MOBILE VIEW  | 

METHADONE AND RELATED AGENTS

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) This management deals with opioid agonists with a long duration of action used in the management of opiate withdrawal:
    1) Methadone hydrochloride is a synthetic opiate agonist with multiple actions. Qualitatively and quantitatively similar to the actions of morphine with the most prominent effect being on the central nervous system and smooth muscle.
    2) Levomethadyl acetate hydrochloride (or levo-alpha-acetyl-methadol known as LAAM) is a derivative of methadone (which it resembles chemically), but has a longer duration of action than methadone (up to 72 hours). It was discontinued in the United States in September 2003 and removed from the European market in March 2001 due to reports of severe cardiac adverse events.

Specific Substances

    A) METHADONE HYDROCHLORIDE
    1) Amidine Hydrochloride
    2) Amidone Hydrochloride
    3) (+/-)-Methadone Hydrochloride
    4) Methadoni Hydrochloridum
    5) Methadonum
    6) Phenadone
    7) Molecular Formula: C21-H27-N-O
    8) CAS 76-99-3 (methadone)
    9) CAS 297-88-1 (methadone, +/-)
    10) CAS 1095-90-5 (methadone hydrochloride)
    11) CAS 125-56-4 (methadone hydrochloride, +/-)
    LEVO ALPHA ACETYL METHADOL
    1) Levo-alpha-acetylmethadol
    2) L-alpha-acetyl-methadol-hydrochloride
    3) levo-alpha-6-dimethylamino-4
    4) 4-diphenyl-3-heptyl acetate hydrochloride
    5) Molecular Formula: C23-H31-N-O2
    6) CAS 1477-40-3 (levomethadyl)
    7) CAS 34433-66-4 (levomethadyl acetate)
    8) CAS 43033-72-3 (levomethadyl acetate hydrochloride)

Available Forms Sources

    A) FORMS
    1) Methadone hydrochloride is available as: 10 mg/mL injection solution; 5 mg/mL, 10 mg/5 mL, and 10 mg/mL oral solution; 5 mg, 10 mg, and 40 mg oral tablets; 40 mg oral tablet for suspension.
    2) The manufacturer discontinued the sale and distribution of levomethadyl hydrochloride acetate (ORLAAM(R)) in the United States in September 2003. ORLAAM(R) was removed from the European market in March 2001. This is due to reports of severe cardiac adverse events including QT interval prolongation, torsades de pointes, and cardiac arrest.
    B) SOURCES
    1) Widespread methadone treatment programs which rely on weekly home treatment have resulted in accidental childhood exposure due to improper methadone storage (Binchy et al, 1994).
    2) Several reports of methadone poisoning among children receiving antibiotic suspensions have occurred as a result of reconstitution errors in which methadone was inadvertently used (Gayle et al, 1991; Lalkin et al, 1999).
    C) USES
    1) Methadone is commonly used in drug treatment programs (both detoxification and maintenance treatment) for narcotic addiction (heroin or other morphine-like drugs) (Prod Info Methadone hydrochloride (Methadone HCL(R), 1996).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Methadone is used to treat chronic pain and to prevent symptoms of opioid withdrawal in opioid dependent patients. It is also rarely used as an antiperistaltic agent.
    B) PHARMACOLOGY: Methadone is a long-lasting mu-receptor agonist with pharmacological properties qualitatively similar to those of morphine. Individuals who receive subcutaneous or oral methadone daily can develop partial tolerance to the nauseant, anorectic, miotic, sedative, respiratory-depressant, and cardiovascular effects.
    C) TOXICOLOGY: The agonist activity at mu-receptor is responsible for the physical dependency, sedation, and respiratory depression. Methadone prolongs the QT interval by interactions with cardiac potassium channels. Most patients develop minor QTc prolongation, a small number develop QTc greater than 500 msec and are at risk for torsades de pointes.
    D) EPIDEMIOLOGY: Poisoning is common with associated major effects and deaths.
    E) WITH THERAPEUTIC USE
    1) COMMON: Sedation, constipation, tolerance and dependence. OTHER ADVERSE EFFECTS: Dizziness, nausea, weakness, diaphoresis, anorexia, rash, urticaria, urinary retention, visual disturbances, headache, insomnia, constipation, bradycardia, elevated liver enzymes, palpitation, ventricular arrhythmias (reported with maintenance dose of 400 mg PO daily), type I hypersensitivity reaction. Withdrawal symptoms (restlessness, irritability, weakness, lacrimation, excessive perspiration, anxiety, depression, dilated pupils, tremors, tachycardia, nausea, vomiting, diarrhea) may occur in dependent individuals if used with opioid antagonist or agonist/antagonist, or with rapid tapering or rapid discontinuation. Prolonged QT interval and limited cases of torsades de pointes have been reported in patients receiving methadone.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Lightheadedness, dizziness, sedation, nausea, vomiting, miosis (variable in children), and constipation.
    2) SEVERE TOXICITY: Coma, respiratory depression, respiratory arrest, pulmonary edema, bradycardia, hypotension, QT prolongation, torsade de pointes, dysrhythmias, ventricular tachycardia, ventricular fibrillation, cardiomyopathy, syncope, rhabdomyolysis, seizure, cardiac arrest and death.
    3) ONSET: May be delayed. DURATION: Prolonged, 12 hours to several days after exposure.
    0.2.7) NEUROLOGIC
    A) Severe somnolence progressing to coma or stupor may occur following overdose with these agents. Central nervous system adverse events (agitation, confusion, insomnia, euphoria, dysphoria, and visual disturbances) have occurred following methadone use. Toxic encephalopathy has been reported following methadone exposure.
    0.2.16) ENDOCRINE
    A) WITH POISONING/EXPOSURE
    a) Nonketotic hyperglycemia was observed in 3 toddlers admitted with nonfebrile coma following methadone overdoses.
    0.2.20) REPRODUCTIVE
    A) Methadone is classified as FDA pregnancy category C. Use of narcotic analgesics and/or methadone during pregnancy, particularly at high doses or for prolonged periods, has been associated with adverse effects in the neonate or infant; reported effects have included physical dependence/withdrawal, intrauterine growth retardation, visual impairment, and respiratory depression; these are similar to adverse effects observed in women who use cocaine during pregnancy. Methadone is excreted into human breast milk and methadone has been detected in infants breastfed by mothers taking methadone. The safety of methadone use while breastfeeding is controversial and recommendations are varied. In animal and human fertility studies, methadone use in males has produced adverse effects including semen abnormalities, increased embryolethality, and neonatal mortality.

Laboratory Monitoring

    A) Monitor vital signs and mental status carefully. Continuous pulse oximetry monitoring, and capnometry if available.
    B) Obtain a 12-lead ECG, and institute continuous cardiac monitoring in patients with moderate to severe toxicity.
    C) Monitor bed side blood glucose.
    D) Obtain a chest x-ray if aspiration or pulmonary edema is suspected.
    E) Methadone is not detected on most opioid urine drug screens.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Symptomatic and supportive care with continuous monitoring of airway patency is the mainstay of treatment in patients who present with mild to moderate methadone toxicity. Carefully monitor respiratory rate and end tidal CO2 to avoid unrecognized respiratory depression.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Administer oxygen and assist ventilation for respiratory depression. Naloxone is the antidote indicated for severe toxicity (respiratory or CNS depression). Early intubation and ventilation assistance should be performed if respiratory depression does not respond to naloxone. Adequate circulatory support with IV fluids and vasopressors (if needed) should be assured if a patient presents with circulatory collapse that does not reverse with naloxone. Treat seizures with benzodiazepines, propofol and/or barbiturates. Monitor for QT prolongation and dysrhythmias, and follow the appropriate ACLS algorithm if needed. Consider extracorporeal membrane oxygenation in cases of pulmonary edema not responsive to mechanical ventilation with high PEEP.
    C) DECONTAMINATION
    1) PREHOSPITAL: GI decontamination is not recommended because of the risk of aspiration with loss of airway protection and risk of seizure.
    2) HOSPITAL: Administer activated charcoal if the patient presents early after a large ingestion with appropriate level of consciousness, patent airway, and can drink the charcoal.
    D) AIRWAY MANAGEMENT
    1) Perform early in patients with severe methadone toxicity who presents with respiratory depression that does not reverse with naloxone. Noninvasive ventilation or endotracheal intubation may be necessary in patients who develop pulmonary edema. Noninvasive ventilation or endotracheal intubation may be necessary in patients who develop pulmonary edema. Consider extracorporeal membrane oxygenation to treat pulmonary edema refractory to standard measures.
    E) ANTIDOTE
    1) Naloxone, an opioid antagonist, can be administered intravenously, intramuscularly, subcutaneously, intranasally or via an endotracheal tube. The usual dose is 0.4 to 2.0 mg IV. In patients with suspected opioid dependence (including most patients taking methadone overdoses) incremental doses of 0.2 mg IV should be administered, titrated to reversal of respiratory depression and coma, to avoid precipitating acute opioid withdrawal. Doses may be repeated every 2 to 3 minutes up to 20 mg. Very high doses are rarely needed. Nebulized naloxone (2 mg naloxone in 3 mL normal saline administered via nebulizer face mask) is also effective. Continuous infusion is likely to be necessary in patients who have ingested methadone, as the duration of action of naloxone is 1 to 2 hours, compared with a duration of action of 24 hours for methadone. The infusion should be started at a rate such that two thirds of the dose effective for initial reversal is administered each hour, and titrated as needed. Naloxone can potentiate withdrawal in opioid-dependent patients. Opioid withdrawal is not life threatening, but is extremely uncomfortable for the patient.
    F) TORSADES DE POINTES
    1) Correct electrolyte abnormalities. Intravenous magnesium and overdrive pacing as needed.
    G) ENHANCED ELIMINATION
    1) Because of the large volume of distribution and increased protein binding, there is no role for enhanced elimination.
    H) PATIENT DISPOSITION
    1) HOME MANAGEMENT: All patients with suspected methadone overdose (deliberate or inadvertent) should be referred to a healthcare facility.
    2) OBSERVATION CRITERIA: Patients with mild toxicity who do not require naloxone should be observed for at least 8 hours. All children who have ingested any amount of methadone need to be observed in an Emergency Department (ED) for at least 8 hours and considered for hospital admission.
    3) ADMISSION CRITERIA: All patients who develop CNS or respiratory depression should be admitted for observation (for at least 24 hours) even after adequate response to naloxone therapy. Patients who required intubation or a naloxone infusion require an intensive care unit admission. Patients should not be discharged until they have remained awake and alert for 4 to 6 hours after the naloxone infusion has been discontinued.
    4) CONSULT CRITERIA: Consult a poison center or medical toxicologist for assistance in managing patients with severe toxicity, or in whom the diagnosis is not clear.
    I) PITFALLS
    1) Failure to recognize the potential lethality of an unintentional ingestion of even small doses in a pediatric population or opioid naive adults.
    2) Inadequate observation for respiratory depression after methadone overdose.
    J) PHARMACOKINETICS
    1) Onset: 10 to 20 minutes (parenteral), 30-60 minutes (orally). Duration of action: methadone intoxication may last more than 24 hours. Absorption: well absorbed; peak concentration after 4 hours. Distribution: Vd 4 - 5 L/Kg. Protein binding: 90% after therapeutic dose. Half-life after therapeutic doses: 15-55 hours. Metabolism: hepatic. Rifampin and phenytoin accelerate the metabolism of methadone and can precipitate withdrawal symptoms. Elimination half-life may be prolonged with alkaline pH and decreased during pregnancy. TOXICOKINETICS: Methadone may accumulate with repeat dosing. There is substantial overlap in the blood concentrations reported during therapeutic dosing and those reported following overdose.
    K) DIFFERENTIAL DIAGNOSIS
    1) Other opioids overdoses such as heroin, morphine, buprenorphine, codeine, tramadol, and pethidine may present with similar presentation. Overdose with other sedating agents (e.g., ethanol/benzodiazepine/barbiturate, gamma hydroxybutyrate, and antipsychotics) or overdose with central alpha-2 agonists (e.g., clonidine, tizanidine, imidazoline decongestants). Other: head injury/hemorrhage, hypoglycemia, and/or hypoxia.

Range Of Toxicity

    A) TOXIC DOSE: There is little margin between a therapeutic methadone dose for an opioid tolerant person and a toxic dose with doses of 30 to 40 mg potentially lethal for a non-tolerant adult. Children are particularly vulnerable to overdose. Inadvertent ingestion of methadone was found to be the most toxic of the opioids; doses as low as a single tablet can lead to death (10 mg can be potentially lethal for a 10 kg toddler). Patients who receive early treatment that prevents hypoxia and respiratory depression generally do well.
    B) THERAPEUTIC DOSE: ADULTS: ORAL: Opioid naive patients: 2.5 mg orally every 8 hours initially, titrated slowly to effect. Opioid dependent patients: 10 to 30 mg orally titrated slowly to effect. PARENTERAL: 2.5 to 10 mg. PEDIATRIC: 0.1 to 0.2 mg/kg orally every 6 hours as needed; maximum of 10 mg.

Summary Of Exposure

    A) USES: Methadone is used to treat chronic pain and to prevent symptoms of opioid withdrawal in opioid dependent patients. It is also rarely used as an antiperistaltic agent.
    B) PHARMACOLOGY: Methadone is a long-lasting mu-receptor agonist with pharmacological properties qualitatively similar to those of morphine. Individuals who receive subcutaneous or oral methadone daily can develop partial tolerance to the nauseant, anorectic, miotic, sedative, respiratory-depressant, and cardiovascular effects.
    C) TOXICOLOGY: The agonist activity at mu-receptor is responsible for the physical dependency, sedation, and respiratory depression. Methadone prolongs the QT interval by interactions with cardiac potassium channels. Most patients develop minor QTc prolongation, a small number develop QTc greater than 500 msec and are at risk for torsades de pointes.
    D) EPIDEMIOLOGY: Poisoning is common with associated major effects and deaths.
    E) WITH THERAPEUTIC USE
    1) COMMON: Sedation, constipation, tolerance and dependence. OTHER ADVERSE EFFECTS: Dizziness, nausea, weakness, diaphoresis, anorexia, rash, urticaria, urinary retention, visual disturbances, headache, insomnia, constipation, bradycardia, elevated liver enzymes, palpitation, ventricular arrhythmias (reported with maintenance dose of 400 mg PO daily), type I hypersensitivity reaction. Withdrawal symptoms (restlessness, irritability, weakness, lacrimation, excessive perspiration, anxiety, depression, dilated pupils, tremors, tachycardia, nausea, vomiting, diarrhea) may occur in dependent individuals if used with opioid antagonist or agonist/antagonist, or with rapid tapering or rapid discontinuation. Prolonged QT interval and limited cases of torsades de pointes have been reported in patients receiving methadone.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: Lightheadedness, dizziness, sedation, nausea, vomiting, miosis (variable in children), and constipation.
    2) SEVERE TOXICITY: Coma, respiratory depression, respiratory arrest, pulmonary edema, bradycardia, hypotension, QT prolongation, torsade de pointes, dysrhythmias, ventricular tachycardia, ventricular fibrillation, cardiomyopathy, syncope, rhabdomyolysis, seizure, cardiac arrest and death.
    3) ONSET: May be delayed. DURATION: Prolonged, 12 hours to several days after exposure.

Vital Signs

    3.3.2) RESPIRATIONS
    A) Both tidal volume and respiratory rate are depressed at higher drug concentrations following an overdose (Wolff, 2002).
    3.3.3) TEMPERATURE
    A) Hypothermia may occur following a methadone overdose (Wolff, 2002).
    B) CASE REPORT: Hypothermia (35.6 degrees C) developed in a 3-year-old girl who became comatose after ingesting an unknown amount of methadone (Mills et al, 2008).
    3.3.4) BLOOD PRESSURE
    A) Hypotension may be seen following an overdose (Wolff, 2002).
    B) CASE REPORT: Hypotension (BP 70/30 mm Hg) developed in a 3-year-old girl after ingesting an unknown amount of methadone (Mills et al, 2008).
    3.3.5) PULSE
    A) A decrease in pulse rate may occur following an overdose (Wolff, 2002).
    B) CASE REPORT: Tachycardia (heart rate 120 beats/min, likely secondary to hypotension and hypoxia) developed in a 3-year-old girl after ingesting an unknown amount of methadone (Mills et al, 2008).

