAL
    a) Phenobarbital has been protective against lethality in mice but there are no human data (Thiessen & Cook, 1973b) (David & Borne, 1984) (Paton et al, 1975).
    C) 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) 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).
    D) 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).
    E) HYPERTENSIVE EPISODE
    1) SUMMARY: Hypertension often resolves once the patient is less agitated (ie, following benzodiazepine administration). If severe hypertension associated with end organ damage 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.
    F) BODY TEMPERATURE ABOVE REFERENCE RANGE
    1) Hyperthermia occurs in severe cases (Dean et al, 2013; Drees et al, 2009). It may be life threatening, and has been contributory to death (Dean et al, 2013). It should be treated aggressively.
    2) Core temperature above 40 degrees C may be life threatening and indicates the need for rapid sedation and cooling.
    3) External cooling measures should be provided by accelerating evaporative heat loss by keeping the patient's skin wet with cool water and placing fans in the room. Rapid cooling measures including placing the patient in an ice bath may be required. Monitor temperature every continuously by rectal probe, or every 30 minutes until below 38 degrees centigrade.
    4) Administer intravenous benzodiazepines. Large doses may be required. Monitor respiratory adequacy and airway. Be prepared to intubate and ventilate if needed.
    5) Intubation with neuromuscular paralysis and sedation may be required in severe cases.
    G) 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).
    H) 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).
    I) 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).
    J) VASOSPASM
    1) Heparinization and intravenous or intraarterial nitroprusside have been used with success (Bowen et al, 1983).
    K) 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).

Enhanced Elimination

    A) LACK OF EFFECT
    1) Hemodialysis and hemoperfusion are not of value because of the large volume of distribution.

Case Reports

    A) ADULT
    1) COMBINED INGESTION (MDA and 2C-I)
    a) A 39-year-old woman presented with rapidly diminishing mental status, agitation, hypothermia, vomiting, urinary incontinence, severe hypertension, vasoconstriction and exterior posturing after ingesting MDA and 2C-I. Drug concentrations measured in the urine were: MDA 5.56 mg/L and 2C-I 0.31mg/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).

Summary

    A) TOXICITY: Severe toxicity has developed following recreational doses in some individuals. A man developed agitation, hallucinations, tachycardia, and fever after insufflating about 0.1 g of 25I-NBOMe powder. He recovered following supportive care. FATALITIES: Two men died after insufflating 30 to 35 mg of 2C-T-7. Fatalities have also been reported following the use of 2C-E, 2C-T-21, and 2C-I-NBOMe (unknown amounts).
    B) TYPICAL DOSES: 2C-B: 12 to 24 mg; 2C-B doses up to 100 mg have also been used and tolerated in some cases. 2C-B powder form doses for insufflation are usually one third of the oral dose. 2C-C: 20 to 40 mg; 2C-D: 20 to 60 mg; 2C-E: 10 to 25 mg; 2C-G: 20 to 35 mg; 2C-G-3: 16 to 25 mg; 2C-G-5: 10 to 16 mg; 2C-I: 14 to 22 mg; 2C-N: 100 to 150 mg; 2C-P: 6 to 10 mg; 2C-SE: approximately 100 mg; 2C-T: 60 to 100 mg; 2C-T-2: 12 to 25 mg; 2C-T-4: 8 to 20 mg; 2C-T-7: 10 to 30 mg; 2C-T-8: 30 to 50 mg; 2C-T-9: 60 to 100 mg; 2C-T-13: 25 to 40 mg; 2C-T-15: greater than 30 mg; 2C-T-17: 60 to 100 mg; 2C-T-21: 8 to 12 mg.

Minimum Lethal Exposure

    A) 2C-T-7
    1) CASE REPORT: A 20-year-old man developed vomiting, hallucinations, agitation, violence/aggression, nasal bleeding, seizures, pulmonary edema and cardiopulmonary arrest, and 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; liver 854 ng/g (Curtis et al, 2003).
    2) CASE REPORT: A 17-year-old man developed vomiting, agitation, violence/aggression, possible hyperthermia, rigidity, and cardiopulmonary arrest, and died after insufflating 30 to 35 mg of 2C-T-7 (Dean et al, 2013).
    B) 2C-T-21
    1) CASE REPORT: A 22-year-old man developed hyperthermia (108 degrees F), seizures and coma, and died after ingesting an unknown amount (dipping his tip of tongue into the powder) of 2C-T-21 (Dean et al, 2013).
    C) Fatalities have also been reported following the use of 2C-E and 2C-I-NBOMe (unknown amounts) (Dean et al, 2013).

Maximum Tolerated Exposure

    A) TYPICAL RECREATIONAL DOSES
    1) Typical doses are listed below (Dean et al, 2013):
    1) 2C-B: 12 to 24 mg
    2) 2C-C: 20 to 40 mg
    3) 2C-D: 20 to 60 mg
    4) 2C-E: 10 to 25 mg
    5) 2C-G: 20 to 35 mg
    6) 2C-G-3: 16 to 25 mg
    7) 2C-G-5: 10 to 16 mg
    8) 2C-I: 14 to 22 mg
    9) 2C-N: 100 to 150 mg
    10) 2C-P: 6 to 10 mg
    11) 2C-SE: approximately 100 mg
    12) 2C-T: 60 to 100 mg
    13) 2C-T-2: 12 to 25 mg
    14) 2C-T-4: 8 to 20 mg
    15) 2C-T-7: 10 to 30 mg
    16) 2C-T-8: 30 to 50 mg
    17) 2C-T-9: 60 to 100 mg
    18) 2C-T-13: 25 to 40 mg
    19) 2C-T-15: greater than 30 mg
    20) 2C-T-17: 60 to 100 mg
    21) 2C-T-21: 8 to 12 mg
    2) 2C-B doses up to 100 mg have also been used and tolerated (Hill & Thomas, 2011; Cole et al, 2002). 2C-B powder form doses for insufflation are usually one third of the oral dose (Cole et al, 2002).
    B) 2C-B
    1) 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 tablet (available as 5 mg tablets) 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).
    C) 25I-NBOMe
    1) CASE REPORT: A 21-year-old man developed agitation, hallucinations, tachycardia, and fever after insufflating about 0.1 g of 25I-NBOMe powder. He recovered gradually following supportive care (Hill et al, 2013).
    2) CASE REPORT: A 29-year-old man presented with agitation, aggression, seizures, tachycardia, hypertension, tachypnea, and fever after the intravenous injection of 3 mL of 25I-NBOMe (unknown concentration). Laboratory results revealed respiratory and metabolic acidosis, elevation of creatine kinase, impaired renal function, and elevated alanine transaminase. A CT scan of the head showed mild cerebral edema. Despite supportive care, he developed anuria with acute kidney injury within the next 2 days and he underwent a continuous veno-venous hemofiltration (CVVH). His condition deteriorated later with fever, high-oxygen and vasopressor requirements and worsening renal impairment. ARDS and pulmonary abscesses were observed in chest radiographs. Following a complicated hospital course and aggressive supportive care, his condition gradually normalized and he was discharged home 43 days after presentation (Hill et al, 2013).

Serum Plasma Blood Concentrations

    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) 2C-T-7
    a) 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).
    2) 25I-NBOMe
    a) In a case report of a man who used 25I-NBOMe (a (n-benzyl) phenethylamine in the 2C family), concentration of 25I-NBOMe in patient's serum was 0.76 ng/mL using high performance liquid chromatography with tandem mass spectrometry (HPLC/MS/MS) analysis. He recovered following supportive care (Rose et al, 2013).