Heent

    3.4.3) EYES
    A) OVERDOSE: MIOSIS is a typical opioid effect (Glatstein et al, 2009; Binchy et al, 1994; Shaw et al, 2011), and may last up to 72 hours. In a case series of 42 children, 17 had pinpoint pupils following accidental exposure to methadone (Glatstein et al, 2009; Binchy et al, 1994). Pinpoint pupils are typical in adults following overdose, but are not always seen in pediatric patients following acute overdose (Brooks et al, 1999). Pupillary response after overdose correlates well with methadone plasma concentrations (Wolff, 2002). An 8-month-old and a 3-year-old child presented with pinpoint pupils after inadvertently drinking juice containing methadone (Schwab & Caggiano, 2001). A 3-month-old presented with miotic pupils after intentional methadone poisoning (Lee & Lam, 2002).
    1) Miosis is not a universal response to an overdose of these drugs. Evenly or unevenly dilated pupils may occur; patients with profound coma may present with dilated pupils (Wolff, 2002).
    B) THERAPEUTIC USE: Visual disturbances including blurred vision have been reported with methadone therapy (Wolff, 2002; Prod Info Methadone hydrochloride (Methadone HCL(R), 1996).
    3.4.4) EARS
    A) WITH POISONING/EXPOSURE
    1) Hearing loss has been reported after methadone overdose.
    a) CASE REPORT: A 37-year-old man presented with nausea, hearing loss, confusion, and a low respiratory rate after ingesting 15 methadone 5 mg tablets. His symptoms improved after receiving a single dose of naloxone; however, he complained of profound hearing loss with a mild tinnitus, but no vestibular symptoms or aural fullness. His neurological examination was normal. Pure tone audiometry showed bilateral sudden sensorineural hearing loss of on average -57 db in the right ear and -40 db in the left ear. His condition continued to improve and a second audiometry was normal on day 10 (van Gaalen et al, 2009).
    b) CASE REPORT: Two patients, a 30-year-old man and a 25-year-old woman, developed bilateral hearing loss after ingesting an unknown amount of methadone. Toxicology screening tests were positive for methadone and tetrahydrocannabinol (THC). Both patients recovered following supportive care (Christenson & Marjala, 2010).
    c) CASE REPORT: A 20-year-old man presented with lethargy, miosis, tachycardia, tachypnea, mild respiratory distress and bilateral hearing loss after an inadvertent methadone overdose. A chest radiograph revealed pulmonary edema, with mild bibasilar infiltrates. Despite IV naloxone (0.4 mg) therapy, his symptoms did not improve. Laboratory analysis revealed leukocytosis of 25,000/dL, serum sodium of 149 mEq/L, a serum creatinine of 1.6 mg/dL, a CK of 2452 international units/L, an alanine aminotransferase of 227 IU/L, and aspartate aminotransferase of 243 IU/L. A urine drug screening immunoassay revealed the presence of methadone and cannabinoids. His venous blood gas analysis showed a respiratory acidosis, with a pH of 7.13 and a pCO of 96 mm Hg. He was treated for aspiration pneumonitis and rhabdomyolysis in the ICU and following further supportive care, his symptoms, including his hearing loss resolved on day 4 (Shaw et al, 2011).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) HYPOTENSIVE EPISODE
    1) WITH POISONING/EXPOSURE
    a) As with other narcotic analgesics, circulatory depression may develop following overdose with methadone (George et al, 2012; Prod Info Methadone hydrochloride (Methadone HCL(R), 1996; Mills et al, 2008; DeBoer et al, 2001). Hypotension can occur with severe overdosage resulting in circulatory collapse and cardiac arrest (Wolff, 2002).
    b) CASE SERIES: In a case series of 44 episodes of accidental methadone exposure in children, 4 developed hypotension (Binchy et al, 1994).
    c) CASE REPORT: In separate cases, a 16-year-old boy and a 19-year-old woman were found unresponsive and hypotensive after intentionally ingesting an unknown amount of methadone. The 16-year-old boy had a blood pressure of 60/30 mmHg. The 19-year-old woman had a blood pressure of 59/36 mmHg and a heart rate of 120 beats/min. Neither individual responded to naloxone. They were intubated and taken to an emergency department where fluid resuscitation and vasopressors were administered. In both cases, hypoxia persisted despite treatment with nitric oxide and high-frequency oscillatory ventilation. However, after 3 days of extracorporeal membrane oxygenation both individuals improved and were discharged home within 3 to 5 days without serious complications (Daugherty, 2011).
    B) BRADYCARDIA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 27-year-old man enrolled in a methadone maintenance program began taking 40 mg methadone daily. He presented, 1 day after initiating therapy, with a severe headache, blood pressure 137/66 mmHg, and pulse rate 90 beats/min, and was diagnosed with a probable subarachnoid hemorrhage. Within 24 hours, the patient's heart rate decreased to 35 beats/min with a blood pressure of 127/77 mmHg. An ECG showed sinus bradycardia, and echocardiogram showed normal heart valves and no septal wall abnormalities. The patient's heart rate improved with withdrawal of methadone therapy and administration of glucagon. Two days later, rechallenge with 45 mg of methadone resulted in recurrence of the bradycardia (32 beats/min). The patient recovered following complete cessation of methadone (Ashwath et al, 2005).
    2) WITH POISONING/EXPOSURE
    a) Circulatory depression including bradycardia may occur in overdose with methadone (Wolff, 2002; Prod Info Methadone hydrochloride (Methadone HCL(R), 1996).
    C) PROLONGED QT INTERVAL
    1) WITH THERAPEUTIC USE
    a) SUMMARY: There have been reports of prolonged QT interval in patients receiving methadone and limited cases of torsades de pointes (Thanavaro & Thanavaro, 2011; Pimentel & Mayo, 2008; Ehret et al, 2006; Lamont & Hunt, 2006). In a small prospective case series, an increase in QTc interval was observed with the start of methadone therapy, but no episodes of torsades de pointes were reported (Fredheim et al, 2006).
    b) In a 3-year prospective study of 130 patients who used a stable low dose of methadone (mean dose: 18.2 mg/day) as pain treatment for at least 1 week, it was determined that 50% of patients were potentially at risk for torsades de pointes; 5% of patients developed QTc times greater or equal to 500 ms and were at a serious risk of developing torsades de pointes. Other potential QTc-prolonging medications were used by about 70% of patients (van den Beuken-van Everdingen et al, 2013).
    c) A single center, cross-sectional study of 180 patients (average age, 32.6 years; 123 men) showed an 8.8% incidence of QT interval prolongation in patients in a methadone maintenance therapy program with a mean methadone dose of 80.4 mg (SD +/- 27.5 mg). Significant increases in QTc interval prolongation were gender specific; males had an 8.3% incidence and females had a 0.5% incidence based on gender specific definitions (greater than 450 msec for males and greater than 470 msec for females). However, 11.1% of the patients had QTc intervals greater than 450 msec compared with approximately 2.5% of the general population. Neither methadone dose nor the presence of cocaine as detected by urine toxicology screen had an effect on incidence of QT interval prolongation (Roy et al, 2012).
    d) In a comparison study of 167 methadone maintenance patients with 80 injection drug users not receiving methadone, the prevalence of QTc prolongation to 0.50 second or longer was 16.2% in the methadone group compared with 0% in the control group. Torsades de pointes was observed in 6 (3.6%) patients in the methadone group. A higher daily methadone dose was weakly but significantly associated with QT prolongation. The lowest daily doses associated with QTc prolongation was 20 mg/day which produced a QTc of 0.46 second and 30 mg/day produced a QTc of 0.50 second. Daily methadone doses from 40 to 200 mg/day produced torsades de pointes in this series.
    1) The authors noted that QT prolongation was associated with a higher daily methadone dose. In addition, hypokalemia, lower prothrombin level which may suggest a decrease in liver function, and use of CYP3A4 inhibitors may act synergistically with methadone to prolong QT (Ehret et al, 2006).
    e) In a small prospective case series of 8 healthy adults with chronic nonmalignant pain, an increase in QTc time was observed with the start of methadone therapy that was statistically, but not clinically significant. The mean QTc time increased from 0.416 at the start of therapy to 0.436 two weeks after beginning therapy. An increase of QTc times above 0.50 seconds were not observed, nor any episodes of dysrhythmias. Follow-up at 9 months, indicated that overall QTc changes were small and were not clinically or statistically significant in this patient population. However, two patients did have QTc increases that were approaching clinical significance (Fredheim et al, 2006).
    f) In a cross-sectional study of 138 patients in methadone maintenance treatment (MMT), no correlation was found between QTc interval and methadone doses and serum levels. All patients were receiving methadone 40 to 290 mg/day (mean dose, 170.9 +/- 50.3 mg/day) for a minimum of 100 days up to 10.7 years. Bazett formula was used to calculate corrected-QT intervals. The mean QTc of all patients was 418.3 +/- 32.8 milliseconds (ms) and the mean serum methadone level was 708.2 +/- 363.1 ng/mL. QTc intervals above 500 ms (prolonged) were observed in 3 patients. Two of these patients died from non-cardiac causes within a few months after the study. Nineteen patients had QTc intervals of between 450 and 499 ms (possibly prolonged). All 22 patients with QTc intervals of 450 ms and greater were receiving methadone doses of greater than 120 mg/day (Peles et al, 2007).
    D) TORSADES DE POINTES
    1) WITH THERAPEUTIC USE
    a) In a review of methadone-induced torsades de pointes reported in the medical literature, it was found that high-dose methadone and co-medication use were the most likely factors associated with torsades de pointes. However, the presence of hypokalemia, hepatic dysfunction, or coadministration of CYP3A4 inhibitors was not associated with methadone-induced torsades de pointes, but it did appear to increase the risk of long QT syndrome (LQTS) (Justo, 2006).
    b) TORSADES DE POINTES MIMICKING SEIZURES: A 29-year-old woman who had been using methadone 60 mg twice daily for 6 years, presented with a 3-month history of frequent convulsive episodes (up to 3 times per week). All laboratory results, including a CT scan of the head, were normal. An ECG revealed a mildly prolonged corrected QT interval (QTc) of 445 msec. EEG and ECG changes were recorded during a typical convulsive episode, which manifested with rapid breathing associated with bilateral dystonic arm posturing, whole body myoclonic jerks and noisy breathing, loss of consciousness, and flaccid and ceased respiration. After 54 seconds, she regained consciousness and resumed breathing. The recorded ECG revealed torsades de pointes (TdP) lasting for 139 seconds and EEG revealed complete voltage suppression for 48 seconds. At this time, she experienced a cardiac arrest, but recovered spontaneously. Another ECG revealed QTc prolongation at 536 msec and symmetrical T-wave inversions in the anterior and inferior leads. Following supportive care, her condition gradually improved and she was discharged 2 days after presentation with a plan to begin buprenorphine with a cardiac followup (Raina et al, 2014).
    c) CASE REPORT: A 55-year-old man treated with methadone (dose not reported) for chronic painful neuropathy presented to the emergency department (ED) with recurrent syncope and ventricular tachycardia noted on Holter monitor. He was defibrillated twice with immediate return of pulses and consciousness. Polymorphic ventricular tachycardia was identified on ECG and he was treated with 150 mg IV bolus amiodarone and 6 g IV magnesium. He was admitted to the ICU while methadone was discontinued and long-acting oral morphine sulfate was initiated. His QRS and QTc normalized to 0.08 and 0.462 respectively within days and he was discharged home without sequelae (Nordt et al, 2011).
    d) CASE REPORT: A 40-year-old woman developed symptomatic torsade de pointes (TdP) and prolonged QT after an increase in her daily dose of methadone to 135 mg/day the previous week. Her hospital course was remarkable for multiple runs of polymorphic ventricular tachycardia consistent with TdP. She also experienced seizure activity and a prolonged episode of syncope immediately after arrival. Following cardioversion and treatment with magnesium sulfate and lidocaine, her condition stabilized. Following the reduction in the patient's methadone dose to 60 mg/day, her QT interval was shortened significantly. Three days after admission, she had an internal cardioverter-defibrillator implanted (Pimentel & Mayo, 2008).
    e) CASE REPORT: A 61-year-old heroin user who had been taking maintenance methadone 110 mg/day, presented after 2 episodes of dizziness and near-syncope. He had a medical history of smoking, diabetes mellitus, hypertension, anemia, posttraumatic stress disorder, and anxiety, and had been taking glyburide, hydrochlorothiazide, lisinopril, and metformin daily. Physical examination showed an irregular pulse, a grade 2/6 holosystolic murmur, and few scattered rhonchi. Laboratory results showed anemia, hypokalemia (2.5 mEq/L), and hypomagnesemia (1.5 mg/dL). An ECG revealed normal sinus rhythm with atrial bigeminy and a prolonged QTc (626 msec). He later developed chest pain and palpitation associated with torsade de pointes (TdP). Following supportive care, including IV potassium and magnesium, TdP resolved and QTc interval became shorter, but remained prolonged until methadone was replaced with a taper of long-acting morphine sulfate and then buprenorphine. Extended-release metoprolol was started after severe left ventricular dysfunction (ejection fraction of 35%) was diagnosed and lisinopril dose was increased. Five months later, his ECG was normal, except for borderline QTc prolongation (443 msec) (Thanavaro & Thanavaro, 2011).
    E) ELECTROCARDIOGRAM ABNORMAL
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 17-year-old man who ingested an unknown quantity of methadone developed hyperkalemia (7.3 mEq/L) and the ECG revealed peaked T-waves diffusely. He was tachycardic and hypotensive on admission to the ED. The patient developed diencephalic seizures. Following a prolonged recovery period, the patient improved (DeBoer et al, 2001).
    F) CARDIAC ARREST
    1) WITH POISONING/EXPOSURE
    a) Profound respiratory depression leading to cardiopulmonary arrest has been reported following methadone overdose (Copeman & Robins, 2000). Respiratory depression may be delayed and may recur after naloxone administration.
    b) CASE REPORT: An extremely obese 3-year-old boy was treated with naloxone infusion following an unintentional 40 mg methadone overdose. Following discontinuation of naloxone, the boy remained symptom-free for 12 hours when he suffered a cardiopulmonary arrest and eventually died, with toxic levels of serum methadone found at autopsy. It was speculated that the biphasic pharmacokinetic properties of methadone and its long half-life were accentuated in this case of obesity (Copeman & Robins, 2000).
    G) TACHYCARDIA
    1) WITH POISONING/EXPOSURE
    a) Tachycardia has been reported following methadone ingestion (Shaw et al, 2011; Daugherty, 2011; Mills et al, 2008; DeBoer et al, 2001).
    b) CASE REPORT: Tachycardia (heart rate 120 beats/min, likely secondary to hypotension and hypoxia) developed in a 3-year-old girl after ingesting an unknown amount of methadone (Mills et al, 2008).
    H) TAKOTSUBO CARDIOMYOPATHY
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 60-year-old man with a history of COPD, hepatitis C, and IV drug abuse treated with high-dose methadone presented to the emergency department with labored breathing and lethargy after inadvertently ingesting double his prescribed dose of methadone. Shortly after treatment with naloxone infusion, he developed symptoms consistent with Takotsubo cardiomyopathy (stress-induced cardiomyopathy). He complained of retrosternal chest pressure and became hypertensive, agitated, and tachycardic. ECG showed ST-segment elevation with T-wave inversions and normal QT interval. Laboratory analysis showed a troponin level of 1.68 nanograms/mL and CK-MB of 30.4 international units/L. However, no significant coronary artery disease was detected with coronary angiography. A postcatheterization echocardiogram revealed an ejection fraction (EF) of 35% along with anteroapical and inferoapical ballooning and distal septal hypokinesis. He was discharged home on day 5 of admission in stable condition. At 1 month follow-up, echocardiogram showed an EF of 55% with no residual hypokinesis and ECG was normal (Saiful et al, 2011).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) ACUTE RESPIRATORY INSUFFICIENCY
    1) WITH POISONING/EXPOSURE
    a) Prolonged toxicity of 24 to 48 hours duration including respiratory depression may occur in individuals overdosing with methadone (Shaw et al, 2011; Ortiz-Gomez & Souto-Ferro, 2006; Lee & Lam, 2002; Wolff, 2002; Hendra et al, 1996; Gayle et al, 1991; Romac, 1986; Sey et al, 1971). Respiratory depression is the hallmark of opioid overdose; however, it is only seen in about 50% of patients with CNS depression (Wolff, 2002).
    1) ADULT
    a) CASE REPORT: Bradypnea (respirations 6/minute) was reported in an elderly opiate-naive woman following inadvertent exposure to 320 mg of methadone. The patient was intubated for 24 hours and required reintubation 12 hours later for hypercapnia. On day 5, pulmonary edema developed. The patient gradually improved and was discharged from the ICU on day 9 (Hantson et al, 2003).
    2) PEDIATRIC
    a) The onset of respiratory depression after methadone overdose was delayed for 6 hours in a 6-year-old child (Geller & Garrettson, 1994).
    b) Several children have developed methadone poisoning after being given an antibiotic suspension which was reconstituted with methadone hydrochloride (Gayle et al, 1991; Lalkin et al, 1999).
    c) CASE REPORT: A 6-year-old boy with autistic disorder developed severe respiratory distress and decreased level of consciousness several hours after receiving a tablespoonful of a cough syrup. It was determined later that the bottle of cough syrup contained methadone that was given to the patient by mistake. Laboratory results revealed the presence of methadone in the patient's urine. Following supportive care, the patient condition gradually improved and he was transferred out of the ICU after 36 hours of observation (Gharib et al, 2014).
    d) CASE REPORT: A 4-year-old was exposed to methadone following an antibiotic dispensing error which resulted in unresponsiveness and decreased breathing, occurring after the second 5 mL dose. Testing revealed a methadone concentration of 2.4 mg/L. Following supportive care, the child made a complete recovery (Lalkin et al, 1999).
    e) CASE REPORT: Normal respiratory rate in a toddler following acute methadone ingestion was reported. Compromised respiratory function was demonstrated by a decrease in minute volume, resulting in CO2 retention, hypoxia, and respiratory acidosis (Brooks et al, 1999).
    f) CASE REPORTS: Two cases of an 8-month-old and a 3-year-old child who drank juice containing methadone are reported. Both presented to the ED unresponsive and with depressed respirations. Both recovered following naloxone injections (Schwab & Caggiano, 2001).
    g) CASE SERIES: In a case series of 42 children accidentally exposed to methadone (therapy was intended for an adult in the home), 10 developed respiratory depression (Binchy et al, 1994).
    B) ACUTE LUNG INJURY
    1) WITH POISONING/EXPOSURE
    a) Acute lung injury may result from overdoses of methadone (Gonzva et al, 2013).
    b) Pulmonary edema was reported in 20 patients following methadone ingestions (Wilen et al, 1975). Pulmonary edema was evident in 38 fatalities in which methadone was detected in blood in a retrospective review (Karch & Stephens, 2000). Adult respiratory distress syndrome has been reported following methadone overdose (Wolff, 2002).
    c) CASE REPORT: In separate cases, a 19-year-old woman with a history of illicit drug use and a 16-year-old boy were found unresponsive and in respiratory distress with O2 saturations of 60% and 50% respectively after intentionally ingesting an unknown amount of methadone. The 16-year-old boy had a blood pressure of 60/30 mmHg. The 19-year-old woman had a blood pressures of 59/36 mmHg and a heart rate of 120 beats/min. Neither individual responded to naloxone. They were intubated and taken to an emergency department where fluid resuscitation and vasopressors were administered. In both cases, hypoxia persisted (PaO2, 40 mmHg and 35 mmHg) despite treatment with nitric oxide and high-frequency oscillatory ventilation. Pulmonary edema was confirmed with chest radiograph in each case. Serum methadone was detected in both cases; however, urine opiates were negative for both. Extracorporeal membrane oxygenation was initiated and continued for 3 days for both individuals. They were discharged within 3 to 5 days without serious complications (Daugherty, 2011).
    d) CASE REPORT: A 54-year-old man developed non-cardiogenic pulmonary edema following an overdose of an unknown amount of methadone. His arterial blood gas revealed a pH of 7.18, PaCO2 of 10.11 kPa (75.8 mmHg), and PaO of 6.13 kPa (45.98 mm Hg). A portable chest x-ray revealed bilateral florid pulmonary edema. Despite initial supportive care that included naloxone, no improvement in his respiratory failure was observed and he remained hypoxic and tachypneic. His pulmonary edema gradually improved following the early use of non-invasive ventilation (Ridgway & Pountney, 2007).
    e) CASE REPORT: A 20-year-old man who was found unconscious at home after ingesting an unknown amount of methadone, in addition to drinking 5 beers and smoking cannabis, regained consciousness and had persistent hypoxia and increased respiratory rate after prehospital naloxone therapy. Chest radiograph showed diffuse infiltrates and echocardiography showed no cardiac function impairment, suggesting a noncardiogenic pulmonary edema. He was treated with naloxone infusion and noninvasive ventilation and recovered within 3 days of methadone use. (Gonzva et al, 2013).
    C) PNEUMONIA
    1) WITH POISONING/EXPOSURE
    a) Bronchopneumonia, occurring very early, has been identified histologically in several methadone overdose cases resulting in death. In one fatality, even though methadone serum levels were not high (0.06 mg/L), death was due to extensive bronchopneumonia and attributed to prolonged unconsciousness following methadone overdose which precipitated aspiration pneumonia (Green et al, 2000).
    D) PULMONARY EDEMA
    1) WITH POISONING/EXPOSURE
    a) Acute pulmonary edema with severely depressed left ventricular contractility was reported in an opiate-naive elderly woman 5 days after inadvertent exposure to 320 mg of methadone. Following supportive care the patient was discharged from the ICU on hospital day 9 (Hantson et al, 2003).
    E) RIGID CHEST
    1) WITH POISONING/EXPOSURE
    a) A neonate, who was born at 32 weeks gestation, was then readmitted to NICU for 4 weeks of age for respiratory distress from RSV, requiring 10 days of mechanical ventilation. On transfer to the ward, he was inadvertently administered methadone 0.5 mg/kg (intended dose 0.05 mg/kg). About 40 minutes post overdose he developed stridor and retractions, and was treated with nasal oxygen. About 100 minutes after the overdose he developed worsening stridor and increasing oxygen requirement that did not respond to nebulized epinephrine and albuterol. He became apneic, but no chest rise could be produced with bag-valve-mask ventilation. Naloxone was administered intramuscularly (0.08 mg) and within a minute chest rise was observed, followed by spontaneous reparations and return of consciousness (Lynch & Hack, 2010).