Toxicologic Mechanism

    A) The 2C series are a large group of chemicals characterized by methoxy groups at positions 2 and 5 of the benzene ring. They differ from D series of substituted amphetamines 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). 2C compounds have an affinity for 5-HT2 and alpha-adrenergic receptors, and they can be either agonists or antagonists. 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 (Dean et al, 2013; Hill & Thomas, 2011; Giroud et al, 1998).

General Bibliography

    1) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    2) Ambrose JB, Bennett HD, Lee HS, et al: Cerebral vasculopathy after 4-bromo-2,5-dimethoxyphenethylamine ingestion. Neurologist 2010; 16(3):199-202.
    3) 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.
    4) Appleton R: Lorazepam vs diazepam in the acute treatment of epileptic seizures and status epilepticus.. Dev Med Child Neuro 1995; 37:682-688.
    5) Bosak A , Lovecchio F , & Levine M : Recurrent Seizures and Serotonin Syndrome Following "2C-I" Ingestion. J Med Toxicol 2013; 9(2):196-198.
    6) Bowen JS, Davis GB, & Kearney TE: Diffuse vascular spasm associated with 4-bromo-2,5-dimethoxyamphetamine ingestion. JAMA 1983; 249:1477-1479.
    7) Boyer EW & Shannon M: The serotonin syndrome. N Eng J Med 2005; 352(11):1112-1120.
    8) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    9) 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.
    10) Bunchman TE, Lynch RE, & Wood EG: Intravenously administered labetalol for treatment of hypertension in children. J Pediatr 1992; 120(1):140-144.
    11) Camp NE: Drug- and toxin-induced Rhabdomyolysis. J Emerg Nurs 2009; 35(5):481-482.
    12) Caudevilla-Galligo F , Riba J , Ventura M , et al: 4-Bromo-2,5-dimethoxyphenethylamine (2C-B): presence in the recreational drug market in Spain, pattern of use and subjective effects. J Psychopharmacol 2012; 26(7):1026-1035.
    13) 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.
    14) 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.
    15) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    16) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    17) Claassen JAHR & Gelissen HPMM: The serotonin syndrome (letter). N Eng J Med 2005; 352(23):2455.
    18) Cole MD , Lea C , & Oxley N : 4-Bromo-2,5-dimethoxyphenethylamine (2C-B): a review of the public domain literature. Sci Justice 2002; 42(4):223-224.
    19) Criddle LM: Rhabdomyolysis. Pathophysiology, recognition, and management. Crit Care Nurse 2003; 23(6):14-22, 24-26, 28.
    20) 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.
    21) Davis WM & Borne RF: Pharmacologic investigation of compounds related to 3,4 methylenedioxyamphetamine. Subst Alcohol Actions Misuse 1984; 5:105-110.
    22) De Negri M & Baglietto MG: Treatment of status epilepticus in children. Paediatr Drugs 2001; 3(6):411-420.
    23) Deal JE , Barratt TM , & Dillon MJ : Management of hypertensive emergencies. Arch Dis Child 1992; 67(9):1089-1092.
    24) Dean BV, Stellpflug SJ, Burnett AM, et al: 2C or not 2C: phenethylamine designer drug review. J Med Toxicol 2013; 9(2):172-178.
    25) Drees JC , Stone JA , & Wu AH : Morbidity involving the hallucinogenic designer amines MDA and 2C-I. J Forensic Sci 2009; 54(6):1485-1487.
    26) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    27) 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.
    28) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    29) Fivush B , Neu A , & Furth S : Acute hypertensive crises in children: emergencies and urgencies. Curr Opin Pediatr 1997; 9(3):233-236.
    30) Flynn JT & Tullus K: Severe hypertension in children and adolescents: pathophysiology and treatment. Pediatr Nephrol 2009; 24(6):1101-1112.
    31) Giang DW & McBride MC : Lorazepam versus diazepam for the treatment of status epilepticus. Pediatr Neurol 1988; 4(6):358-361.
    32) Gillman PK: Ecstasy, serotonin syndrome and the treatment of hyperpyrexia (letter). MJA 1997; 167:109-111.