Neurologic

    3.7.1) SUMMARY
    A) Severe somnolence progressing to coma or stupor may occur following overdose with these agents. Central nervous system adverse events (agitation, confusion, insomnia, euphoria, dysphoria, and visual disturbances) have occurred following methadone use. Toxic encephalopathy has been reported following methadone exposure.
    3.7.2) CLINICAL EFFECTS
    A) CENTRAL NERVOUS SYSTEM DEFICIT
    1) WITH THERAPEUTIC USE
    a) The following adverse effects have been observed with the therapeutic use of methadone: euphoria, dysphoria, irritability, insomnia, agitation, headache, disorientation, and somnolence (Pimentel & Mayo, 2008; Prod Info Methadone hydrochloride (Methadone HCL(R), 1996; Eissenberg et al, 1997).
    b) IRRITABILITY or hyperstimulation (dysphoria, nervousness, mental confusion, and insomnia) have been reported with methadone (Ling et al 1978; (Tennant et al, 1986).
    c) COGNITIVE DEFICITS have been reported following methadone maintenance in a study comparing methadone maintenance patients to matched control subjects of non-heroin users. The methadone maintenance group had significantly higher rates of cognitive impairment (measurements of neuropsychological domains) than controls of alcohol dependence, heroin overdose and head injury (Darke et al, 2000).
    2) WITH POISONING/EXPOSURE
    a) Severe somnolence progressing to prolonged coma (more than 24 hours) or stupor may occur following overdose with these agents (Gonzva et al, 2013; van Gaalen et al, 2009; Shaw et al, 2011; Wolff, 2002; Wolff, 2002; Lee & Lam, 2002; Gayle et al, 1991; Romac, 1986; Sey et al, 1971). Due to its prolonged duration of action and long half-life (15 to 40 hours) (Haddad et al, 1998), methadone overdose has resulted in a relapse of decreased mental status after an acute overdose (Hendra et al, 1996). Other CNS overdose symptoms may include hallucinations, confusion, light-headedness, and headache (Wolff, 2002).
    b) ADULTS
    1) CASE REPORT: A 22-year-old man died 44 hours after an acute overdose of 420 mg methadone. A naloxone infusion was discontinued approximately 3 hours prior to his death, as he appeared stable (Hendra et al, 1996).
    c) PEDIATRIC
    1) Severe toxicity can develop in children exposed to small amounts of methadone (Tiras et al, 2006; Binchy et al, 1994). In one study of methadone patients with small children in their home, only 50% were found to properly store their medication (Calman et al, 1996).
    2) Lethargy has been reported in children following methadone exposure (Glatstein et al, 2009; Brooks et al, 1999; Lorenzo & Weinstock, 1995).
    3) CASE REPORT: A 6-year-old boy with autistic disorder developed severe respiratory distress and decreased level of consciousness several hours after receiving a tablespoonful of a cough syrup. It was determined later that the bottle of cough syrup contained methadone that was given to the patient by mistake. Laboratory results revealed the presence of methadone in the patient's urine. Following supportive care, the patient condition gradually improved and he was transferred out of the ICU after 36 hours of observation (Gharib et al, 2014).
    4) CASE REPORT: A 4-year-old was accidentally exposed to methadone following a dispensing error while preparing an oral antibiotic suspension. Unresponsiveness and decreased breathing occurred following a second 5 mL dose; testing revealed a methadone concentration of 2.4 mg/L. Following supportive care the child made a complete recovery (Lalkin et al, 1999).
    5) CASE REPORT: A 30-month-old child was reported to be lethargic, with an ataxic gait after ingesting a mouthful of methadone liquid. A single 2 mg naloxone injection resulted in prompt awakening with no further relapses (Brooks et al, 1999).
    6) CASE REPORT: A 4-year-old was initially agitated but quickly developed lethargy and difficulty breathing and was unresponsive several hours later after a 40 mg ingestion of methadone. A delay in medical treatment was reported and postmortem blood methadone level was 0.5 mg/L (Lorenzo & Weinstock, 1995).
    7) CASE REPORTS: Four pediatric fatalities were reported, ranging in age from 4 months to 5 years old, following unintentional methadone ingestions. Three of the children were found unresponsive and one child vomited and developed apnea (Li et al, 2000).
    B) SEIZURE
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: A 40-year-old woman developed symptomatic torsade de pointes, a prolonged QT, syncope and seizure activity after an increase in her daily dose of methadone to 135 mg/day the previous week (Pimentel & Mayo, 2008).
    2) WITH POISONING/EXPOSURE
    a) Methadone overdose has resulted in seizure-type activity (DeBoer et al, 2001). Seizure activity has been reported in infants exposed to methadone (Lee & Lam, 2002; Lorenzo & Weinstock, 1995).
    b) CASE SERIES: In a case series of 42 children accidentally exposed to methadone, 2 had seizures (Binchy et al, 1994).
    c) CASE REPORT: Following the ingestion of an unknown quantity of methadone, a 17-year-old boy developed paroxysmal sympathetic storm (diencephalic seizures) the day after hospital admission. On presentation he had been profoundly hypoxic and hypotensive. Decerebrate posturing and transient hypertension (following hypotension treated with dopamine) occurred. EEG revealed diffuse theta and delta wave slowing, but no focal findings or epileptic activity. Repeat head CT scan showed anoxic brain injury. Recovery was slow (3 months) (DeBoer et al, 2001).
    d) CASE REPORT: A 20-day-old premature neonate (born at a gestational age of 24.5 weeks) who was receiving fentanyl and morphine for analgesia and sedation after several tube thoracostomies for pneumothorax, developed respiratory depression within 2 hours of receiving 7.8 mg of methadone (about 200-fold the prescribed dose). Despite supportive therapy, he developed 4 episodes of clonic movements of the left arm and leg; however, no seizure activities were observed in an EEG during the clonic period. Following treatment with phenobarbital for presumed seizures and dopamine infusion for hemodynamic instability, his condition improved gradually (George et al, 2012).
    C) TOXIC ENCEPHALOPATHY
    1) WITH POISONING/EXPOSURE
    a) Methadone-induced acute toxic leukoencephalopathy has been reported in several patients (Metkees et al, 2015; Gheuens et al, 2010; Zanin et al, 2010; Anselmo et al, 2006). In MRI brain imaging, methadone-induced acute leukoencephalopathy usually affects the white matter more than the gray matter. This would allow the clinician to differentiate it from hypoxic injury or other toxicities (eg, carbon monoxide) that more commonly affect gray matter (Metkees et al, 2015).
    b) CASE REPORT: A 15-year-old girl presented in an unconscious state (Glasgow Coma Scale score of 3) after ingesting an unknown quantity of methadone. Her vital signs included a blood pressure of 147/109 mmHg, heart rate of 121 beats/min, and a respiratory rate of 26 breaths/min. Laboratory results revealed metabolic acidosis and hyperkalemia. Despite treatment with naloxone, her mental status did not improve. A brain MRI revealed diffuse nonenhancing T2 hyperintensities and restricted diffusion in the white matter of both hemispheres with sparing of subcortical U fibers, suggesting toxic leukoencephalopathy. Hypoxic-ischemic etiology was excluded because of lack of deep gray nuclear, cortical or cerebellar involvement. She later developed irreversible cerebral edema and eventually died despite supportive care (Metkees et al, 2015).
    c) CASE REPORT: A 49-year-old woman was admitted to an intensive care unit with respiratory failure after intentionally ingesting an amount of methadone 10 times her normal dose. She was placed on mechanical ventilation and seemed to recover as she was weaned off ventilation and transferred into psychiatric programs. However, about 2 weeks after transfer, she developed symptoms consistent with toxic spongiform leukoencephalopathy. Her condition deteriorated gradually over weeks beginning with akinetic syndrome and progressing to a full akinetic catatonic mute state and complete immobilization requiring tube feedings. At symptom onset, brain MRI showed bilateral diffuse white matter changes and lumbar puncture showed elevated lactate (2 mmol/L; reference less than 1.7 mmol/L) and decreased protein (18 mg/dL; reference 27 to 60 mg/dL). Brain MRI one month later showed progression of white matter changes and MR spectroscopy findings were consistent with neuronal damage. Brain biopsy was performed; histologic examination showed findings of toxic spongiform leukoencephalopathy. After 3 months of supportive physical therapy and botox injections, she regained normal function of her proximal muscle groups but kept dystonic position of her hands and feet. Brain MRI showed no improvement and frontal cognitive impairment persisted (Gheuens et al, 2010).
    d) CASE REPORT: A 3-year-old boy was inadvertently exposed to methadone and was found comatose. An initial head CT revealed acute obstructive hydrocephalus due to massive cerebellar edema and supratentorial lesions. Mannitol and dexamethasone were administered with no change in neurologic status. Surgical drainage resulted in immediate improvement in consciousness with residual right-sided hemiplegia and mutism. Hemiplegia slowly resolved. By day 6, high dose methylprednisolone was begun to treat the cerebellar lesions. The patient was discharged to home by day 16 with residual ataxia which slowly resolved over 4 weeks (Anselmo et al, 2006).
    e) CASE REPORT: A 3-year-old girl developed severe cerebellitis after ingesting an unknown amount of methadone. She presented unconscious (GCS 3/15) with labored breathing and hypothermia (35.6 degrees C), hypotension (BP 70/30 mm Hg), tachycardia (heart rate 120 beats/min), and acidosis (pH 6.8). CT revealed diffuse symmetrical edema of the cerebellum, resulting in compression of the fourth ventricle and hydrocephalus. An MRI brain scan confirmed these changes with the addition of bilateral watershed infarcts and extensive restricted diffusion throughout the cerebral grey matter. She underwent an urgent decompressive occipital craniotomy. On day 4, she experienced severe spasticity in all four limbs and had severe cortical visual impairment. An MRI brain scan two months after the methadone ingestion revealed maturation of the initial white matter changes and generalized atrophy of the cerebrum and cerebellum. Her condition gradually improved over the next 4 months (Mills et al, 2008).
    f) CASE REPORT: A 30-month-old girl presented unconscious (Glasgow Coma Scale 3/15) after ingesting 20 mg of methadone (methadone blood level: 0.063 mcg/mL). She recovered after treatment with naloxone infusion for 8 hours. Five days postingestion, she developed severe wide dystonic movements, psychomotor agitation, gait and balance impairment, and slurred speech. All neuroimaging studies were negative at this time and a diagnosis of toxic methadone encephalopathy was made. Despite treatment with lorazepam and clonazepam, only mild improvement was observed after 9 days. A repeat MRI, 19 days postingestion, revealed symmetric signal abnormalities in the temporomesial regions, basal ganglia, and substantia nigra. Her symptoms gradually improved, but another MRI performed 6 months postingestion revealed same cerebellar signal abnormalities and atrophic changes of the basal ganglia. She was completely asymptomatic at the 12-month follow-up examination (Zanin et al, 2010).
    D) INTRATHECAL INJECTION
    1) WITH POISONING/EXPOSURE
    a) Severe neurological symptoms including lower extremity weakness, numbness, back pain, and mental status changes occurred in a series of eight patients who inadvertently received intrathecal methadone. Implanted intrathecal catheter pumps were intended to be filled with morphine but due to a compounding error in the pharmacy, the pumps were filled with methadone and morphine. Other contaminants (ethanol and methanol) were also found in the refill solution. Three patients required laminectomy for removal of sterile abscesses from the exposure and as a result have paralysis or permanent leg weakness (Jones et al, 2002).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) DRUG-INDUCED ILEUS
    1) WITH THERAPEUTIC USE
    a) Most gastric disturbances observed (abdominal pain, constipation, nausea and vomiting) are a result of decreased gastric motility (Prod Info Methadone hydrochloride (Methadone HCL(R), 1996; Haddad et al, 1998).
    b) Symptoms of constipation are related to decreased peristalsis in the small intestine and colon (Prod Info Methadone hydrochloride (Methadone HCL(R), 1996; Haddad et al, 1998).
    B) SPASM OF SPHINCTER OF ODDI
    1) WITH THERAPEUTIC USE
    a) Spasm of the sphincter of Oddi can occur with methadone therapy (Haddad et al, 1998; Prod Info Methadone hydrochloride (Methadone HCL(R), 1996).
    C) NAUSEA AND VOMITING
    1) WITH POISONING/EXPOSURE
    a) Nausea and vomiting are typical signs following overdoses of these drugs (van Gaalen et al, 2009; Wolff, 2002).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) ABNORMAL LIVER FUNCTION
    1) WITH THERAPEUTIC USE
    a) Although infrequent, hepatotoxicity has been reported after therapeutic use of methadone (Walter, 1969; Lapiere, 1969).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) ABNORMAL SEXUAL FUNCTION
    1) WITH THERAPEUTIC USE
    a) Difficulty ejaculating and impotence have occurred with methadone during therapeutic use (Prod Info Methadone hydrochloride (Methadone HCL(R), 1996).
    B) RETENTION OF URINE
    1) WITH THERAPEUTIC USE
    a) Urinary retention and hesitancy have occurred following therapeutic use (Prod Info Methadone hydrochloride (Methadone HCL(R), 1996).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) RESPIRATORY ACIDOSIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Lethargy and respiratory acidosis (pH 7.26; pCO2 58 mm Hg; pO2 45 mm Hg; bicarbonate 25 mmol/L) developed in an 11-month-old infant after exposure to an unknown amount of 5-mg methadone tablets. The child's condition normalized after receiving an IV dose of 0.1 mg/kg naloxone (Glatstein et al, 2009).
    b) CASE REPORT: A 2-month-old male infant who was born to a mother with narcotic dependency, developed respiratory failure from suspected intentional methadone administration. The mother was on a methadone maintenance treatment program (methadone concentration: 60 mg/15 mL). Laboratory results revealed mixed respiratory and metabolic acidosis (pH 7.24; PCO2 59 mmHg; PO2 44 mmHg; base excess -4%). A urine toxicity screen confirmed the presence of methadone. Following supportive therapy, including naloxone therapy (0.005 mg/kg loading dose, 0.0025 mg/kg/hour continuous infusion), he recovered after 3 days of hospitalization (Siew et al, 2012).
    c) CASE REPORT: A 20-year-old man presented with lethargy, miosis, tachycardia, tachypnea, mild respiratory distress and bilateral hearing loss after an inadvertent methadone overdose. A chest radiograph revealed pulmonary edema, with mild bibasilar infiltrates. Despite IV naloxone (0.4 mg) therapy, his symptoms did not improve. Laboratory analysis revealed leukocytosis of 25,000/dL, serum sodium of 149 mEq/L, a serum creatinine of 1.6 mg/dL, a CK of 2452 international units/L, an alanine aminotransferase of 227 IU/L, and aspartate aminotransferase of 243 IU/L. A urine drug screening immunoassay revealed the presence of methadone and cannabinoids. His venous blood gas analysis showed a respiratory acidosis, with a pH of 7.13 and a pCO of 96 mm Hg. He was treated for aspiration pneumonitis and rhabdomyolysis in the ICU and following further supportive care, his symptoms, including his hearing loss resolved on day 4 (Shaw et al, 2011).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) ERUPTION
    1) WITH POISONING/EXPOSURE
    a) Pruritus may occur following methadone overdose (Wolff, 2002).
    B) INJECTION SITE REACTION
    1) WITH THERAPEUTIC USE
    a) Patients receiving subcutaneous injections of methadone may experience injection site reactions including erythema and induration. Incidence of local toxicity is high, and is not predictable. It may be uniformly managed by site rotation and concurrent injection of dexamethasone (Mathew & Storey, 1999).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) RHABDOMYOLYSIS
    1) WITH POISONING/EXPOSURE
    a) Methadone, like other opioids, may cause rhabdomyolysis following overdoses (Hoffman et al, 2000).
    b) CASE REPORT: A 34-year-old man developed limb ischemia and rhabdomyolysis (CK peaked at 166,000) after accidentally injecting a solution of ten 10-mg methadone tablets dissolved in water into his femoral artery instead of the femoral vein. An angiogram of the lower extremity showed focal areas of narrowing, suggesting spasm and evidence of distal small-particle embolization. Following supportive care, he was discharged 6 days later with patchy mottling throughout the lower extremity and a CK of 30,000 (Gramenz et al, 2009).
    B) MUSCLE RIGIDITY
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A neonate developed life-threatening rigid-chest syndrome after receiving a methadone dose of 10 times the ordered amount of 0.05 mg/kg per dose every 6 hours. He recovered completely after receiving naloxone therapy (Lynch & Hack, 2010).

Endocrine

    3.16.1) SUMMARY
    A) WITH POISONING/EXPOSURE
    a) Nonketotic hyperglycemia was observed in 3 toddlers admitted with nonfebrile coma following methadone overdoses.
    3.16.2) CLINICAL EFFECTS
    A) HYPERGLYCEMIA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORTS: Nonketotic hyperglycemia was observed in 3 toddlers admitted with nonfebrile coma following unintentional methadone exposure. Initial blood glucoses ranged from 378 mg/dL to 571 mg/dL, which normalized with an insulin drip. Hyperglycemia was found 4 to 8 hours after exposure in these patients.
    1) Although two of the toddlers recovered uneventfully, the third one had an anterolateral myocardial infarct and developed cerebral edema resulting in permanent neurologic injury. The authors suggested that the presence of hyperglycemia may have a role in precipitating cerebral edema due to its cerebral osmotic dysregulation (Tiras et al, 2006).

Reproductive

    3.20.1) SUMMARY
    A) Methadone is classified as FDA pregnancy category C. Use of narcotic analgesics and/or methadone during pregnancy, particularly at high doses or for prolonged periods, has been associated with adverse effects in the neonate or infant; reported effects have included physical dependence/withdrawal, intrauterine growth retardation, visual impairment, and respiratory depression; these are similar to adverse effects observed in women who use cocaine during pregnancy. Methadone is excreted into human breast milk and methadone has been detected in infants breastfed by mothers taking methadone. The safety of methadone use while breastfeeding is controversial and recommendations are varied. In animal and human fertility studies, methadone use in males has produced adverse effects including semen abnormalities, increased embryolethality, and neonatal mortality.
    3.20.2) TERATOGENICITY
    A) VISUAL ABNORMALITIES
    1) Congenital nystagmus was reported in 2 infants following in utero exposure to methadone. In both cases, the mothers used methadone throughout the duration of the pregnancy. The first infant was born at 33-weeks gestation. Concurrent diazepam use was reported by the mother. While neonatal abstinence syndrome (NAS) was not reported at birth, significant pendular horizontal nystagmus was reported within 3 months. Within 12 months, the infant was able to find null positions and visual acuity and cognitive development improved. The second infant was born at 40-weeks gestation with NAS present at birth and difficulty with visual following was noted. Within 8 months, improvement in visual function was observed and neurological development was normal (Tinelli et al, 2013).
    2) Visual defects and abnormal clinical visual assessments were discovered in more infants of drug-misusing mothers who were administered substitute methadone during pregnancy compared with nonmaternal-exposed infants. At the 6-month follow up, 32 (40%) of the 81 drug-exposed infants failed visual assessments (relative risk of abnormal assessment, 5.1; 95% CI, 1.3 to 20) compared with 2 of 26 healthy, nonmaternal-exposed infants. Strabismus was the most prevalent visual impairment, found in 20 infants (25%) of the drug-exposed cohort, 12 of whom also had exotropia and 8 of whom had esotropia. Reduced visual acuity (greater than 0.9 on logarithmic size progression chart) was found in 18 infants (22%) in the drug-exposed cohort, 11 of whom had other associated ophthalmic comorbidities. Nystagmus was detected in 9 drug-exposed infants (11%). Only one of these infants was exposed to methadone alone; most were exposed to other illicit drugs in utero. Delayed visual maturation (DVM) was present in 2 infants and suspected in 8 others, based on parental and caretaker reports of visual inattentiveness (McGlone et al, 2014).
    3) One study found a higher rate of strabismus in the infants of mothers who attended a methadone maintenance clinic than in the general population (24% vs 2.8% to 5.3%) (Nelson et al, 1987).
    B) HEAD CIRCUMFERENCE
    1) A retrospective study that evaluated early growth and developmental outcome of infants with in-utero exposure to high-dose methadone (100 mg or higher per day), low dose-methadone (less than 100 mg/day), and buprenorphine, reported lower head circumference z scores in infants prenatally exposed to methadone doses greater than 100 mg at 3 to 5 months of age. This finding can have a negative impact on motor skill development during early infancy (Bier et al, 2015).
    C) LACK OF EFFECT
    1) Based on data from the Teratogen Information System (TERIS), while maternal use of methadone during pregnancy as part of a supervised, therapeutic regimen may not pose a substantial teratogenic risk, data are insufficient to completely rule out risk. Methadone levels detected in amniotic fluid and cord plasma have been proportional to maternal plasma concentrations. Levels in the urine of neonates were lower than that in maternal urine (Prod Info DOLOPHINE(R) oral tablets, 2014; Prod Info DISKETS(R) dispersible tablets for oral suspension, 2007; Prod Info METHADOSE(TM) oral concentrate, sugar-free oral concentrate, 2007).
    2) In a retrospective review of Canadian databases from January 2006 through December 2010, there was no increased risk for mortality in infants less than 1 year old who were exposed to methadone in utero and experienced neonatal abstinence syndrome (NAS) as compared with the general population of Ontario (odds ratio, 1.45; 95% CI, 0.47 to 4.46; p=0.56). During this time period in Ontario, 8 deaths of infants younger than 1 year were reported to be associated with in utero exposure to methadone while a total of 1103 cases of NAS were recorded. Thus, a mortality rate of 8 deaths per 1103 cases diagnosed or 0.725% was calculated for infants exposed to methadone in utero. The infant mortality rate in Ontario for the same time period was 5.2 per every 1000 live births up to 1 year of age according to Statistics Canada (Kelly et al, 2012).
    D) ANIMAL STUDIES
    1) RATS AND RABBITS: Methadone did not demonstrate teratogenicity in rats or rabbits at doses approximately 3 and 6 times the human daily oral dose, respectively. High doses of methadone produced teratogenicity in guinea pigs, hamsters (reduced number of fetuses per litter, higher incidence of congenital malformations and maternal death), and mice (exencephaly) (Prod Info DISKETS(R) dispersible tablets for oral suspension, 2007; Prod Info METHADOSE(TM) oral concentrate, sugar-free oral concentrate, 2007; Prod Info DOLOPHINE(R) HYDROCHLORIDE oral tablets, 2006).
    2) RODENTS: Methadone was teratogenic in hamsters at 2 times the human dose and in mice at a dose equal to the human dose. Offspring of male rodents treated prior to mating showed increased neonatal mortality and behavioral differences (Prod Info DOLOPHINE(R) oral tablets, 2014).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    1) The manufacturer has classified methadone as FDA pregnancy category C (Prod Info DOLOPHINE(R) oral tablets, 2014; Prod Info DISKETS(R) dispersible tablets for oral suspension, 2007; Prod Info METHADOSE(TM) oral concentrate, sugar-free oral concentrate, 2007).
    2) Use of narcotic analgesics and/or methadone during pregnancy, particularly at high doses or for prolonged periods, has been associated with adverse effects in neonates, including physical dependence/withdrawal, intrauterine growth retardation, and respiratory depression (Cunningham et al, 1993). Due to the potential for lower trough plasma methadone concentrations during pregnancy, higher doses or shorter dose intervals of methadone may be warranted to avoid withdrawal symptoms (Prod Info DOLOPHINE(R) oral tablets, 2014). Use methadone during pregnancy only if the potential benefit to the mother outweighs the potential risk to the fetus (Prod Info methadone hydrochloride oral solution, 2008; Prod Info DOLOPHINE(R) oral tablets, 2014). If prolonged use during pregnancy is required, monitor the newborn for symptoms of neonatal opioid withdrawal syndrome(Prod Info DOLOPHINE(R) oral tablets, 2014).
    3) Methadone crosses the placenta and may produce respiratory depression in neonates. Therefore, it is not for use during or immediately preceding labor. Shorter acting analgesics and techniques are more appropriate (Prod Info DOLOPHINE(R) oral tablets, 2014)
    B) SPECIFIC AGENT
    1) METHADONE MAINTENANCE IN PREGNANCY
    a) To determine any dose-related neonatal effects from maternal use of methadone, researchers retrospectively compared 70 pregnancies historically monitored by a perinatal intervention program for pregnant women with a history of drug abuse. Nearly two-thirds of the pregnancies were maintained on methadone (median daily dose 20 mg; range 0 to 150 mg), while the remainder opted for detoxification. Methadone dose did not correlate with gestational age at delivery, preterm birth, birth weight, cesarean delivery, meconium staining of the amniotic fluid, or low Apgar score; increasing doses were, however, significantly associated with birth weight below the tenth percentile and positive neonatal toxicology screen for opioids or cocaine (all P values less than 0.05). Among the 46% of the neonates treated for withdrawal, nearly all of the infants were born to mothers taking at least 40 mg of methadone daily. Pregnant mothers who supplemented with heroin delivered infants more likely to require treatment for withdrawal (Dashe et al, 2002).
    b) In a study of pregnant women receiving methadone maintenance, similar birth outcomes (birth weight, obstetric complications, and neonatal morbidity and mortality) were observed as pregnant women using cocaine. Three neonates in the methadone group had major congenital anomalies with 2 of the 3 resulting in mortality. The authors attributed this finding to ongoing abuse of illicit drugs in this patient population throughout pregnancy (Brown et al, 1998).
    c) When compared with a control group matched for sex, gestational age, and similar toxicologic exposure, 42 neonates born to mothers in a methadone maintenance program showed increased risks for smaller head circumference, lower birth weight, and shorter length (p less than 0.01 for each measure) (Arevalo et al, 2003).
    d) Eight deaths were reported for infants younger than 1 year and whose mothers were on methadone maintenance therapy in a retrospective review of Canadian databases from January 2006 through December 2010. In these 8 infants, there was a significant increase in both right and left lung weight at the time of autopsy as compared with published age-specific reference organ weights (p=0.035, p=0.007, respectively) (Kelly et al, 2012).
    e) Use of narcotic analgesics and/or methadone during pregnancy, particularly at high doses or for prolonged periods, has been associated with adverse effects in the neonate; reported effects have included physical dependence/withdrawal, intrauterine growth retardation, and respiratory depression (Cunningham et al, 1993).
    f) Similar results had previously been shown in infants born to methadone-exposed mothers compared with infants born to non-drug exposed mothers matched for race, age and socioeconomic status; however, at 6 months of age there was no statistically significant difference in mental development among the infants (Kaltenbach & Finnegan, 1987).
    2) METHADONE TREATMENT PROGRAMS -
    a) In a study of methadone use during pregnancy, neonatal withdrawal was more infrequent in mothers who participated in a methadone detoxification program during pregnancy than in those who practiced uncontrolled opiate abuse until delivery (Maas et al, 1990).
    3) NEONATAL ABSTINENCE SYNDROME
    a) A double-blind, double-dummy, flexible-dosing, randomized, controlled study of 131 pregnant women exposed to buprenorphine or methadone (n=58 and n=73, respectively) did not demonstrate a significant difference in the percentage of neonates in need of neonatal abstinence syndrome (NAS) treatment, peak NAS scores, or neonate head circumference between treatment groups. There was a significant 89% decrease in the total amount of morphine required to treat NAS in neonates exposed to buprenorphine compared with neonates exposed to methadone. In addition, neonates exposed to buprenorphine spent, on average, a significant 43% less time in the hospital when compared with neonates exposed to methadone. The significance of the results did not vary when the analysis was adjusted for selected covariates. An analysis for secondary outcomes revealed neonates exposed to buprenorphine required, on average, a significant 58% less time in the hospital receiving treatment for NAS when compared with neonates exposed to methadone. There were no significant differences between the 2 treatment groups in the incidence of maternal and/or neonatal adverse effects (Jones et al, 2010).
    b) In a prospective, cohort study of pregnant women enrolled in Norwegian opioid maintenance treatment programs (n=38), neither the incidence of neonatal abstinence syndrome (NAS) nor the duration of NAS treatment was associated with the maternal dose of methadone (n=26) or buprenorphine (n=12). NAS was reported in 58% and 67% of infants following maternal methadone and buprenorphine use, respectively. NAS treatment lasted for a mean of 43 and 37 days in the methadone- and buprenorphine-exposed groups, respectively. The maternal daily dose of methadone and buprenorphine (mean of 90 mg and 13.3 mg, respectively, before delivery) did not significantly correlate with duration of NAS treatment (p=0.054 and p=0.31, respectively). There were also no significant differences in occurrence (p=0.73) or duration (p=0.64) of NAS treatment between the methadone and buprenorphine groups (Bakstad et al, 2009).
    4) NEONATAL WITHDRAWAL SYMPTOMS
    a) Methadone use during pregnancy has been associated with neonatal withdrawal symptoms, which have occurred following as few as 10 days of methadone therapy, and as soon as 3 hours after birth (Rahbar, 1975; Rajegowda et al, 1972; Pierson et al, 1972). Symptoms reportedly have persisted 7 to 10 days, and included tremor, irritability, hyperactivity, jitteriness, shrill cry, vomiting, diarrhea, sustained ankle clonus, hyperthermia, hypertoxicity, and convulsions (Rahbar, 1975; Rajegowda et al, 1972). Sudden death in infants born to methadone-maintained mothers have been reported (Pierson et al, 1972).
    5) SUDDEN INFANT DEATH SYNDROME
    a) The evidence of an increased incidence of sudden infant death syndrome in infants of methadone-treated women is conflicting. Abnormal fetal non-stress tests have occurred more than in control groups, when tested between 1 and 2 hours after a maintenance dose of methadone in late pregnancy (Prod Info DOLOPHINE(R) oral tablets, 2014).
    6) FETAL/NEONATAL ADVERSE REACTIONS
    a) In a prospective cohort study, poorer neurodevelopment at 6 months of age was observed in infants (n=81) exposed to methadone in utero compared with non-exposed infants (n=26). On Griffiths Mental Development Scale assessment, 8 (9.9%) of methadone exposed infants scored less than 85 compared with 100% of non-exposed infants scoring greater than or equal to 95. Of these 8 infants, 6 had one or more coexisting visual problem, including reduced visual acuity which was reported in all 6 infants. Results for infants requiring treatment for neonatal abstinence syndrome (NAS) were significantly poorer compared with infants that did not require treatment. Additionally, exposure to multiple illicit drugs in utero resulted in poorer development, particularly locomotor development and hand-eye skills. Development in accommodated infants (n=20) was poorer compared with infants still in the care of their birth parents; however, accommodated infants were likely to have received treatment for NAS and been exposed to multiple drugs in utero (McGlone & Mactier, 2015).
    7) CONDUCTION DISTURBANCES
    a) CASE REPORT: A 21-year-old primiparous woman on methadone maintenance therapy (50 mg/day) delivered a term baby in good condition except for noted bradycardia (heart rate, 80 to 90 beats/min (bpm)). No other cardiovascular complications were noted. An ECG revealed a corrected QT interval (QTc) prolongation of 510 msec (newborn upper limit 440 msec). The infant also experienced mild withdrawal symptoms which required supportive care. Several ECG's were performed and the QT interval progressively shortened. On day 5, the QTc was 380 msec and the infant's heart rate also improved to 120 to 140 bpm. At two months of age, the infant was healthy with a normal QTc interval (Hussain & Ewer, 2007).
    8) EFFECTS ON PREGNANCY
    a) Methadone elimination is significantly greater during pregnancy, according to a study of approximately 30 pregnant women being treated with oral methadone during the second and third trimesters. Increased body clearance of methadone was demonstrated in pregnant patients compared with the same patients postpartum or with non-pregnant opioid-dependent women. During the second and third trimesters, terminal half-life of methadone was decreased. The resulting lower methadone trough levels during pregnancy may lead to withdrawal symptoms in some pregnant patients (Prod Info DOLOPHINE(R) oral tablets, 2014; Prod Info DISKETS(R) dispersible tablets for oral suspension, 2007).
    C) LACK OF EFFECT
    1) A retrospective review of pregnant, opiate-dependent women (n=101) who received inpatient opiate detoxification with methadone did not find an increased risk in second-trimester miscarriage or third-trimester premature delivery (Prod Info DOLOPHINE(R) oral tablets, 2014; Prod Info methadone hydrochloride oral solution, 2008).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) Methadone is excreted into human breast milk. Maternal doses of 10 to 80 mg/day have resulted in methadone milk concentrations of 50 to 570 mcg/L, which were lower than maternal steady-state serum levels. Peak methadone concentrations occur approximately 4 to 5 hours following an oral dose. Based on an average milk consumption of 150 mL/kg/day, a nursing infant would ingest approximately 17.4 mcg/kg/day (2% to 3%) of the oral maternal methadone dose (Prod Info DOLOPHINE(R) oral tablets, 2014; Prod Info DISKETS(R) dispersible tablets for oral suspension, 2007; Prod Info METHADOSE(TM) oral concentrate, sugar-free oral concentrate, 2007; Prod Info DOLOPHINE(R) HYDROCHLORIDE oral tablets, 2006). Exercise caution when administering methadone to a nursing woman. Reports of sedation and respiratory depression have occurred in infants exposed to methadone through breast milk. Mothers should be instructed on how to recognize respiratory depression and/or sedation in their infants. For women who are already breastfeeding, who are to be treated with methadone, they should be advised on how to gradually wean their babies to prevent withdrawal symptoms in the infant (Prod Info DOLOPHINE(R) oral tablets, 2014; Prod Info DISKETS(R) dispersible tablets for oral suspension, 2007).
    2) Therapeutic doses of methadone are considered to be compatible with breastfeeding by the American Academy of Pediatrics (Anon, 2001).
    3) Based upon 12 lactating women who were taking daily doses of methadone from 20 to 80 mg, nursing infants received a mean of 2.79% of the maternal dose (Wojnar-Horton et al, 1997).
    4) Maintenance doses of methadone 10 to 40 mg/day produced low breast milk concentrations of 50 to 170 ng/mL; doses of 50 to 80 mg/day produced concentrations of 230 to 570 ng/mL (Prod Info DOLOPHINE(R) oral tablets, 2014; Blinick et al, 1975).
    5) The death of a nursing infant was attributed to methadone (plasma concentration 0.04 mg/dL), presumably ingested during breast feeding. Malnutrition and abnormalities of liver and brain were other possible causes of death (Smialek et al, 1977).
    B) LACK OF EFFECT
    1) In a study describing 12 lactating women (mean age of 27.2 years) treated with a mean single daily methadone dose of 75.8 mg (range, 40 to 110 mg) that was initiated 1 month prior to delivery, the infant plasma concentrations of methadone were small in daily breast milk samples submitted on days 1 through 4 of nursing. Of the 12 women, three women received one psychotropic medication during pregnancy and one woman received two medications (sertraline (2), fluoxetine (2), and olanzapine (1)). All 12 infants had appropriate birth weights, head circumference measurements, and Apgar scores for their gestational ages; one infant was hospitalized for 10 days, 6 of which were for the treatment of mild to moderate neonatal abstinence syndrome (NAS). Breast milk to plasma (BM/P) methadone concentration ratios ranged from 0.1 to 2.7 at trough (just prior to daily methadone dose) and 0.1 to 1.2 at peak (3 hours after daily methadone dose). Although there was a significant increase in methadone concentration in breast milk over time for the peak post breastfeeding time (t(22)=2.4), methadone concentrations in breast milk ranged from 20.6 to 314.2 nanograms (ng)/mL and were not related to maternal dose. No significant correlations between maternal methadone dose and maternal plasma concentrations were demonstrated for peak or trough samples at any time. Authors suggest that these results support the use of methadone, regardless of dose, in nursing mothers (Jansson et al, 2007).
    3.20.5) FERTILITY
    A) SEMEN ABNORMAL
    1) In males treated with methadone, reduced ejaculate volume and seminal vesicle and prostate secretions have been observed. There have also been reports of decreased serum testosterone levels and sperm motility, and abnormalities in sperm morphology (Prod Info DOLOPHINE(R) oral tablets, 2014; Prod Info DISKETS(R) dispersible tablets for oral suspension, 2007; Prod Info METHADOSE(TM) oral concentrate, sugar-free oral concentrate, 2007).
    B) ANIMAL STUDIES
    1) Animal studies have demonstrated that methadone treatment of males can alter reproductive function. In male mice and rats, methadone has produced significant regression of sex accessory organs and testes. Embryolethality and neonatal mortality were increased after male rats were treated with methadone once daily for three consecutive days (Prod Info DOLOPHINE(R) oral tablets, 2014; Prod Info DISKETS(R) dispersible tablets for oral suspension, 2007; Prod Info METHADOSE(TM) oral concentrate, sugar-free oral concentrate, 2007).
    2) When uterine contents of methadone-naive female mice that were bred with methadone-treated mice were examined, the findings indicated that methadone treatment increased the rate of preimplantation deaths in all post-meiotic states (Prod Info DISKETS(R) dispersible tablets for oral suspension, 2007; Prod Info METHADOSE(TM) oral concentrate, sugar-free oral concentrate, 2007; Prod Info DOLOPHINE(R) HYDROCHLORIDE oral tablets, 2006).

Carcinogenicity

    3.21.4) ANIMAL STUDIES
    A) LACK OF EFFECT
    1) LAAM - Two-year carcinogenicity studies in rats and mice given oral doses of LAAM, 13 mg/kg and 30 mg/kg, respectively, revealed NO carcinogenic changes (Prod Info Orlaam(R), 2000).

Genotoxicity

    A) LAAM - No mutagenic activity was apparent in several mutagenic tests (Ames, unscheduled DNA synthesis and repair test mouse lymphoma cells in vitro, or chromosomal aberration tests in rats in vivo); LAAM was positive in a forward mutation assay in N. crassa, the significance of which is unknown (Prod Info Orlaam(R), 2000).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor vital signs and mental status carefully. Continuous pulse oximetry monitoring, and capnometry if available.
    B) Obtain a 12-lead ECG, and institute continuous cardiac monitoring in patients with moderate to severe toxicity.
    C) Monitor bed side blood glucose.
    D) Obtain a chest x-ray if aspiration or pulmonary edema is suspected.
    E) Methadone is not detected on most opioid urine drug screens.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) Monitor CPK in patients with prolonged coma or repeated seizure activity.
    B) TOXICITY
    1) Routine laboratory testing for opiates will not detect methadone. It is necessary to order a special toxicology screen for detection of methadone in urine, serum, or blood. These tests have reported sensitivities of 10 to 300 ng/mL(Schwab & Caggiano, 2001).
    4.1.4) OTHER
    A) OTHER
    1) ECG
    a) Obtain a 12-lead ECG, and institute continuous cardiac monitoring in patients with moderate to severe toxicity. QTc interval prolongation and torsades de pointes have been reported with therapeutic methadone use.
    b) Periodic ECGs have been recommended in patients receiving methadone in doses exceeding 100 mg/day, or who have unexplained syncope or seizures (Krantz et al, 2009).
    2) MONITORING
    a) Continuous pulse oximetry monitoring, and capnometry if available. Monitor arterial blood gases and chest x-ray in patients with evidence of pulmonary edema.
    b) Monitor bed side blood glucose.
    3) IMMUNOASSAY INTERFERENCE
    a) A 9-year-old boy who was being treated with diphenhydramine for nasal congestion and difficulty sleeping, presented with an altered level of consciousness. An immunoassay toxicological screen (One Step Multi-Drug, Multi-Line Screen Test Device, ACON Laboratories) was positive for methadone, however, his parents denied any risk of drug or toxin exposure except for the diphenhydramine. Gas chromatography/mass spectrometry of the patient's urine did not show the presence of methadone (Rogers et al, 2010).

Radiographic Studies

    A) CHEST RADIOGRAPH
    1) Chest x-ray should be obtained in symptomatic patients for detection of possible acute lung injury following an overdose.
    B) MRI
    1) Methadone-induced acute toxic leukoencephalopathy has been reported in several patients (Metkees et al, 2015; Gheuens et al, 2010; Zanin et al, 2010; Anselmo et al, 2006). In MRI brain imaging, methadone-induced acute leukoencephalopathy usually affects the white matter more than the gray matter. This would allow the clinician to differentiate it from hypoxic injury or other toxicities (eg, carbon monoxide) that more commonly affect gray matter (Metkees et al, 2015).

Methods

    A) MULTIPLE ANALYTICAL METHODS
    1) Morphine-specific opiate immunoassay screening tests cannot detect methadone or its metabolite, 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP). Immunoassays such as DRI methadone assay (Microgenics Corp, Freemont, CA) may be used to detect methadone. False positive results have been reported when tapentadol reacted with the DRI methadone assay; however, cross-reactivity in the assay is low (Milone, 2012).
    2) Semiquantitative and qualitative EMIT(R) homogeneous enzyme immunoassays are available for measurement of the class of opioids in urine. The assays detect methadone and other opioids (morphine, morphine glucuronide, codeine, and hydromorphone, and higher concentrations of nalorphine and meperidine). The assays do NOT detect long-acting methadone, L-alpha-acetyl-methadol (LAAM), or its metabolites. CDC proficiency testing and clinical studies show the EMIT(R) method to correlate well with GC, GLC, HPLC, RIA, and TLC. Adulterants may also cause false negatives (Mikkelsen & Ash, 1988). Crane et al (1993) reported false negative readings for opiates when the drug, mefenamic acid, has been ingested prior to an EMIT urine drug assay (Crane et al, 1993).
    3) Rogers et al (1997) described the use of GC/MS method to detect methadone in three cases of methadone/benzodiazepine overdoses. Additionally, alprazolam was only detected by modification of an existing HPLC procedure (Rogers et al, 1997).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) All patients who develop CNS or respiratory depression should be admitted for observation (for at least 24 hours) even after adequate response to naloxone therapy. Patients who required intubation or a naloxone infusion require an intensive care unit admission. Patients should not be discharged until they have remained awake and alert for 4 to 6 hours after the naloxone infusion has been discontinued.
    6.3.1.2) HOME CRITERIA/ORAL
    A) All patients with suspected methadone overdose (deliberate or inadvertent) should be referred to a healthcare facility.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Consult a poison center or medical toxicologist for assistance in managing patients with severe toxicity, or in whom the diagnosis is not clear.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Patients with mild toxicity who do not require naloxone should be observed for at least 8 hours. All children who have ingested any amount of methadone need to be observed in an Emergency Department (ED) for at least 8 hours and considered for hospital admission.