    33) Gillman PK: Successful treatment of serotonin syndrome with chlorpromazine (letter). MJA 1996; 165:345.
    34) Giroud C , Augsburger M , Rivier L , et al: 2C-B: a new psychoactive phenylethylamine recently discovered in Ecstasy tablets sold on the Swiss black market. J Anal Toxicol 1998; 22(5):345-354.
    35) Goldberg RJ & Huk M: Serotonin syndrome from trazodone and buspirone (letter). Psychosomatics 1992; 33:235-236.
    36) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    37) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    38) Graham PM: Successful treatment of the toxic serotonin syndrome with chlorpromazine (letter). Med J Australia 1997; 166:166-167.
    39) Grinspoon L & Hedblom P: The speed culture: amphetamine use and abuse in America, Harvard University Press, Cambridge, MA, 1975, pp 40-61.
    40) Hari P & Sinha A: Hypertensive emergencies in children. Indian J Pediatr 2011; 78(5):569-575.
    41) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    42) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    43) Hill SL & Thomas SH: Clinical toxicology of newer recreational drugs. Clin Toxicol (Phila) 2011; 49(8):705-719.
    44) Hill SL , Doris T , Gurung S , et al: Severe clinical toxicity associated with analytically confirmed recreational use of 25I-NBOMe: case series. Clin Toxicol (Phila) 2013; 51(6):487-492.
    45) Homsi E, Barreiro MF, Orlando JM, et al: Prophylaxis of acute renal failure in patients with rhabdomyolysis. Ren Fail 1997; 19(2):283-288.
    46) Huang HH & Bai YM: Persistent psychosis after ingestion of a single tablet of '2C-B'. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35(1):293-294.
    47) Huerta-Alardin AL, Varon J, & Marik PE: Bench-to-bedside review: Rhabdomyolysis -- an overview for clinicians. Crit Care 2005; 9(2):158-169.
    48) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    49) Kerrigan S , Mellon MB , Banuelos S , et al: Evaluation of commercial enzyme-linked immunosorbent assays to identify psychedelic phenethylamines. J Anal Toxicol 2011; 35(7):444-451.
    50) 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.
    51) Kline SS, Mauro LS, & Scala-Barnett DM: Serotonin syndrome versus neuroleptic malignant syndrome as a cause of death. Clin Pharmac 1989; 8:510-514.
    52) Koch-Weser J: Hypertensive emergencies. N Engl J Med 1974; 290:211.
    53) 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.
    54) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    55) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    56) 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.
    57) Meyer MR & Maurer HH: Metabolism of designer drugs of abuse: an updated review. Curr Drug Metab 2010; 11(5):468-482.
    58) 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.
    59) Mills KC: Serotonin syndrome: a clinical update. Med Toxicol 1997; 13:763-783.
    60) Mitchell WG: Status epilepticus and acute repetitive seizures in children, adolescents, and young adults: etiology, outcome, and treatment. Epilepsia 1996; 37(S1):S74-S80.
    61) Miyajima M, Matsumoto T, & Ito S: 2C-T-4 intoxication: acute psychosis caused by a designer drug. Psychiatry Clin Neurosci 2008; 62(2):243-.
    62) 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.
    63) 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.
    64) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    65) 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.
    66) 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.
    67) 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.
    68) Pichini S , Pujadas M , Marchei E , et al: Liquid chromatography-atmospheric pressure ionization electrospray mass spectrometry determination of "hallucinogenic designer drugs" in urine of consumers. J Pharm Biomed Anal 2008; 47(2):335-342.
    69) Polderman KH: Acute renal failure and rhabdomyolysis. Int J Artif Organs 2004; 27(12):1030-1033.