Monitoring

    A) Monitor vital signs and mental status carefully. Continuous pulse oximetry monitoring, and capnometry if available.
    B) Obtain a 12-lead ECG, and institute continuous cardiac monitoring in patients with moderate to severe toxicity.
    C) Monitor bed side blood glucose.
    D) Obtain a chest x-ray if aspiration or pulmonary edema is suspected.
    E) Methadone is not detected on most opioid urine drug screens.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) GI decontamination is not recommended because of the risk of aspiration with lose of airway protection and risk of seizure. .
    6.5.2) PREVENTION OF ABSORPTION
    A) Administer activated charcoal if the patient present early after large ingestion with appropriate level of consciousness, patent airway, and can drink the charcoal.
    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) NALOXONE
    1) NALOXONE/SUMMARY
    a) Naloxone, a pure opioid antagonist, reverses coma and respiratory depression from all opioids. It has no agonist effects and can safely be employed in a mixed or unknown overdose where it can be diagnostic and therapeutic without risk to the patient.
    b) Indicated in patients with mental status and respiratory depression possibly related to opioid overdose (Hoffman et al, 1991).
    c) DOSE: The initial dose of naloxone should be low (0.04 to 0.4 mg) with a repeat dosing as needed or dose escalation to 2 mg as indicated due to the risk of opioid withdrawal in an opioid-tolerant individual; if delay in obtaining venous access, may administer subcutaneously, intramuscularly, intranasally, via nebulizer (in a patient with spontaneous respirations) or via an endotracheal tube (Vanden Hoek,TL,et al).
    d) Recurrence of opioid toxicity has been reported to occur in approximately 1 out of 3 adult ED opioid overdose cases after a response to naloxone. Recurrences are more likely with long-acting opioids (Watson et al, 1998)
    2) NALOXONE DOSE/ADULT
    a) INITIAL BOLUS DOSE: Because naloxone can produce opioid withdrawal in an opioid-dependent individual leading to severe agitation and hypertension, the initial dose of naloxone should be low (0.04 to 0.4 mg) with a repeat dosing as needed or dose escalation to 2 mg as indicated (Vanden Hoek,TL,et al).
    1) This dose can also be given intramuscularly or subcutaneously in the absence of intravenous access (Howland & Nelson, 2011; Prod Info naloxone HCl IV, IM, subcutaneous injection solution, 2008; Maio et al, 1987; Wanger et al, 1998).
    b) Larger doses may be needed to reverse opioid effects. Generally, if no response is observed after 8 to 10 milligrams has been administered, the diagnosis of opioid-induced respiratory depression should be questioned (Howland & Nelson, 2011; Prod Info naloxone HCl IV, IM, subcutaneous injection solution, 2008). Very large doses of naloxone (10 milligrams or more) may be required to reverse the effects of a buprenorphine overdose (Gal, 1989; Jasinski et al, 1978).
    1) Single doses of up to 24 milligrams have been given without adverse effect (Evans et al, 1973).
    c) REPEAT DOSE: The effective naloxone dose may have to be repeated every 20 to 90 minutes due to the much longer duration of action of the opioid agonist used(Howland & Nelson, 2011).
    1) OPIOID DEPENDENT PATIENTS: The goal of naloxone therapy is to reverse respiratory depression without precipitating significant withdrawal. Starting doses of naloxone 0.04 mg IV, or 0.001 mg/kg, have been suggested as appropriate for opioid-dependent patients without severe respiratory depression (Howland & Nelson, 2011). If necessary the dose may be repeated or increased gradually until the desired response is achieved (adequate respirations, ability to protect airway, responds to stimulation but no evidence of withdrawal) (Howland & Nelson, 2011). In the presence of opioid dependence, withdrawal symptoms typically appear within minutes of naloxone administration and subside in about 2 hours. The severity and duration of the withdrawal syndrome are dependant upon the naloxone dose and the degree and type of dependence.(Prod Info naloxone HCl IV, IM, subcutaneous injection solution, 2008)
    2) PRECAUTION should be taken in the presence of a mixed overdose of a sympathomimetic with an opioid. Administration of naloxone may provoke serious sympathomimetic toxicity by removing the protective opioid-mediated CNS depressant effects. Arrhythmogenic effects of naloxone may also be potentiated in the presence of severe hyperkalemia (McCann et al, 2002).
    d) NALOXONE DOSE/CHILDREN
    1) LESS THAN 5 YEARS OF AGE OR LESS THAN 20 KG: 0.1 mg/kg IV/intraosseous/IM/subcutaneously maximum dose 2 mg; may repeat dose every 2 to 5 minutes until symptoms improve (Kleinman et al, 2010; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008)
    2) 5 YEARS OF AGE OR OLDER OR GREATER THAN 20 KG: 2 mg IV/intraosseous/IM/subcutaneouslymay repeat dose every 2 to 5 minutes until symptoms improve (Kleinman et al, 2010; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Krauss & Green, 2006). Although naloxone may be given via the endotracheal tube for pediatric resuscitation, optimal doses are unknown. Some experts have recommended using 2 to 3 times the IV dose (Kleinman et al, 2010)
    3) AVOIDANCE OF OPIOID WITHDRAWAL: In cases of known or suspected chronic opioid therapy, a lower dose of 0.01 mg/kg may be considered and titrated to effect to avoid withdrawal: INITIAL DOSE: 0.01 mg/kg body weight given IV. If this does not result in clinical improvement, an additional dose of 0.1 mg/kg body weight may be given. It may be given by the IM or subQ route if the IV route is not available (Prod Info naloxone HCl IV, IM, subcutaneous injection solution, 2008)
    e) NALOXONE DOSE/NEONATE
    1) The American Academy of Pediatrics recommends a neonatal dose of 0.1 mg/kg IV or intratracheally from birth until age 5 years or 20 kilograms of body weight (AAP, 1989; Kleinman et al, 2010).
    2) Smaller doses (10 to 30 mcg/kg IV) have been successful in the setting of exposure via maternal administration of narcotics or administration to neonates in therapeutic doses for anesthesia (Wiener et al, 1977; Welles et al, 1984; Fischer & Cook, 1974; Brice et al, 1979).
    3) POTENTIAL OF WITHDRAWAL: The risk of precipitating withdrawal in an addicted neonate should be considered. Withdrawal seizures have been provoked in infants from opioid-abusing mothers when the infants were given naloxone at birth to stimulate breathing (Gibbs et al, 1989).
    4) In cases of inadvertent administration of an opioid overdose to a neonate, larger doses may be required. In one case of oral morphine intoxication, 0.16 milligram/kilogram/hour was required for 5 days (Tenenbein, 1984).
    f) NALOXONE/ALTERNATE ROUTES
    1) If intravenous access cannot be rapidly established, naloxone can be administered via subcutaneous or intramuscular injection, intranasally, or via inhaled nebulization in patients with spontaneous respirations.
    2) INTRAMUSCULAR/SUBCUTANEOUS ROUTES: If an intravenous line cannot be secured due to hypoperfusion or lack of adequate veins then naloxone can be administered by other routes.
    3) The intramuscular or subcutaneous routes are effective if hypoperfusion is not present (Prod Info naloxone HCl IV, IM, subcutaneous injection solution, 2008). The delay required to establish an IV, offsets the slower rate of subcutaneous absorption (Wanger et al, 1998).
    4) Naloxone Evzio(TM) is a hand-held autoinjector intended for the emergency treatment of known or suspected opioid overdose. The autoinjector is equipped with an electronic voice instruction system to assist caregivers with administration. It is available as 0.4 mg/0.4 mL solution for injection in a pre-filled auto-injector (Prod Info EVZIO(TM) injection solution, 2014).
    5) INTRANASAL ROUTE: Intranasal naloxone has been shown to be effective in opioid overdose; bioavailability appears similar to the intravenous route (Kelly & Koutsogiannis, 2002). Based on several case series of patients with suspected opiate overdose, the average response time of 3.4 minutes was observed using a formulation of 1 mg/mL/nostril by a mucosal atomization device (Kerr et al, 2009; Kelly & Koutsogiannis, 2002). However, a young adult who intentionally masticated two 25 mcg fentanyl patches and developed agonal respirations (6 breaths per minute), decreased mental status and mitotic pupils did not respond to intranasal naloxone (1 mg in each nostril) administered by paramedics. After 11 minutes, paramedics placed an IV and administered 1 mg of IV naloxone; respirations normalized and mental status improved. Upon admission, 2 additional doses of naloxone 0.4 mg IV were needed. The patient was monitored overnight and discharged the following day without sequelae. Its suggested that intranasal administration can lead to unpredictable absorption (Zuckerman et al, 2014).
    a) Narcan(R) nasal spray is supplied as a single 4 mg dose of naloxone hydrochloride in a 0.1 mL intranasal spray (Prod Info NARCAN(R) nasal spray, 2015).
    b) FDA DOSING: Initial dose: 1 spray (4 mg) intranasally into 1 nostril. Subsequent doses: Use a new Narcan(R) nasal spray and administer into alternating nostrils. May repeat dose every 2 to 3 minutes. Requirement for repeat dosing is dependent on the amount, type, and route of administration of the opioid being antagonized. Higher or repeat doses may be required for partial agonists or mixed agonist/antagonists (Prod Info NARCAN(R) nasal spray, 2015).
    c) AMERICAN HEART ASSOCIATION GUIDELINE DOSING: Usual dose: 2 mg intranasally as soon as possible; may repeat after 4 minutes (Lavonas et al, 2015). Higher doses may be required with atypical opioids (VandenHoek et al, 2010).
    d) ABSORPTION: Based on limited data, the absorption rate of intranasal administration is comparable to intravenous administration. The peak plasma concentration of intranasal administration is estimated to be 3 minutes which is similar to the intravenous route (Kerr et al, 2009). In rare cases, nasal absorption may be inhibited by injury, prior use of intranasal drugs, or excessive secretions (Kerr et al, 2009).
    6) NEBULIZED ROUTE: DOSE: A suggested dose is 2 mg naloxone with 3 mL of normal saline for suspected opioid overdose in patients with some spontaneous respirations (Weber et al, 2012).
    7) ENDOTRACHEAL ROUTE: Endotracheal administration of naloxone can be effective(Tandberg & Abercrombie, 1982), optimum dose unknown but 2 to 3 times the intravenous dose had been recommended by some (Kleinman et al, 2010).
    g) NALOXONE/CONTINUOUS INFUSION METHOD
    1) A continuous infusion of naloxone may be employed in circumstances of opioid overdose with long acting opioids (Howland & Nelson, 2011; Redfern, 1983).
    2) The patient is given an initial dose of IV naloxone to achieve reversal of opioid effects and is then started on a continuous infusion to maintain this state of antagonism.
    3) DOSE: Utilize two-thirds of the initial naloxone bolus on an hourly basis (Howland & Nelson, 2011; Mofenson & Caraccio, 1987). For an adult, prepare the dose by multiplying the effective bolus dose by 6.6, and add that amount to 1000 mL and administer at an IV infusion rate of 100 mL/hour (Howland & Nelson, 2011).
    4) Dose and duration of action of naloxone therapy varies based on several factors; continuous monitoring should be used to prevent withdrawal induction (Howland & Nelson, 2011).
    5) Observe patients for evidence of CNS or respiratory depression for at least 2 hours after discontinuing the infusion (Howland & Nelson, 2011).
    h) NALOXONE/PREGNANCY
    1) In general, the smallest dose of naloxone required to reverse life threatening opioid effects should be used in pregnant women. Naloxone detoxification of opioid addicts during pregnancy may result in fetal distress, meconium staining and fetal death (Zuspan et al, 1975). When naloxone is used during pregnancy, opioid abstinence may be provoked in utero (Umans & Szeto, 1985).
    3) CASE REPORTS
    a) Patients with methadone overdose may require prolonged administration of naloxone and careful observation for relapse after naloxone is discontinued.
    b) NEBULIZED NALOXONE: A 46-year-old woman in a methadone maintenance program who was taking methadone 75 mg daily (525 mg weekly), presented with lethargy and respiratory depression after ingesting an unknown amount of methadone. She had a medical history of HIV and chronic obstructive pulmonary disease. Because of scarred peripheral veins, it was difficult to obtain a venous access. She received 2 mg of nebulized naloxone (mixed with 3 mL normal saline via nebulizer face mask) and her condition improved within 5 minutes of therapy. A second dose of nebulized naloxone was administered after she became lethargic again 55 minutes after the first dose of naloxone. Once again, her condition improved rapidly. No opioid withdrawal was noted after each nebulized naloxone dose. In the ICU, she was treated with an IV naloxone infusion (2 mg/hr) after the patient's oxygen saturation decreased and respirations became shallow 50 minutes after the second dose of naloxone. She experienced severe opioid withdrawal 5 minutes later and developed pulmonary edema. Following further symptomatic therapy, she gradually recovered and was discharged a week later (Mycyk et al, 2003).
    c) CASE REPORT: A 22-year-old man died approximately 44 hours following an acute methadone overdose. His naloxone infusion had been discontinued about 3 hours prior to his death because he appeared to be oriented and stabilized. Due to the long half-life of methadone, his respiratory rate and mental status again declined, resulting in death (Hendra et al, 1996).
    d) CASE REPORT: A 13-year-old child who overdosed on methadone required a cumulative dose of 0.65 mg/kg naloxone over 65.5 hours of continuous infusion and recovered (Romac, 1986).
    B) NALMEFENE
    1) NALMEFENE: The recommended dose for opioid toxicity in a non-opioid dependent patient is 0.5 mg/70 kg slow intravenously, intramuscularly or subcutaneously. This may be followed by a second dose 2 to 5 minutes later. If after following a total dose of 1.5 mg/70 kg, no clinical response has been achieved, it is unlikely that further doses will have an effect. Do not administer more than is required to restore the respiratory rate to normal, in order to minimize the likelihood of cardiovascular stress and precipitated withdrawal syndrome (Prod Info Revex(R), nalmefene, 1998).
    a) The half-life of methadone exceeds the half-life of nalmefene; prolonged monitoring (even if the patient responds to the initial dose) is indicated since the patient could have recurrent somnolence after the nalmefene wears off.
    C) NALTREXONE
    1) Naltrexone is NOT recommended for methadone overdoses in an opiate-dependent person due to possible precipitation of opiate withdrawal. Naloxone is recommended in these patients. Conservative management with naltrexone has been reported in methadone overdose in an opiate-naive patient. An adult male inadvertently received 125 mg of methadone. One hour after the overdose, he was given 100 mg of oral naltrexone, followed by another 100 mg about 10 hours later. The patient remained asymptomatic and was discharged the following day (Dhopesh et al, 2002).
    D) MONITORING OF PATIENT
    1) Monitor vital signs and mental status carefully. Continuous pulse oximetry monitoring, and capnometry if available.
    2) Obtain a 12-lead ECG, and institute continuous cardiac monitoring in patients with moderate to severe toxicity.
    3) Monitor bed side blood glucose.
    4) Obtain a chest x-ray if aspiration or pulmonary edema is suspected.
    5) Methadone is not detected on most opioid urine drug screens.
    E) CONTRAINDICATED TREATMENT
    1) Nalorphine and levallorphan have agonist and antagonist properties including respiratory depression. They should no longer be used because if it is not an opioid that is being treated, CNS depression may ensue.
    F) ACUTE LUNG INJURY
    1) Noninvasive ventilation or endotracheal intubation may be necessary in patients who develop pulmonary edema. Consider extracorporeal membrane oxygenation to treat pulmonary edema refractory to standard measures.
    a) CASE REPORT: A 20-year-old man who was found unconscious at home after ingesting an unknown amount of methadone, in addition to drinking 5 beers and smoking cannabis, regained consciousness and had increased respiratory rate, persistent hypoxia and respiratory distress after prehospital naloxone therapy. Chest radiograph showed bilateral alveolar and interstitial infiltrates and echocardiography showed no cardiac function impairment, suggesting noncardiogenic pulmonary edema. He was successfully treated with a naloxone infusion and noninvasive ventilation (Gonzva et al, 2013).
    2) ONSET: Onset of acute lung injury after toxic exposure may be delayed up to 24 to 72 hours after exposure in some cases.
    3) NON-PHARMACOLOGIC TREATMENT: The treatment of acute lung injury is primarily supportive (Cataletto, 2012). Maintain adequate ventilation and oxygenation with frequent monitoring of arterial blood gases and/or pulse oximetry. If a high FIO2 is required to maintain adequate oxygenation, mechanical ventilation and positive-end-expiratory pressure (PEEP) may be required; ventilation with small tidal volumes (6 mL/kg) is preferred if ARDS develops (Haas, 2011; Stolbach & Hoffman, 2011).
    a) To minimize barotrauma and other complications, use the lowest amount of PEEP possible while maintaining adequate oxygenation. Use of smaller tidal volumes (6 mL/kg) and lower plateau pressures (30 cm water or less) has been associated with decreased mortality and more rapid weaning from mechanical ventilation in patients with ARDS (Brower et al, 2000). More treatment information may be obtained from ARDS Clinical Network website, NIH NHLBI ARDS Clinical Network Mechanical Ventilation Protocol Summary, http://www.ardsnet.org/node/77791 (NHLBI ARDS Network, 2008)
    4) FLUIDS: Crystalloid solutions must be administered judiciously. Pulmonary artery monitoring may help. In general the pulmonary artery wedge pressure should be kept relatively low while still maintaining adequate cardiac output, blood pressure and urine output (Stolbach & Hoffman, 2011).
    5) ANTIBIOTICS: Indicated only when there is evidence of infection (Artigas et al, 1998).
    6) EXPERIMENTAL THERAPY: Partial liquid ventilation has shown promise in preliminary studies (Kollef & Schuster, 1995).
    7) CALFACTANT: In a multicenter, randomized, blinded trial, endotracheal instillation of 2 doses of 80 mL/m(2) calfactant (35 mg/mL of phospholipid suspension in saline) in infants, children, and adolescents with acute lung injury resulted in acute improvement in oxygenation and lower mortality; however, no significant decrease in the course of respiratory failure measured by duration of ventilator therapy, intensive care unit, or hospital stay was noted. Adverse effects (transient hypoxia and hypotension) were more frequent in calfactant patients, but these effects were mild and did not require withdrawal from the study (Wilson et al, 2005).
    8) However, in a multicenter, randomized, controlled, and masked trial, endotracheal instillation of up to 3 doses of calfactant (30 mg) in adults only with acute lung injury/ARDS due to direct lung injury was not associated with improved oxygenation and longer term benefits compared to the placebo group. It was also associated with significant increases in hypoxia and hypotension (Willson et al, 2015).
    9) EXTRACORPOREAL MEMBRANE OXYGENATION (ECMO): In separate cases, a 16-year-old boy and a 19-year-old woman with a history of illicit drug use were found unresponsive and hypotensive with O2 saturations of 50% and 60% (respectively) after intentionally ingesting an unknown amount of methadone. Administrations of naloxone were ineffective and hypoxia persisted (PaO2 40 mmHg and 35 mmHg) despite treatment with nitric oxide and high-frequency oscillatory ventilation. Pulmonary edema was confirmed with chest radiograph in both cases. ECMO was initiated in each case with a cardiac output of 4 to 5 L/min and continued for 3 days. Both individuals were discharged within 3 to 5 days of admission without serious complications (Daugherty, 2011).
    G) SEIZURE
    1) Correct hypoxia and administer naloxone. If seizures persist or recur, anticonvulsants may be needed.
    2) 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).
    3) 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 .
    4) 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).
    5) 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, 2009; Chin et al, 2008).
    6) 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).
    7) 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).
    H) 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).
    I) PROLONGED QT INTERVAL
    1) METHADONE
    a) Obtain serial ECGs following a significant exposure, institute continuous cardiac monitoring.
    b) In a limited number of studies QTc interval prolongation has been reported in patients receiving therapeutic doses of methadone. In some cases, a correlation between high-dose therapy and QTc prolongation was observed. Torsades has been observed infrequently (Ehret et al, 2006; Lamont & Hunt, 2006).
    2) SUMMARY
    a) Withdraw the causative agent. Hemodynamically unstable patients with Torsades de pointes (TdP) require electrical cardioversion. Emergent treatment with magnesium (first-line agent) or atrial overdrive pacing is indicated. Detect and correct underlying electrolyte abnormalities (ie, hypomagnesemia, hypokalemia, hypocalcemia). Correct hypoxia, if present (Drew et al, 2010; Neumar et al, 2010; Keren et al, 1981; Smith & Gallagher, 1980).
    b) Polymorphic VT associated with acquired long QT syndrome may be treated with IV magnesium. Overdrive pacing or isoproterenol may be successful in terminating TdP, particularly when accompanied by bradycardia or if TdP appears to be precipitated by pauses in rhythm (Neumar et al, 2010). In patients with polymorphic VT with a normal QT interval, magnesium is unlikely to be effective (Link et al, 2015).
    3) MAGNESIUM SULFATE
    a) Magnesium is recommended (first-line agent) for the prevention and treatment of drug-induced torsades de pointes (TdP) even if the serum magnesium concentration is normal. QTc intervals greater than 500 milliseconds after a potential drug overdose may correlate with the development of TdP (Charlton et al, 2010; Drew et al, 2010). ADULT DOSE: No clearly established guidelines exist; an optimal dosing regimen has not been established. Administer 1 to 2 grams diluted in 10 milliliters D5W IV/IO over 15 minutes (Neumar et al, 2010). Followed if needed by a second 2 gram bolus and an infusion of 0.5 to 1 gram (4 to 8 mEq) per hour in patients not responding to the initial bolus or with recurrence of dysrhythmias (American Heart Association, 2005; Perticone et al, 1997). Rate of infusion may be increased if dysrhythmias recur. For persistent refractory dysrhythmias, a continuous infusion of up to 3 to 10 milligrams/minute in adults may be given (Charlton et al, 2010).
    b) PEDIATRIC DOSE: 25 to 50 milligrams/kilogram diluted to 10 milligrams/milliliter for intravenous infusion over 5 to 15 minutes up to 2 g (Charlton et al, 2010).
    c) PRECAUTIONS: Use with caution in patients with renal insufficiency.
    d) MAJOR ADVERSE EFFECTS: High doses may cause hypotension, respiratory depression, and CNS toxicity (Neumar et al, 2010). Toxicity may be observed at magnesium levels of 3.5 to 4.0 mEq/L or greater (Charlton et al, 2010).
    e) MONITORING PARAMETERS: Monitor heart rate and rhythm, blood pressure, respiratory rate, motor strength, deep tendon reflexes, serum magnesium, phosphorus, and calcium concentrations (Prod Info magnesium sulfate heptahydrate IV, IM injection, solution, 2009).
    4) OVERDRIVE PACING
    a) Institute electrical overdrive pacing at a rate of 130 to 150 beats per minute, and decrease as tolerated. Rates of 100 to 120 beats per minute may terminate torsades (American Heart Association, 2005). Pacing can be used to suppress self-limited runs of TdP that may progress to unstable or refractory TdP, or for override refractory, persistent TdP before the potential development of ventricular fibrillation (Charlton et al, 2010). In a case series overdrive pacing was successful in terminating TdP associated with bradycardia and drug-induced QT prolongation (Neumar et al, 2010).
    5) POTASSIUM REPLETION
    a) Potassium supplementation, even if serum potassium is normal, has been recommended by many experts (Charlton et al, 2010; American Heart Association, 2005). Supplementation to supratherapeutic potassium concentrations of 4.5 to 5 mmol/L has been suggested, although there is little evidence to determine the optimal range in dysrhythmia (Drew et al, 2010; Charlton et al, 2010).
    6) ISOPROTERENOL
    a) Isoproterenol has been successful in aborting torsades de pointes that was resistant to magnesium therapy in a patient in whom transvenous overdrive pacing was not an option (Charlton et al, 2010) and has been successfully used to treat torsades de pointes associated with bradycardia and drug induced QT prolongation (Keren et al, 1981; Neumar et al, 2010). Isoproterenol may have a limited role in pharmacologic overdrive pacing in select patients with drug-induced torsades de pointes and acquired long QT syndrome (Charlton et al, 2010; Neumar et al, 2010). Isoproterenol should be avoided in patients with polymorphic VT associated with familial long QT syndrome (Neumar et al, 2010).
    b) DOSE: ADULT: 2 to 10 micrograms/minute via a continuous monitored intravenous infusion; titrate to heart rate and rhythm response (Neumar et al, 2010).
    c) PRECAUTIONS: Correct hypovolemia before using; contraindicated in patients with acute cardiac ischemia (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    1) Contraindicated in patients with preexisting dysrhythmias; tachycardia or heart block due to digitalis toxicity; ventricular dysrhythmias that require inotropic therapy; and angina. Use with caution in patients with coronary insufficiency (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    d) MAJOR ADVERSE EFFECTS: Tachycardia, cardiac dysrhythmias, palpitations, hypotension or hypertension, nervousness, headache, dizziness, and dyspnea (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    e) MONITORING PARAMETERS: Monitor heart rate and rhythm, blood pressure, respirations and central venous pressure to guide volume replacement (Prod Info Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, 2013).
    7) OTHER DRUGS
    a) Mexiletine, verapamil, propranolol, and labetalol have also been used to treat TdP, but results have been inconsistent (Khan & Gowda, 2004).
    8) AVOID
    a) Avoid class Ia antidysrhythmics (eg, quinidine, disopyramide, procainamide, aprindine), class Ic (eg, flecainide, encainide, propafenone) and most class III antidysrhythmics (eg, N-acetylprocainamide, sotalol) since they may further prolong the QT interval and have been associated with TdP.
    9) LAAM
    a) The manufacturer discontinued the sale and distribution of levomethadyl hydrochloride acetate (ORLAAM(R)) in the United States in September 2003. ORLAAM(R) was removed from the European market in March 2001. This is due to reports of severe cardiac adverse events including QT interval prolongation, torsades de pointes, and cardiac arrest.
    J) DRUG WITHDRAWAL
    1) SIGNS OF WITHDRAWAL INCLUDE lacrimation, restlessness, diaphoresis, mydriasis, piloerection, fasciculations, abdominal pain, tachycardia, hypertension, nausea, diarrhea, dehydration, hyperglycemia, and fetal posturing. Death rarely occurs (Freitas, 1985).
    2) Seizures and hallucinations are almost always associated with concomitant withdrawal from sedative/hypnotics except in neonates where seizures are noted.
    3) NALOXONE: Naloxone Should be used with caution in patients that may be opioid abusers. Its use may precipitate opioid withdrawal (Popper et al, 1989).
    4) Management of withdrawal in the confirmed addict may be accomplished with the administration of clonidine, or with the reintroduction of the original addicting agent if available through a recognized drug withdrawal program.
    a) A tapered course over 3 weeks will accomplish this goal.
    5) CLONIDINE: In well controlled settings with compliant (selected, not randomized) patients, clonidine 6 micrograms/kilogram acutely and 10 to 17 micrograms/kilogram/day chronically (for 10 days) has been effective in preventing opioid withdrawal syndrome (Riordan & Kleber, 1980; Gold et al, 1980; Gold et al, 1978a; Gold et al, 1978b; Gold et al, 1979).
    K) DRUG DEPENDENCE
    1) NALTREXONE: The usual dose to block opioid effects is 50 mg/day orally for several weeks, followed by a schedule of 3 doses per week: 100 mg on Monday, 100 mg on Wednesday, and 150 mg on Friday. Before initiating therapy the patient should be opioid-free for at least 1 week to avoid a withdrawal reaction (Landsberg et al, 1976; Willette, 1982; Prod Info ReVia(R), 1999).

Enhanced Elimination

    A) HEMODIALYSIS
    1) Because of the large volume of distribution, and protein binding, there is no role for enhanced elimination.

Case Reports

    A) ADULT
    1) ADULT: A 30-year-old known drug addict who had used methadone for several years was found unresponsive and pronounced dead on admission to the hospital (Jensen & Gregersen, 1991). Postmortem exam revealed no obvious "fresh" injection sites. The cause of death was concluded to be poisoning by methadone injection. Necrotic groin wounds were thought to be old injection sites; however, microscopic exam revealed the presence of fresh bleeding and tablet residue which were believed to act as fistulas allowing direct administration into large veins.
    B) PEDIATRIC
    1) INFANT: A 3-month-old girl was admitted to the ED with respiratory failure, unconsciousness, and generalized seizures; she was also hypotonic with miotic pupils. Mechanical ventilation was started. The infant had previously been treated at birth for narcotic withdrawal, which was stopped at 3 weeks of age. She was not breast-fed. Initial blood counts, serum biochemistries, and CSF examinations were normal. Chest radiograph, CT scan of the brain, and microbiological studies were also normal. Narcotic poisoning was suspected, and a naloxone infusion (0.05 mg/kg/hr) was started. The infant's condition improved and mechanical ventilation was discontinued. A urine toxicology screen revealed the presence of methadone, and physical abuse by methadone poisoning was suspected. The actual dose of methadone remained unknown. The infant improved and was discharged (Lee & Lam, 2002).
    2) CHILD ABUSE
    a) Child abuse resulting in death has been reported. The deaths of a 14-month-old and a 5-month-old were attributed to methadone-related asphyxia. In both cases, the mothers were the perpetrators (Kintz et al, 2005).

Summary

    A) TOXIC DOSE: There is little margin between a therapeutic methadone dose for an opioid tolerant person and a toxic dose with doses of 30 to 40 mg potentially lethal for a non-tolerant adult. Children are particularly vulnerable to overdose. Inadvertent ingestion of methadone was found to be the most toxic of the opioids; doses as low as a single tablet can lead to death (10 mg can be potentially lethal for a 10 kg toddler). Patients who receive early treatment that prevents hypoxia and respiratory depression generally do well.
    B) THERAPEUTIC DOSE: ADULTS: ORAL: Opioid naive patients: 2.5 mg orally every 8 hours initially, titrated slowly to effect. Opioid dependent patients: 10 to 30 mg orally titrated slowly to effect. PARENTERAL: 2.5 to 10 mg. PEDIATRIC: 0.1 to 0.2 mg/kg orally every 6 hours as needed; maximum of 10 mg.

Therapeutic Dose

    7.2.1) ADULT
    A) METHADONE
    1) MODERATE TO SEVERE PAIN
    a) GUIDELINE DOSING: For opioid-naive patients, the usual methadone initial dose is 2.5 mg every 8 hours (oral, IV, IM, SubQ), with dose increases occurring no more frequently than weekly. In opioid-tolerant patients, starting methadone doses should generally not exceed 30 to 40 mg a day, even in patients on high doses of other opioids. Extreme caution should be used to avoid overdose, taking into consideration methadone's long elimination half-life (Chou et al, 2009)
    b) MANUFACTURER DOSING: For opioid-naive patients, the usual methadone initial dose is 2.5 to 10 mg every 8 to 12 hours, slowly titrated to effect. More frequent administration may be necessary during methadone initiation to maintain adequate analgesia, and extreme caution should be used to avoid overdose, taking into consideration methadone's long elimination half-life. Methadone may be administered intramuscularly, but the absorption has not been well defined and may be unpredictable; local tissue reactions may occur (Prod Info methadone hcl injection, 2006).
    2) DRUG DETOXIFICATION
    a) 15 to 30 mg ORALLY as an initial dose, additional 5 to 10 mg can be given 2 to 4 hr later if needed; adjust dose cautiously over the first week based upon control of withdrawal 2 to 4 hr post dose; usual total daily dose is 40 mg/day; keep on stable dose for 2 to 3 days then decrease in 1 to 2 day intervals according to response; detoxification treatment usually occurs over 21 days (Prod Info DOLOPHINE(R) HYDROCHLORIDE oral tablets, 2006; Prod Info methadone hydrochloride oral solution, 2008; Prod Info methadone hcl tablets for oral suspension, 2005)
    3) OPIOID ABUSE, MAINTENANCE THERAPY
    a) Maintenance doses must be individualized; methadone doses should be titrated to a dose where symptoms are prevented for 24 hr; usual maintenance doses range from 80 to 120 mg/day (Prod Info DOLOPHINE(R) HYDROCHLORIDE oral tablets, 2006; Prod Info methadone hydrochloride oral solution, 2008; Prod Info methadone hcl tablets for oral suspension, 2005)
    B) LEVO ALPHA ACETYL METHADOL (LAAM)
    1) LAAM: The manufacturer discontinued the sale and distribution of levomethadyl hydrochloride acetate (ORLAAM(R)) in the United States in September 2003. ORLAAM(R) was removed from the European market in March 2001. This is due to reports of severe cardiac adverse events including QT interval prolongation, torsades de pointes, and cardiac arrest.
    7.2.2) PEDIATRIC
    A) ROUTE OF ADMINISTRATION
    1) METHADONE
    a) INTRAMUSCULAR, INTRAVENOUS or SUBCUTANEOUS
    1) PAIN MANAGEMENT
    a) Initial Dosing: 0.1 to 0.2 mg/kg per dose. Repeat as needed (usually every 4 to 12 hours). Maximum 5 mg/dose (Kraemer & Rose, 2009; Greco & Berde, 2005; Golianu et al, 2000; Berde et al, 1991; Yaster et al, 1989).
    b) Supplemental doses as small as 0.03 to 0.08 mg/kg IV have been used successfully for postoperative pain management in children (Berde et al, 1991).
    b) ORAL
    1) Initial dose: 0.1 to 0.2 mg/kg per dose. Repeat as needed (usually every 4 to 12 hours). Maximum 10 mg/dose (Kraemer & Rose, 2009; Greco & Berde, 2005; Golianu et al, 2000; Shir et al, 1998; Miser & Miser, 1985).
    2) Opioid experienced children with cancer-related pain may require every 4 to 8 hour dosing intervals (Davies et al, 2008; Miser & Miser, 1985).
    2) NEONATAL ABSTINENCE SYNDROME
    a) Initial dose: 0.05 to 0.2 mg/kg per dose orally every 12 to 24 hours (Burgos & Burke, 2009; Anand, 2007; Wunsch, 2006).
    b) Reduce dose by 10% to 20% per week over 4 to 6 weeks. Adjust weaning schedule based on signs and symptoms of withdrawal (Burgos & Burke, 2009).
    3) OPIOID WITHDRAWAL
    a) Initial dose: 0.05 to 0.1 mg/kg orally every 6 hours and adjust dose by 0.05 mg/kg if necessary to control symptoms. After 24 to 48 hours, extend interval to 8 to 12 hours, followed by a 10 to 14 day taper of the dose (Lugo et al, 2001; Anand & Arnold, 1994; Tobias et al, 1990).
    4) DETOXIFICATION TREATMENT
    a) The safety and efficacy of methadone in treatment of adolescents under the age of 18 with drug addictions has not been adequately studied. Treatment is based on individual consideration and by law requires parental consent (Prod Info Methadone hydrochloride (Methadone HCL(R), 1996).

Minimum Lethal Exposure

    A) PEDIATRIC
    1) 10 mg of methadone has been lethal in a young child (Harding-Pink, 1993).
    2) 20 to 30 mg, which is only 20 to 30 mL of liquid methadone preparations used by opioid-dependent adults in some countries, can be lethal to a small child (Molyneux et al, 1991).
    3) Approximately 40 mg of methadone was fatal in a 4-year-old boy; postmortem methadone blood level was 0.5 mg/L (Lorenzo & Weinstock, 1995). A fatality was reported in a 4-year-old boy who drank 40 mg of methadone and slept for several hours before he was discovered dead (Li et al, 2000).
    B) ADULT
    1) A standard day's maintenance dose of methadone (50 to 100 mg) can cause life-threatening poisoning in non-tolerant adults (Drummer et al, 1992; Harding-Pink, 1993).

Maximum Tolerated Exposure

    A) CASE REPORT: A 20-day-old premature neonate (born at a gestational age of 24.5 weeks) who was receiving fentanyl and morphine for analgesia and sedation after several tube thoracostomies for pneumothorax, developed respiratory depression, clonic movements, and hemodynamic instability after receiving 7.8 mg of methadone (about 200-fold the prescribed dose) instead of 39 mcg every 6 hours. He recovered following supportive care (George et al, 2012).
    B) PEDIATRIC: A 30-month-old girl presented unconscious (Glasgow Coma Scale 3/15) after ingesting 20 mg of methadone (methadone blood level: 0.063 mcg/mL). She recovered after treatment with naloxone infusion for 8 hours. Five days postingestion, she developed delayed methadone encephalopathy. She recovered gradually after supportive care (Zanin et al, 2010).
    C) PEDIATRIC: A neonate developed life-threatening rigid-chest syndrome after receiving a methadone dose of 10 times the ordered amount of 0.05 mg/kg per dose every 6 hours. He recovered completely after receiving naloxone therapy (Lynch & Hack, 2010).
    D) CASE REPORT: A 35-year-old man who was taking methadone 20 mg/day, sertraline, and venlafaxine, ingested 200 mg of methadone in a suicide attempt and developed symptoms of serotonin syndrome (panic, confusion, disorientation, hallucinations, elevated blood pressure, pulse, and respiration) 5 hours post-ingestion. On presentation, a narcotic urine screen was negative, therefore, no decontamination or antagonist therapy was administered. Ten hours post-ingestion, he was found unconscious after aspirating his stomach contents. Despite supportive therapy, he died 5 days post-ingestion (Martinez & Martinez, 2008).
    E) CASE REPORT: A 37-year-old man presented with nausea, hearing loss, confusion, and a low respiratory rate after ingesting 15 methadone 5 mg tablets. His symptoms improved after receiving a single dose of naloxone; however, he complained of profound hearing loss with a mild tinnitus, but no vestibular symptoms or aural fullness. His neurological examination was normal. Pure tone audiometry showed bilateral sudden sensorineural hearing loss of on average -57 db in the right ear and -40 db in the left ear. His condition continued to improve and a second audiometry was normal on day 10 (van Gaalen et al, 2009).
    F) CASE REPORT: A 76-year-old woman with breast cancer and chronic lymphocytic leukemia was inadvertently exposed to 320 mg of methadone taken over a 12-hour period. CNS and respiratory depression developed, and the patient's clinical course was complicated by persistent respiratory insufficiency and pulmonary edema. The patient gradually improved and was discharged from the ICU on day 9 (Hantson et al, 2003).
    G) CASE REPORT/EPIDURAL INJECTION: A 75-year-old man with a history of prostate cancer, received an inadvertent epidural methadone injection of 40 mg instead of 4 mg and developed mild drowsiness. Sixteen hours after exposure, euphoria developed, but the patient returned to baseline within 6 hours. He never developed respiratory depression or hypotension (Ortiz-Gomez & Souto-Ferro, 2006).

Serum Plasma Blood Concentrations

    7.5.1) THERAPEUTIC CONCENTRATIONS
    A) THERAPEUTIC CONCENTRATION LEVELS
    1) Therapeutic methadone drug concentrations when treating opiate withdrawal are generally in the range of 100 to 400 ng/mL (Dhopesh et al, 2002).
    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) ADULT
    a) A 19-year-old woman with a history of illicit drug use intentionally ingested an unknown amount of methadone and was found obtunded with agonal respirations and bloody oral secretions. Laboratory analysis upon arrival at the emergency department showed a serum methadone concentration of 48 ng/mL but a urine drug screen was negative for opiates. She recovered after 3 days of treatment with extracorporeal membrane oxygenation and was extubated and discharged on day 5 (Daugherty, 2011).
    b) A 49-year old woman developed respiratory failure after intentionally ingesting an amount of methadone 10 times her normal dose. Laboratory analysis at the time of admission showed a serum methadone concentration of 817 ng/mL. Urine and blood analysis for other substances including heroin, cocaine, and cannabis were negative. She recovered from the respiratory failure; however, a few weeks later she developed symptoms consistent with toxic spongiform leukoencephalopathy. After 3 months of supportive physical therapy and botox injections, brain MRI showed no improvement and frontal cognitive impairment persisted (Gheuens et al, 2010).
    c) COMBINATION EXPOSURE: A 23-year-old man was found unconscious and pulseless, and later developed multiorgan failure with acute necrosis of the bilateral globi pallidi in the brain, and systemic rhabdomyolysis after reportedly ingesting 40 mg of methadone and insufflating 3 mg of alprazolam. Because of his poor prognosis, life support measures were withdrawn about 24 hours after presentation, and he died soon after. Laboratory results revealed blood methadone concentration of 154 ng/mL (therapeutic range, 50 to 1000 ng/mL), alprazolam concentration of 14.1 ng/mL (therapeutic range, 10 to 40 ng/mL), and cannabinoid carboxytetrahydrocannabinol of 5.9 ng/mL (Corliss et al, 2013).
    2) PEDIATRIC
    a) CASE REPORT: A 20-day-old premature neonate (born at a gestational age of 24.5 weeks) who was receiving fentanyl and morphine for analgesia and sedation after several tube thoracostomies for pneumothorax, developed respiratory depression, clonic movements, and hemodynamic instability after receiving 7.8 mg of methadone (about 200-fold the prescribed dose). Peak serum methadone concentration was 340 ng/mL 20 hours after methadone overdose. Urine methadone concentration was greater than 5000 ng/mL for the first 73 hours and decreased to 389 ng/mL about 141 hours after overdose (George et al, 2012).
    b) A 6-year-old boy was pronounced dead hours after he was inadvertently administered methadone, rather than 50 mg meperidine, in a prescribed oral dental cocktail also containing 50 mg hydroxyzine and 375 mg chloral hydrate. Postmortem toxicological analyses of femoral blood showed methadone (0.51 mcg/mL), hydroxyzine (less than 0.54 mcg/mL), and trichloroethanol (8.3 mcg/mL). Further analyses of liver tissue showed 2.2 mcg/g of methadone and gastric contents showed less than 1 mcg/mL of methadone. Analyses of drug residual in the syringe used to administer the solution showed presence of hydroxyzine, chloral hydrate, and methadone. Meperidine, hydroxyzine, and chloral hydrate were detected in a control syringe compounded by the same pharmacy for a different patient (Kupiec et al, 2011).
    c) Postmortem blood methadone concentrations of 2 children (3.5 and 15 months of age) were 277 and 298 ng/mL, respectively. Both patients died after receiving unknown quantities of methadone (Mistry et al, 2010).
    d) In several case reports (children 5 weeks to 13 years), postmortem blood methadone concentrations of 60 to 1100 ng/mL were reported (Mistry et al, 2010).
    e) A 30-month-old girl presented unconscious after ingesting 20 mg of methadone (methadone blood level: 0.063 mcg/mL or 63 ng/mL). She recovered after treatment with naloxone infusion for 8 hours. Five days postingestion, she developed delayed methadone encephalopathy. She recovered gradually after supportive care (Zanin et al, 2010).
    3) An inadvertent dosing of methadone (125 mg) to an opiate-naive 52-year-old man resulted in the following serial methadone blood concentrations (Dhopesh et al, 2002):
    TIME AFTER DOSEBLOOD CONCENTRATION
    2 hours470 ng/mL
    8 hours256 ng/mL
    18 hours199 ng/mL

    4) POSTMORTEM BLOOD CONCENTRATION
    a) Postmortem methadone blood concentrations ranged from 0.4 to 1.8 milligrams/liter in 10 fatal cases attributed to methadone (Manning et al, 1976).
    b) In a series of 10 fatal cases, postmortem methadone blood concentrations ranged from 0.87 to 8.1 millimoles/L with a mean of 2.1 millimoles/L (Drummer et al, 1992). Additional depressant drugs were found in five cases.
    c) In a series of fatalities due to methadone only (15 cases) or methadone plus a benzodiazepine (30 cases), mean postmortem methadone blood concentrations were 0.34 and 0.74 mg/L, respectively, in the methadone only group and the methadone plus benzodiazepine group. Higher methadone concentrations in the cases with both methadone and benzodiazepines might be due to tolerance from chronic abuse or benzodiazepine induced inhibition of the enzymes that metabolize methadone (Mikolaenko et al, 2002).
    d) Postmortem methadone blood level of 1400 ng/mL was reported in a 27-year-old woman who had been treated with 180 mg/day of methadone for greater than one year (Perret et al, 2000).
    e) Postmortem methadone blood level of 1900 ng/mL was reported in a 40-year-old man with hepatic encephalopathy who had been taking an average methadone dose of 120 mg/day for 2 years (Perret et al, 2000).
    f) A 21-year-old man was reported to have postmortem blood methadone level of 800 ng/mL after being in a methadone treatment program for 3 weeks and abusing methadone by injecting his oral dose (Perret et al, 2000).
    g) Postmortem methadone blood level of 1400 ng/mL was reported in a 38-year-old HIV positive male, who had been on methadone 80 mg/day for 3 years. Autopsy revealed bronchopneumonia, pleuritic and pericardia effusion (Perret et al, 2000).
    h) One study, using a linear regression model, showed a linear relationship between weight-adjusted methadone dose and post-mortem blood methadone concentrations; however, there appeared to be a significant difference in post-mortem blood concentrations between men and women. Given the same dose per kilogram of methadone, women appeared to have significantly higher post-mortem blood methadone concentrations than men. Also, the estimated post-mortem blood concentrations of male and female maintenance patients were at least two and three times, respectively, the estimated trough levels of living patients. It is believed that post-mortem methadone redistribution may be the primary cause for the differences between the post-mortem blood concentrations between men and women, and for the differences between estimated blood concentrations of living and deceased patients (Caplehorn & Drummer, 2002).
    i) PEDIATRIC: Postmortem blood methadone concentration was 0.5 mg/L in a 4-year-old boy following an accidental ingestion of approximately 40 mg of methadone (Lorenzo & Weinstock, 1995).