    70) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    71) Product Information: ATIVAN(R) injection, lorazepam injection. Baxter Healthcare Corporation, Deerfield, IL, 2003.
    72) Product Information: BREVIBLOC(TM) intravenous injection, esmolol HCl intravenous injection. Baxter Healthcare Corporation (per FDA), Deerfield, IL, 2012.
    73) Product Information: Cordarone(R) oral tablets, amiodarone HCl oral tablets. Wyeth Pharmaceuticals Inc (per FDA), Philadelphia, PA, 2015.
    74) Product Information: Lidocaine HCl intravenous injection solution, lidocaine HCl intravenous injection solution. Hospira (per manufacturer), Lake Forest, IL, 2006.
    75) Product Information: NITROPRESS(R) injection for IV infusion, Sodium Nitroprusside injection for IV infusion. Hospira, Inc., Lake Forest, IL, 2007.
    76) Product Information: NITROPRESS(R) injection, sodium nitroprusside injection. Hospira,Inc, Lake Forest, IL, 2004.
    77) Product Information: Phentolamine Mesylate IM, IV injection Sandoz Standard, phentolamine mesylate IM, IV injection Sandoz Standard. Sandoz Canada (per manufacturer), Boucherville, QC, 2005.
    78) Product Information: Trandate(R) IV injection, labetalol hydrochloride IV injection. Prometheus Laboratories Inc., San Diego, CA, 2010.
    79) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    80) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    81) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    82) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    83) 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.
    84) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    85) Rhoney D & Peacock WF: Intravenous therapy for hypertensive emergencies, part 1. Am J Health Syst Pharm 2009; 66(15):1343-1352.
    86) Rose SR, Poklis JL, & Poklis A: A case of 25I-NBOMe (25-I) intoxication: a new potent 5-HT2A agonist designer drug. Clin Toxicol (Phila) 2013; 51(3):174-177.
    87) Sacks J, Ray MJ, Williams S, et al: Fatal toxic leukoencephalopathy secondary to overdose of a new psychoactive designer drug 2C-E ("Europa"). Proc (Bayl Univ Med Cent) 2012; 25(4):374-376.
    88) 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.
    89) 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.
    90) Shannon M: Methylenedioxymethamphetamine (MDMA, "Ecstasy"). Ped Emerg Care 2000; 16:377-380.
    91) Singh D, Akingbola O, Yosypiv I, et al: Emergency management of hypertension in children. Int J Nephrol 2012; 2012:420247.
    92) 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.
    93) Solursh LP & Clement WR: Hallucinogenic drug abuse - manifestations and management. Can Med Assoc J 1968; 98:407-410.
    94) 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.
    95) Sternbach H: The serotonin syndrome. Am J Psychiatr 1991; 148:705-713.
    96) Temple ME & Nahata MC: Treatment of pediatric hypertension. Pharmacotherapy 2000; 20(2):140-150.
    97) 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.
    98) 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.
    99) Van Vrancken MJ, Benavides R, & Wians FH Jr: Identification of designer drug 2C-E (4-ethyl-2, 5-dimethoxy-phenethylamine) in urine following a drug overdose. Proc (Bayl Univ Med Cent) 2013; 26(1):58-61.
    100) Vanholder R, Sever MS, Erek E, et al: Rhabdomyolysis. J Am Soc Nephrol 2000; 11(8):1553-1561.
    101) Walter LA & Catenacci MH: Rhabdomyolysis. Hosp Physician 2008; 44(1):25-31.
    102) Watson JD, Ferguson C, & Hinds CJ: Exertional heat stroke induced by amphetamine analogues. Anaesthesia 1993; 48:1057-1060.
    103) Wheless JW : Treatment of status epilepticus in children. Pediatr Ann 2004; 33(6):376-383.
    104) 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.
    105) de Boer D & Bosman I: A new trend in drugs-of-abuse; the 2C-series of phenethylamine designer drugs. Pharm World Sci 2004; 26(2):110-113.
    106) de Boer D, Gijzels MJ, Bosman IJ, et al: More data about the new psychoactive drug 2C-B. J Anal Toxicol 1999; 23(3):227-228.
    107) 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.