Toxicity Information

    7.7.1) TOXICITY VALUES
    A) LD50- (INTRAPERITONEAL)MOUSE:
    1) 35 mg/kg (RTECS, 2001)
    B) LD50- (ORAL)MOUSE:
    1) 70 mg/kg (RTECS, 2001)
    C) LD50- (SUBCUTANEOUS)MOUSE:
    1) 35 mg/kg (RTECS, 2001)
    D) LD50- (INTRAPERITONEAL)RAT:
    1) 18 mg/kg (RTECS, 2001)
    E) LD50- (ORAL)RAT:
    1) 86 mg/kg (RTECS, 2001)
    F) LD50- (SUBCUTANEOUS)RAT:
    1) 30 mg/kg (RTECS, 2001)

Pharmacologic Mechanism

    A) Methadone is a synthetic opioid similar in structure to propoxyphene (Fraser, 1990). It is a mu receptor type (Haddad et al, 1998). Methadone's primary actions are similar to morphine in providing analgesia, however, the onset of action is longer following methadone dosing (Prod Info Methadone hydrochloride (Methadone HCL(R), 1996).
    B) LAAM: The manufacturer discontinued the sale and distribution of levomethadyl hydrochloride acetate (ORLAAM(R)) in the United States in September 2003. ORLAAM(R) was removed from the European market in March 2001. This is due to reports of severe cardiac adverse events including QT interval prolongation, Torsades de Pointes, and cardiac arrest.

Physical Characteristics

    A) METHADONE - white crystalline powder

Molecular Weight

    A) METHADONE HYDROCHLORIDE: 345.90

General Bibliography

    1) AAP: Emergency drug doses for infants and children and naloxone use in newborns: clarification. AAP: Pediatrics 1989; 83:803.
    2) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    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) Anand KJ & Arnold JH: Opioid tolerance and dependence in infants and children. Crit Care Med 1994; 22(2):334-342.
    5) Anand KJ: Pharmacological approaches to the management of pain in the neonatal intensive care unit. J Perinatol 2007; 27(Suppl 1):S4-S11.
    6) Anggard E, Gunne LM, & Homstrand J: Disposition of methadone in methadone maintenance. Clin Pharmacol Ther 1975; 17:258-266.
    7) Anon: American academy of pediatrics committee on drugs: transfer of drugs and other chemicals into human milk. Pediatrics 2001; 108(3):776-789.
    8) Anselmo M, Campos RA, doCarmo V, et al: Methadone intoxication in a child: toxic encephalopathy?. J Child Neurol 2006; 21(7):618-620.
    9) Arevalo R, Emralino F, Lezcano L et al: In utero methadone exposure diminishes growth potentials in full-term neonates (abstract). Presented at the Pediatric Academic Societies' Annual Meeting; May 3-6, 2003; Seattle, WA, USA, 2003.
    10) Artigas A, Bernard GR, Carlet J, et al: The American-European consensus conference on ARDS, part 2: ventilatory, pharmacologic, supportive therapy, study design strategies, and issues related to recovery and remodeling.. Am J Respir Crit Care Med 1998; 157:1332-1347.
    11) Ashwath ML, Ajjan M, & Culclasure T: Methadone-induced bradycardia. J Emerg Med 2005; 29:73-75.
    12) Bakstad B, Sarfi M, Welle-Strand GK, et al: Opioid Maintenance Treatment during Pregnancy: Occurrence and Severity of Neonatal Abstinence Syndrome. A National Prospective Study. Eur Addict Res 2009; 15(3):128-134.
    13) Barrett DH, Luk AJ, & Parrish RG: An investigation of medical examiner cases in which methadone was detected, Harris county, Texas, 1987-1992. J Forensic Sci 1996; 41:442-448.
    14) Baselt RC: Disposition of Toxic Drugs and Chemicals in Man, 5th ed, Chemical Toxicology Institute, Foster City, CA, 2000.
    15) Berde CB, Beyer JE, Bournaki MC, et al: Comparison of morphine and methadone for prevention of postoperative pain in 3- to 7-year-old children. J Pediatr 1991; 119(1 Pt 1):136-41.
    16) Bier JB , Finger AS , Bier BA , et al: Growth and developmental outcome of infants with in-utero exposure to methadone vs buprenorphine. J Perinatol 2015; 35(8):656-659.
    17) Binchy JM, Molyneux EM, & Manning J: Accidental ingestion of methadone by children in Merseyside. Br Med J 1994; 308:1335-1336.
    18) Blinick G, Inturrisi CE, Jerez E, et al: Methadone assays in pregnant women and progeny. Am J Obstet Gynecol 1975; 121:617.
    19) Brice JEH, Moreland TA, Parija AC, et al: Plasma naloxone levels in the newborn after intravenous and intramuscular administration. Br J Clin Pharmacol 1979; 8:412P-413P.
    20) Brooks DE, Roberge R, & Spear A: Clinical nuances of pediatric methadone intoxication. Vet Human Toxicol 1999; 41:388-390.
    21) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    22) Brower RG, Matthay AM, & Morris A: Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Eng J Med 2000; 342:1301-1308.
    23) Brown HL, Britton KA, & Mahaffey D: Methadone maintenance in pregnancy: A reappraisal. Am J Obstet Gynecol 1998; 179:459-463.
    24) Bunten H, Liang WJ, Pounder DJ, et al: OPRM1 and CYP2B6 gene variants as risk factors in methadone-related deaths. Clin Pharmacol Ther 2010; 88(3):383-389.
    25) Burgos AE & Burke BL: Neonatal Abstinence Syndrome. Neoreviews 2009; 10(5):e222-e229.
    26) Calman L, Finch E, & Powis B: Only half of patients store methadone in safe place (letter). Br Med J 1996; 313:1481.
    27) Caplehorn JRM & Drummer OH: Methadone dose and post-mortem blood concentration. Drug Alcohol Rev 2002; 21:329-333.
    28) Caplehorn JRM & Drummer OH: Mortality associated with New South Wales methadone programs in 1994: lives lost and saved. MJA 1999; 170:104-109.
    29) Cataletto M: Respiratory Distress Syndrome, Acute(ARDS). In: Domino FJ, ed. The 5-Minute Clinical Consult 2012, 20th ed. Lippincott Williams & Wilkins, Philadelphia, PA, 2012.
    30) 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.
    31) Charlton NP , Lawrence DT , Brady WJ , et al: Termination of drug-induced torsades de pointes with overdrive pacing. Am J Emerg Med 2010; 28(1):95-102.
    32) 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.
    33) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    34) Chou R , Fanciullo GJ , Fine PG , et al: Clinical guidelines for the use of chronic opioid therapy in chronic noncancer pain. J Pain 2009; 10(2):113-130.
    35) Christenson BJ & Marjala AR: Two cases of sudden sensorineural hearing loss after methadone overdose. Ann Pharmacother 2010; 44(1):207-210.
    36) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    37) Copeman AJ & Robins AW: A fatal case of methadone toxicity. Is obesity implicated?. Arch Dis Child 2000; 82 (Suppl 1):A9.
    38) Corliss RF, Mandal R, & Soriano BJ: Bilateral acute necrosis of the globi pallidi and rhabdomyolysis due to combined methadone and benzodiazepine toxicity. Am J Forensic Med Pathol 2013; 34(1):1-4.
    39) Crane T, Badminton MN, & Dawson CM: Mefenamic acid prevents assessment of drug abuse with EMIT(R) assays (letter). Clin Chemistry 1993; 39:549.
    40) Cunningham FG, MacDonald PC, Leveno KF, et alCunningham FG, MacDonald PC, Leveno KF, et al (Eds): Williams Obstetrics, 19th. Appleton & Lange, Norwalk, CT, 1993.
    41) Daniels AM: Doctors in prison must be careful when prescribing methadone. Br Med J 1997; 315:603.
    42) Darke S, Sims J, & McDonald S: Cognitive impairment among methadone maintenance patients. Addiction 2000; 95:687-695.
    43) Dashe JS, Sheffield JS, Olscher DA, et al: Relationship between maternal methadone dosage and neonatal withdrawal. Obstet Gynecol 2002; 100(6):1244-1249.
    44) Daugherty LE: Extracorporeal membrane oxygenation as rescue therapy for methadone-induced pulmonary edema. Pediatr Emerg Care 2011; 27(7):633-634.
    45) Davies D , DeVlaming D , & Haines C : Methadone analgesia for children with advanced cancer. Pediatr Blood Cancer 2008; 51(3):393-397.
    46) DeBoer LG, Engebretsen KM, & Harris CR: Paroxysmal sympathetic storm (diencephalic seizures) following methadone overdose (abstract). J Toxicol-Clin Toxicol 2001; 39:314.
    47) Dhopesh V, Yu E, & Fudala PJ: Conservative management with naltrexone of an iatrogenic methadone overdose in an opiate-naive patient (letter). J Clin Psychopharmacol 2002; 22:231-232.
    48) Drew BJ, Ackerman MJ, Funk M, et al: Prevention of torsade de pointes in hospital settings: a scientific statement from the American Heart Association and the American College of Cardiology Foundation. J Am Coll Cardiol 2010; 55(9):934-947.
    49) Drummer OH, Opeskin K, & Syrjanen M: Methadone toxicity causing death in ten subjects starting on a methadone maintenance program. Am J Forensic Med & Pathol 1992; 13:346-350.
    50) Eap CB: Comment: opiate withdrawal symptoms induced by antiretroviral drugs (letter). Ann Pharmacother 2000; 34:1077-1078.
    51) Ehret GB, Voide C, Gex-Fabry M, et al: Drug-induced long QT syndrome in injection drug users receiving methadone: high frequency in hospitalized patients and risk factors. Arch Intern Med 2006; 166(12):1280-1287.
    52) Eissenberg T, Bigelow GE, & Strain EC: Dose-related efficacy of levomethadyl acetate for treatment of opioid dependence - a randomized trial. JAMA 1997; 277:1945-1951.
    53) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    54) Evans LE, Swainson CP, & Roscoe P: Treatment of drug overdosage with naloxone, a specific narcotic antagonist. Lancet 1973; 1:452-455.
    55) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    56) Fischer CG & Cook DR: The respiratory and narcotic antagonistic effects of naloxone in infants. Anesth Analg 1974; 53:849-852.
    57) Fraser AD: Clinical toxicology of drugs used in the treatment of opiate dependency. Clin in Lab Med 1990; 10:375-387.
    58) Fredheim OM, Borchgrevink PC, Hegrenaes L, et al: Opioid switching from morphine to methadone causes a minor but not clinically significant increase in QTc time: A prospective 9-month follow-up study. J Pain Symptom Manage 2006; 32(2):180-185.
    59) Freitas PM: Narcotic withdrawal in the emergency department. Am J Emerg Med 1985; 3:456-460.
    60) Gal TJ: Naloxone reversal of buprenorphine-induced respiratory depression. Clin Pharmacol Ther 1989; 45:66-71.
    61) Gayle MO, Ryan CA, & Nazarali S: Unusual cause of methadone poisoning. Acta Paediatr Scand 1991; 80:486-487.
    62) Geletko SM & Erickson AD: Decreased methadone effect after ritonavir initiation. Pharmacother 2000; 20:93-94.
    63) Geller RJ & Garrettson LK: Delayed onset of toxicity after methadone ingestion due to therapeutic error (abstract). Vet Hum Toxicol 1994; 36:367.
    64) George M, Kitzmiller JP, Ewald MB, et al: Methadone toxicity and possible induction and enhanced elimination in a premature neonate. J Med Toxicol 2012; 8(4):432-435.
    65) Gharib B, Farshadmoghadam H, Hosseini F, et al: Be careful of lies: a six-year-old boy with respiratory distress and decreased level of consciousness. Acta Med Iran 2014; 52(6):481-483.
    66) Gheuens S, Michotte A, Flamez A, et al: Delayed akinetic catatonic mutism following methadone overdose. Neurotoxicology 2010; 31(6):762-764.
    67) Gibbs J, Newson T, & Williams J: Naloxone hazard in infant of opioid abuser (letter). Lancet 1989; 2:159-160.
    68) Glatstein M, Finkelstein Y, & Scolnik D: Accidental methadone ingestion in an infant: case report and review of the literature. Pediatr Emerg Care 2009; 25(2):109-111.
    69) Gold MS, Pottash AC, & Sweeney DR: Opiate withdrawal using clonidine. JAMA 1980; 243:343-346.
    70) Gold MS, Redmond DE Jr, & Kleber HD: Clonidine blocks acute opiate withdrawal symptoms. Lancet 1978b; 2:599.
    71) Gold MS, Redmond DE Jr, & Kleber HD: Clonidine in opiate withdrawal. Lancet 1978a; 1:929-930.
    72) Gold MS, Redmond DE Jr, & Kleber HD: Noradrenergic hyperactivity in opiate withdrawal supported by clonidine reversal of opiate withdrawal. Am J Psychiatry 1979; 136:100.
    73) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    74) Golianu B, Krane EJ, Galloway KS, et al: Pediatric acute pain management. Pediatr Clin North Am 2000; 47(3):559-587.
    75) Gonzva J, Prunet B, Deniel C, et al: Early antidote use associated with noninvasive ventilation in prehospital treatment of methadone intoxication. Am J Emerg Med 2013; 31(2):448.
    76) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    77) Gramenz P, Roberts D, & Schrag L: Intra-arterial self-injection of methadone tablets into the femoral artery. J Emerg Med 2009; Epub:--.
    78) Greco C & Berde C: Pain management for the hospitalized pediatric patient. Pediatr Clin North Am 2005; 52(4):995-1027.
    79) Green H, James RA, & Gilbert JD: Methadone maintenance programs - a two-edged sword?. Am J Foren Med Pathol 2000; 21:359-361.
    80) Haas CF: Mechanical ventilation with lung protective strategies: what works?. Crit Care Clin 2011; 27(3):469-486.
    81) Haddad LM, Shannon MW, & Winchester JF: Clinical Management of Poisoning and Drug Overdose (3rd ed), W.B.Saunders Company, Philadelphia, PA, 1998.
    82) Hantson P, Vanbinst R, & Wallemacq P: Accidental methadone overdose in an opiate-naive elderly patient (letter). Intens Care Med 2003; 29:2105.
    83) Harding-Pink D: Opioid toxicity - Methadone: one person's maintenance dose is another's poison (letter). Lancet 1993; 341:665-666.
    84) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    85) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    86) Henderson GL & Wilson BK: Excretion of methadone and metabolites in human sweat. Res Commun Chem Pathol Pharmacol 1973; 5:1.
    87) Hendra TJ, Gerrish SP, & Forrest ARW: Fatal methadone overdose. Br Med J 1996; 313:481-482.
    88) Hillman AD, Witenko CJ, Sultan SM, et al: Serotonin syndrome caused by fentanyl and methadone in a burn injury. Pharmacotherapy 2015; 35(1):112-117.
    89) Hoffman JR, Schriger DL, & Luo JS: The empiric use of naloxone in patients with altered mental status: A reappraisal. Ann Emerg Med 1991; 20:246-252.
    90) Hoffman RJ, Nelson L, & Hoffman RS: Life-threatening levo-alpha-acetylmethadol (LAAM) overdose (abstract), EAPCCT XX International Congress, Amsterdam, The Netherlands, 2000.
    91) Howland MA & Nelson LS: Opioid Antagonists. In: Nelson LS, Lewin NA, Howland MA, et al, eds. Goldfrank’s Toxicologic Emergencies, McGraw Hill, New York, NY, 2011, pp 579-585.
    92) Hussain T & Ewer AK: Maternal methadone may cause arrhythmias in neonates. Acta Paediatr 2007; 96(5):768-769.
    93) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    94) Inturrisi CE & Verebely K: Disposition of methadone in man after a single oral dose. Clin Pharmacol Ther 1972b; 13:923.
    95) Inturrisi CE & Verebely K: The levels of methadone in the plasma in methadone maintenance. Clin Pharmacol Ther 1972a; 13:633-637.
    96) Inturrisi CE, Colburn WA, & Kaiko RF: Pharmacokinetics and pharmacodynamics of methadone in patients with chronic pain. Clin Pharmacol Ther 1987; 41:392-401.
    97) Irey NS & Froede RC: Evaluation of deaths from drug overdose: a clinicopathologic study. Am J Clin Pathol 1974; 61:778-784.
    98) Jansson LM, Choo RE, Harrow C, et al: Concentrations of methadone in breast milk and plasma in the immediate perinatal period. J Hum Lact 2007; 23(2):184-190.
    99) Jasinski DR, Pevnick JS, & Griffith ID: Human pharmacology and abuse potential of analgesic buprenorphine. Arch Gen Psychiatry 1978; 35:501-516.
    100) Jensen S & Gregersen M: Fatal poisoning with intravenously injected methadone and no fresh injection marks found. Int J Leg Med 1991; 104:299-301.
    101) Jones HE, Kaltenbach K, Heil SH, et al: Neonatal abstinence syndrome after methadone or buprenorphine exposure. N Engl J Med 2010; 363(24):2320-2331.
    102) Jones TF, Feler CA, Simmons BP, et al: Neurologic complications including paralysis after a medication error involving implanted intrathecal catheters. Am J Med 2002; 112(1):31-36.
    103) Justo D.: Methadone-induced long QT syndrome vs methadone-induced torsades de pointes. Arch Intern Med 2006; 166(20):2288-2290.
    104) Kaltenbach K & Finnegan LP: Perinatal and developmental outcome of infants exposed to methadone in-utero. Neurotoxicol Tetratol 1987; 9:311-313.
    105) Karch SB & Stephens BG: Toxicology and pathology of deaths related to methadone: retrospective review. West J Med 2000; 172:11-14.
    106) Kelly AM & Koutsogiannis Z: Intranasal naloxone for life threatening opioid toxicity. Emerg Med J 2002; 19:375.
    107) Kelly LE, Rieder MJ, Bridgman-Acker K, et al: Are infants exposed to methadone in utero at an increased risk for mortality?. J Popul Ther Clin Pharmacol 2012; 19(2):e160-e165.
    108) Keren A, Tzivoni D, & Gavish D: Etiology, warning signs and therapy of torsade de pointes: a study of 10 patients. Circulation 1981; 64:1167-1174.
    109) Kerr D , Kelly AM , Dietze P , et al: Randomized controlled trial comparing the effectiveness and safety of intranasal and intramuscular naloxone for the treatment of suspected heroin overdose. Addiction 2009; 104(12):2067-2074.
    110) Khan IA & Gowda RM: Novel therapeutics for treatment of long-QT syndrome and torsade de pointes. Int J Cardiol 2004; 95(1):1-6.
    111) Kintz P, Villain M, Dumestre-Toulet Ve, et al: Methadone as a chemical weapon: two fatal cases involving babies. Ther Drug Monit 2005; 27(6):741-743.
    112) 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.
    113) Kollef MH & Schuster DP: The acute respiratory distress syndrome. N Engl J Med 1995; 332:27-37.
    114) Kraemer FW & Rose JB: Pharmacologic management of acute pediatric pain. Anesthesiol Clin 2009; 27(2):241-268.
    115) Krantz MJ, Martin J, Stimmel B, et al: QTc interval screening in methadone treatment. Ann Intern Med 2009; 150(6):387-95.
    116) Krauss B & Green SM: Procedural sedation and analgesia in children. Lancet 2006; 367(9512):766-780.
    117) Kupiec TC, Kemp P, Raj V, et al: A fatality due to an accidental methadone substitution in a dental cocktail. J Anal Toxicol 2011; 35(7):512-515.
    118) Lalkin A, Kapur B, & Koren G: Contamination of antibiotics resulting in severe pediatric methadone poisoning. Annal Pharmacother 1999; 33:314-316.
    119) Lamont P & Hunt SC: A twist on torsade: a prolonged QT interval on methadone. J Gen Intern Med 2006; 21(11):9-12.
    120) Landsberg R, Taintor Z, Plumb M et al: "An analysis of naltrexone use -- its efficacy, safety and potential," In: Julius D & Renault P (Ed): Narcotic Antagonists: Naltrexone Progress Report, Research Monograph 9, Rockville, MD. National Institute on Drug Abuse, 1976, 1976.
    121) Lavonas EJ, Drennan IR, Gabrielli A, et al: Part 10: Special Circumstances of Resuscitation: 2015 American Heart Association Guidelines Update for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. Circulation 2015; 132(18 Suppl 2):S501-S518.
    122) Lee AC & Lam SY: Nonaccidental methadone poisoning (letter). Clin Pediatr 2002; 41:365-366.
    123) Li L, Levine B, & Smialek JE: Fatal methadone poisoning in children: Maryland 1992-1996. Substance Use Misuse 2000; 35:1141-1148.
    124) 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.
    125) LoVecchio F, Pizon A, Riley B, et al: Onset of symptoms after methadone overdose. Am J Emerg Med 2007; 25(1):57-59.
    126) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    127) Lorenzo JA & Weinstock DJ: Pertinent medical intelligence: methadone ingestion in children. Maryland Med J 1995; 44:44-46.
    128) Lugo RA, MacLaren R, Cash J, et al: Enteral methadone to expedite fentanyl discontinuation and prevent opioid abstinence syndrome in the PICU. Pharmacotherapy 2001; 21(12):1566-1573.
    129) Lynch RE & Hack RA: Methadone-induced rigid-chest syndrome after substantial overdose. Pediatrics 2010; 126(1):e232-e234.
    130) Maas U, Kattner E, Weingart-Jesse B, et al: Infrequent neonatal opiate withdrawal following maternal methadone detoxification during pregnancy.. J Perinatol Med 1990; 18:111-118.
    131) Maio RF, Gaukel B, & Freeman B: Intralingual naloxone injection for narcotic-induced respiratory depression. Ann Emerg Med 1987; 16:572-573.
    132) Manning T, Bidanset JH, & Cohen S: Evaluation of the Abuscreen for methadone. J Forensic Sci 1976; 21:112-120.
    133) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    134) Martinez TT & Martinez DN : A case of serotonin syndrome associated with methadone overdose. Proc West Pharmacol Soc 2008; 51:42-44.
    135) Mathew P & Storey P: Subcutaneous methadone in terminally ill patients: manageable local toxicity. J Pain Symptom Manage 1999; 18:49-52.
    136) McCann B, Hunter R, & McCann J: Cocaine/heroin induced rhabdomyolysis and ventricular fibrillation. Emerg Med J 2002; 19:264-265.
    137) McCarthy JH: More people die from methadone misuse than from heroin misuse. Br Med J 1997; 315:603.
    138) McGlone L & Mactier H: Infants of opioid-dependent mothers: neurodevelopment at six months. Early Hum Dev 2015; 91(1):19-21.
    139) McGlone L, Hamilton R, McCulloch DL, et al: Visual outcome in infants born to drug-misusing mothers prescribed methadone in pregnancy. Br J Ophthalmol 2014; 98(2):238-245.
    140) Metkees M, Meesa IR, & Srinivasan A: Methadone-induced acute toxic leukoencephalopathy. Pediatr Neurol 2015; 52(2):256-257.
    141) Mikkelsen SL & Ash KO: Adulterants causing false negatives in illicit drug testing. Clin Chem 1988; 34:2333-2336.
    142) Mikolaenko I, Robinson CA, & Davis GG: A review of methadone deaths in Jefferson County, Alabama. Am J Foren Med Pathol 2002; 23:299-304.
    143) Mills F, MacLennan SC, Devile CJ, et al: Severe cerebellitis following methadone poisoning. Pediatr Radiol 2008; 38(2):227-229.
    144) Milone MC: Laboratory testing for prescription opioids. J Med Toxicol 2012; 8(4):408-416.
    145) Miser AW & Miser JS : The Use of Oral Methadone to Control Moderate and Severe Pain in Children and Young Adults with Malignancy. Clin J Pain 1985; 1(4):243-248.
    146) Mistry V, Jeffery AJ, Madira W, et al: Methadone toxicity in infants: a report of two fatalities. Forensic Sci Med Pathol 2010; 6(2):116-120.
    147) Mofenson HC & Caraccio TR: Continuous infusion of intravenous naloxone (letter). Ann Emerg Med 1987; 16:374-375.
    148) Molyneux E, Ahern R, & Baldwin B: Accidental ingestion of methadone. Br Med J 1991; 303:922-923.
    149) Mycyk MB, Szyszko AL, & Aks SE: Nebulized naloxone gently and effectively reverses methadone intoxication. J Emerg Med 2003; 24(2):185-187.
    150) NHLBI ARDS Network: Mechanical ventilation protocol summary. Massachusetts General Hospital. Boston, MA. 2008. Available from URL: http://www.ardsnet.org/system/files/6mlcardsmall_2008update_final_JULY2008.pdf. As accessed 2013-08-07.
    151) Neeleman J & Farrell M: Fatal methadone and heroin overdoses: time trends in England and Wales. J Epidemol & Comm Health 1997; 51:435-437.
    152) Nelson LB, Ehrlich S, & Calhoun JH: Occurrence of strabismus in infants born to drug-dependent women. Am J Dis Child 1987; 141:175-178.
    153) 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.
    154) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    155) Nordt SP, Zilberstein J, & Gold B: Methadone-induced torsade de pointes. Am J Emerg Med 2011; 29(4):476e1-e2.
    156) Olsen G: Methadone binding to human plasma proteins. Clin Pharmacol Ther 1973; 14:338.
    157) Ortiz-Gomez JR & Souto-Ferro JM: Epidural methadone overdose. Rev Esp Anestesiol Reanim 2006; 53(6):389-390.
    158) P Robinson : Viramune (nevirapine)/methadone interaction. Dear Doctor. Boehringer Ingelheim Roxane Laboratories. Ridgefield, CT. 1999. Available from URL: http://www.fda.gov/medwatch/safety/1999/viramu.pdf.
    159) 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.
    160) Peles E, Bodner G, Kreek MJ, et al: Corrected-QT intervals as related to methadone dose and serum level in methadone maintenance treatment (MMT) patients: a cross-sectional study. Addiction 2007; 102(2):289-300.
    161) Perret G, Deglon JJ, & Kreek MJ: Lethal methadone intoxications in Geneva, Switzerland, from 1994 to 1998. Addiction 2000; 95:1647-1653.
    162) Perticone F, Ceravolo R, & Cuccurullo O: Prolonged magnesium sulfate infusion in the treatment of ventricular tachycardia in acquired long QT syndrome. Clin Drug Inverst 1997; 13:229-236.
    163) Pierson PS, Howard P, & Kleber HD: Sudden deaths in infants born to methadone-maintained addicts. JAMA 1972; 220:1733-1734.
    164) Pimentel L & Mayo D: Chronic methadone therapy complicated by torsades de pointes: a case report. J Emerg Med 2008; 34(3):287-290.
    165) Pinzani V, Faucherre V, & Peyriere H: Methadone withdrawal symptoms with nevirapine and efavirenz (letter). Ann Pharmacother 2000; 34:405-407.
    166) Pohland A, Boaz HE, & Sullivan HR: Synthesis and identification of metabolites resulting from the biotransformation of DL-methadone in man and in the rat. J Med Chem 1971; 14:194.
    167) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    168) Popper C, Kelen GD, & Cunningham G: Naloxone hazard in drug abuser (letter). Lancet 1989; 2:446.
    169) Product Information: DISKETS(R) dispersible tablets for oral suspension, methadone hydrochloride tablets for oral suspension. Boehringer Ingelheim Roxane,Inc., Columbus, OH, 2007.
    170) Product Information: DOLOPHINE(R) HYDROCHLORIDE oral tablets, methadone hcl oral tablets. Roxane Laboratories,Inc, Columbus, OH, 2006.
    171) Product Information: DOLOPHINE(R) oral tablets, methadone HCl oral tablets. Roxane Laboratories, Inc. (per FDA), Columbus, OH, 2014.
    172) Product Information: EVZIO(TM) injection solution, naloxone HCl injection solution. Kaleo, Inc. (per FDA), Richmond, VA, 2014.
    173) Product Information: Isuprel(TM) intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection, isoproterenol HCl intravenous injection, intramuscular injection, subcutaneous injection, intracardiac injection. Hospira, Inc. (per FDA), Lake Forest, IL, 2013.
    174) Product Information: METHADOSE(TM) oral concentrate, sugar-free oral concentrate, methadone hydrochloride oral concentrate. Mallinckrodt,Inc., Hazelwood, MO, 2007.
    175) Product Information: Methadone hydrochloride (Methadone HCL(R). Roxane Laboratories, Inc, Columbus, OH, 1996.
    176) Product Information: NARCAN(R) nasal spray, naloxone HCl nasal spray. Adapt Pharma (per FDA), Radnor, PA, 2015.
    177) Product Information: Orlaam(R), levomethadyl acetate HC. Roxane Laboratories, Columbus, OH, 2000.
    178) Product Information: ReVia(R), Dupont Pharmaceuticals, Wilmington, Delaware (PI revised 5/1999) reviewed 10/2000. ReVia(R), Dupont Pharmaceuticals, Wilmington, Delaware, 1999.
    179) Product Information: Revex(R), nalmefene. Ohmeda Pharmaceuticals Products, Liberty Corner, NJ, 1998.
    180) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    181) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    182) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    183) Product Information: magnesium sulfate heptahydrate IV, IM injection, solution, magnesium sulfate heptahydrate IV, IM injection, solution. Hospira, Inc. (per DailyMed), Lake Forest, IL, 2009.
    184) Product Information: methadone hcl injection, methadone hcl injection. Xanodyne Pharmaceuticals,Inc, Newport, KY, 2006.
    185) Product Information: methadone hcl tablets for oral suspension, methadone hcl tablets for oral suspension. Mallinckrodt Inc, St. Louis, MO, 2005.
    186) Product Information: methadone hydrochloride oral solution, methadone hydrochloride oral solution. Roxane Laboratories Inc, Columbus, OH, 2008.
    187) Product Information: naloxone HCl IV, IM, subcutaneous injection solution, naloxone HCl IV, IM, subcutaneous injection solution. Hospira, Inc (per DailyMed), Lake Forest, IL, 2008.
    188) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    189) RTECS: Registry of Toxic Effects of Chemical Substances. National Institute for Occupational Safety and Health. Cincinnati, OH (Internet Version). Edition expires 2001; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    190) Rahbar F: Observations on methadone withdrawal in 16 neonates. Clin Pediatr 1975; 14:369-371.
    191) Raina S, Paydak H, Al-Lahham T, et al: Methadone induced Torsades de Pointes mimiking seizures in clinical presentation. Am J Drug Alcohol Abuse 2014; 40(6):490-492.
    192) Rajegowda BK, Glass L, Evans HE, et al: Methadone withdrawal in newborn infants. J Pediatr 1972; 81:532-534.
    193) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    194) Redfern N: Dihydrocodeine overdose treated with naloxone infusion. Br Med J 1983; 287:751-752.
    195) Ridgway ZA & Pountney AJ: Acute respiratory distress syndrome induced by oral methadone managed with non-invasive ventilation. Emerg Med J 2007; 24(9):681-.
    196) Riordan CE & Kleber HB: Rapid opiate detoxification with clonidine and naloxone. Lancet 1980; 1:1079.
    197) Rogers SC, Pruitt CW, Crouch DJ, et al: Rapid urine drug screens: diphenhydramine and methadone cross-reactivity. Pediatr Emerg Care 2010; 26(9):665-666.
    198) Rogers WO, Hall MA, & Brissie RM: Case Report: Detection of alprazolam in three cases of methadone/benzodiazepine overdose. J Forensic Sci 1997; 42:155-156.
    199) Romac DR: Safety of prolonged, high-dose infusion of naloxone hydrochloride for severe methadone overdose. Clin Pharm 1986; 5:251-254.
    200) Roy AK , McCarthy C , Kiernan G , et al: Increased incidence of QT interval prolongation in a population receiving lower doses of methadone maintenance therapy. Addiction 2012; 107(6):1132-1139.
    201) Saiful FB, Lafferty J, Jun CH, et al: Takotsubo cardiomyopathy due to iatrogenic methadone withdrawal. Rev Cardiovasc Med 2011; 12(3):164-167.
    202) Scheidegger C, Pietrzak J, & Frei R: Disseminated Candidiasis after intravenous use of oral methadone. Ann Intern Med 1991; 115:576.
    203) Schwab J & Caggiano AO: Pediatric methadone poisoning revisited. Clin Pediatr 2001; 40:119-120.
    204) 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.
    205) Sey MJ, Rubinstein D, & Smith DS: Accidental methadone intoxication in a child. Pediatrics 1971; 48:294-296.
    206) Shaw KA, Babu KM, & Hack JB: Methadone, another cause of opioid-associated hearing loss: a case report. J Emerg Med 2011; 41(6):635-639.
    207) Shir Y , Shenkman Z , Shavelson V , et al: Oral methadone for the treatment of severe pain in hospitalized children: a report of five cases. Clin J Pain 1998; 14(4):350-353.
    208) Siew LT, Auerbach M, Baum CR, et al: Respiratory failure caused by a suspicious white powder: a case report of intentional methadone poisoning in an infant. Pediatr Emerg Care 2012; 28(9):918-920.
    209) Smialek J, MonForte JR, Aronow R, et al: Methadone death in children. JAMA 1977; 238:2516.
    210) Smith WM & Gallagher JJ: "Les torsades de pointes": an unusual ventricular arrhythmia. Ann Intern Med 1980; 93:578-584.
    211) 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 2009; Epub:Epub.
    212) Stolbach A & Hoffman RS: Respiratory Principles. In: Nelson LS, Hoffman RS, Lewin NA, et al, eds. Goldfrank's Toxicologic Emergencies, 9th ed. McGraw Hill Medical, New York, NY, 2011.
    213) Sunjic S & Zador D: Methadone-related deaths in New South Wales (letter). MJA 1997; 166:54-55.
    214) Tandberg D & Abercrombie D: Treatment of heroin overdose with endotracheal naloxone. Ann Emerg Med 1982; 11:443-445.
    215) Tenenbein M: Continuous naloxone infusion for opiate poisoning in infancy. J Pediatr 1984; 105:645-648.
    216) Tennant FS, Rawson RA, & Pumphrey E: Clinical experiences with 959 opioid-dependent patients treated with levo-alpha-acetylmethadol (LAAM). J Substa Abuse Treat 1986; 3:195-202.
    217) Thanavaro KL & Thanavaro JL: Methadone-induced torsades de pointes: a twist of fate. Heart Lung 2011; 40(5):448-453.
    218) Tinelli F, Gamucci A, Battini R, et al: Congenital nystagmus in two infants born from mothers exposed to methadone during pregnancy. Ital J Pediatr 2013; 39:40.
    219) Tiras S, Haas V, Chevret L, et al: Nonketotic hyperglycemic coma in toddlers after unintentional methadone ingestion. Ann Emerg Med 2006; 48(4):448-451.
    220) Tobias JD, Schleien CL, & Haun SE: Methadone as treatment for iatrogenic narcotic dependency in pediatric intensive care unit patients. Crit Care Med 1990; 18(11):1292-1293.
    221) Umans JG & Szeto HH: Precipitated opiate abstinence in utero. Am J Obstet Gynecol 1985; 151:441-444.
    222) Vanden Hoek,TL; Morrison LJ; Shuster M; et al: Part 12: Cardiac Arrest in Special Situations 2010 American Heart Association Guidelines for Cardiopulmonary Resuscitation and Emergency Cardiovascular Care. American Heart Association. Dallas, TX. 2010. Available from URL: http://circ.ahajournals.org/cgi/reprint/122/18_suppl_3/S829. As accessed 2010-10-21.
    223) VandenHoek TL , Morrison LJ , Shuster M , et al: 2010 American Heart Association guidelines for cardiopulmonary resuscitation and emergency cardiovascular care science. Part 12: cardiac arrest in special situations. Circulation 2010; 122(18 Suppl 3):S829-S861.
    224) Verebely K, Volavka J, Mule S, et al: Methadone in man: pharmacokinetic and excretion studies in acute and chronic treatment. Clin Pharmacol Ther 1975; 18:180-190.
    225) Wanger K, Brough L, & Macmillan I: Intravenous vs subcutaneous naloxone for out-of-hospital management of presumed opioid overdose. Acad Emerg Med 1998; 5:293-299.
    226) Ward J, Mattick RP, & Hall W: The effectiveness and safety of methadone maintenance (letter). Addiction 1996; 91:1727-1729.
    227) Watson WA, Steele MT, & Muelleman RL: Opioid toxicity recurrence after an initial response to naloxone. Clin Toxicol 1998; 36:11-17.
    228) Weber JM, Tataris KL, Hoffman JD, et al: Can nebulized naloxone be used safely and effectively by emergency medical services for suspected opioid overdose?. Prehosp Emerg Care 2012; 16(2):289-292.
    229) Welles B, Belfrage P, & de Chateau P: Effects of naloxone on newborn infant behavior after maternal analgesia with pethidine during labor. Acta Obstet Gynecol Scand 1984; 63:617-619.
    230) Wiener PC, Hogg MIJ, & Rosen M: Effects of naloxone on pethidine-induced neonatal depression. Part I. Br Med J 1977; 2:228-229.
    231) Wilen SB, Ulreich S, & Rabinowitz JG: Roentgenographic manifestations of methadone-induced pulmonary edema. Radiology 1975; 114:51-55.
    232) Willette RE: Narcotic antagonists: an alternative for treating opioid dependence. Am Pharm 1982; NS22:44-46.
    233) Williamson PA, Foreman KJ, & White JM: Methadone-related overdose deaths in South Australia, 1984-1994. How safe is methadone prescribing?. MJA 1997; 166:302-305.
    234) Willson DF, Truwit JD, Conaway MR, et al: The adult calfactant in acute respiratory distress syndrome (CARDS) trial. Chest 2015; 148(2):356-364.
    235) Wilson DF, Thomas NJ, Markovitz BP, et al: Effect of exogenous surfactant (calfactant) in pediatric acute lung injury. A randomized controlled trial. JAMA 2005; 293:470-476.
    236) Wojnar-Horton RE, Kristensen JH, Yapp P, et al: Methadone distribution and excretion into breast milk of clients in a methadone maintenance programme. Br J Clin Pharmacol 1997; 44:543-547.
    237) Wolff K: Characterization of methadone overdose: clinical considerations and the scientific evidence. Ther Drug Monitor 2002; 24:457-470.
    238) Worm K, Steentoft A, & Kringsholm B: Methadone and drug addicts. Int J Leg Med 1993; 106:119-123.
    239) Wunsch MJ : A chart review comparing paregoric to methadone in the treatment of neonatal opioid withdrawal. J Addict Dis 2006; 25(3):27-33.
    240) Yaster M , Deshpande JK , & Maxwell LG : The pharmacologic management of pain in children. Compr Ther 1989; 15(10):14-26.
    241) Zanin A, Masiero S, Severino MS, et al: A delayed methadone encephalopathy: clinical and neuroradiological findings. J Child Neurol 2010; 25(6):748-751.
    242) Zuckerman M, Weisberg SN, & Boyer EW: Pitfalls of intranasal naloxone. Prehosp Emerg Care 2014; 18(4):550-554.
    243) Zuspan GP, Gumpel JA, & Mejia-Zelaya A: Fetal stress from methadone withdrawal. Am J Obstet Gynecol 1975; 122:43-46.
    244) de Giacomo M, Gaspari R, & Stefanelli A: Emergency therapeutical approach simulating ultrarapid opioid detoxification in methadone withdrawal precipitated by erroneous administration of naltrexone. Eur J Emerg Med 1999; 6:153-155.
    245) van Gaalen FA, Compier EA, & Fogteloo AJ: Sudden hearing loss after a methadone overdose. Eur Arch Otorhinolaryngol 2009; 266(5):773-774.
    246) van den Beuken-van Everdingen MH, Geurts JW, & Patijn J: Prolonged QT interval by methadone: relevance for daily practice? A prospective study in patients with cancer and noncancer pain. J Opioid Manag 2013; 9(4):263-267.