MOBILE VIEW  | 

OPIOIDS/OPIOID ANTAGONIST

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) Opioid agonists are analgesic drugs that bind and activate various opioid receptor sites, including mu, kappa, delta, and lambda. A weak or partial agonist at an opioid receptor may act as an antagonist at that receptor site.
    B) The following agents and opioid withdrawal have their own specific managements, please refer to them as indicated.
    1) Body packing and body stuffing, particularly concerning heroin
    2) Buprenorphine, a partial agonist opioid
    3) Butorphanol, a partial agonist-antagonist opioid
    4) Codeine, opioid agonist
    5) Dihydrocodeine
    6) Fentanyl, opioid agonist
    7) Hydrocodone, opioid agonist
    8) Hydromorphone, opioid agonist
    9) Levorphanol, opioid agonist
    10) Long-acting opioid antagonists (nalmefene, naltrexone, nalmexone)
    11) Meperidine, a narcotic analgesic with actions similar to morphine
    12) Methadone and related agents, including LAAM, with a long duration of action and used in the management of opiate withdrawal
    13) Morphine, an pure opioid agonist
    14) Naloxone, an opioid receptor antagonist
    15) Opioid withdrawal
    16) Oxycodone, a pure opioid agonist
    17) Oxymorphone
    18) Pentazocine
    19) Propoxyphene (recommended removed from the US market (November 2010))
    20) Tramadol

Specific Substances

    A) OPIATES
    1) Opioids
    2) Opioid antagonists
    ALFENTANIL HYDROCHLORIDE (synonym)
    1) R-39209
    2) CAS 71195-58-9 (alfentanil)
    3) CAS 69049-06-5 (alfentanil hydrochloride, anhydrous)
    4) CAS 70879-28-6 (alfentanil hydrochloride, monohydrate)
    ALPHAPRODINE (synonym)
    1) Alphaprodine
    ALVIMOPAN (synonym)
    1) ADL-8-2698
    2) Alvimopanum
    3) LY-246736
    4) CAS 156053-89-3 (anhydrous alvimopan)
    5) CAS 170098-38-1 (alvimopan dihydrate)
    ANILERIDINE HYDROCHLORIDE (synonym)
    1) CAS 126-12-5 (anileridine hydrochloride)
    2) CAS 144-14-9 (anileridine) (synonym)
    BUTYRFENTANYL (synonym)
    1) CAS 1169-70-6
    DIPRENORPHINE (synonym)
    1) CAS 14357-78-9
    ETORPHINE (synonym)
    1) 6,14-endo-Ethenotetrahydrooripavine
    2) 7-alpha-(1-hydroxy-1-methylbutyl)-
    3) CAS 14521-96-1
    HEROIN (synonym)
    1) Diacetylmorphine hydrochloride
    2) Diamorphine hydrochloride
    3) Morphine diacetyl
    4) Diacetylmorphin
    5) Morphin diacetyl
    6) CAS 561-27-3 (diamorphine)
    7) CAS 1502-95-0 (diamorphine hydrochloride)
    KETOBEMIDONE HYDROCHLORIDE (synonym)
    1) Cetobemidon
    2) Cetobemidone
    3) Ciba 7115
    4) Cliradon
    5) Cliradone
    6) Cymidon
    7) Hoechst 10720
    8) Molecular Formula: C15-H21-N-O2
    9) CAS 469-79-4 (ketobemidone)
    10) CAS 5965-49-1 (ketobemidone hydrochloride)
    MEPTAZINOL HYDROCHLORIDE (agonist/antagonist) (synonym)
    1) IL-22811 (meptazinol)
    2) WY-22811 (meptazinol)
    3) CAS 54340-58-8 (meptazinol)
    4) CAS 59263-76-2 (meptazinol hydrochloride)
    5) CAS 34154-59-1 (+/--meptazinol hydrochloride)
    METHYLFENTANYL (synonym)
    1) Methylfentanyl
    NALBUPHINE HYDROCHLORIDE (agonist/antagonist) (synonym)
    1) EN-2234A
    2) CAS 20594-83-6 (nalbuphine)
    3) CAS 23277-43-2 (nalbuphine hydrochloride)
    NALORPHINE HYDROCHLORIDE (synonym)
    1) CAS 57-29-4 (nalorphine hydrochloride)
    2) CAS 62-67-9 (nalorphine)
    OPIUM (synonym)
    1) Gum Opium
    2) Opium, Powdered
    3) Papaver somniferum (latex from unripe capsules)
    4) Raw Opium
    PAREGORIC (synonym)
    1) CAS 8029-99-0
    PHOLCODINE (synonym)
    1) Morpholinylethyl-morphine
    2) CAS 509-67-1
    REMIFENTANIL HYDROCHLORIDE (synonym)
    1) GI-87084B
    2) 4-Carboxyl-4-(N-phenylpropionamide)-1-piperidine
    3) propionic acid dimethyl ester monohydrate
    4) Molecular formula: C2-O-(H2)8-N2-O5,HCl
    5) CAS 132539-07-2
    SUFENTANIL CITRATE (synonym)
    1) R-33800
    2) R-30730 (sufentanil)
    3) CAS 56030-54-7 (sufentanil)
    4) CAS 60561-17-3 (sufentanil citrate)

    1.2.1) MOLECULAR FORMULA
    1) ALFENTANIL HYDROCHLORIDE: C21H33ClN6O3
    2) ELUXADOLINE: C32H35N5O5
    3) NALBUPHINE HYDROCHLORIDE: C21H27NO4
    4) SUFENTANIL CITRATE: C22H30N2O2S.C6H8O7 (Prod Info sufentanil citrate intravenous injection, epidural injection, 2011)

Available Forms Sources

    A) FORMS
    1) Most opioids are available in both oral and parenteral forms, both legally and illicitly.
    2) Street preparations of drugs may contain variable amounts of active drug causing an overdosage when the preparation is more concentrated than expected.
    3) The street preparations may contain adulterants such as strychnine, PCP, scopolamine, cocaine, clenbuterol, thiamine, dextromethorphan or quinine.
    B) SOURCES
    1) AH-7921: Nine patients were found dead after using AH-7921, a designer opioid with about 80% of morphine's mu-agonist activity. Six patients also used other drugs, such as benzodiazepines, antidepressants, and analgesics; however, it was concluded that poly-drug use (all agents with postmortem therapeutic concentrations) was not a major contributing factor for the fatal outcome (Kronstrand et al, 2014).
    2) ALPHAPRODINE is a synthetic opioid analgesic that is no longer marketed in the US. It has a potency and chemical structure similar to meperidine (Baselt, 2000).
    3) ALVIMOPAN, a selective antagonist of the cloned human mu-opioid receptor, is available orally for short-term use in hospitalized patients to accelerate the time to upper and lower gastrointestinal recovery following partial large or small bowel resection surgery or primary anastomosis (Prod Info ENTEREG(R) oral capsules, 2008).
    4) ANILERIDINE is a synthetic opioid analgesic that is no longer marketed in the US and is reported several times more potent than meperidine (Baselt, 2000).
    5) BUTYRFENTANYL
    a) Butyrfentanyl, a potent short-acting opioid and an analog of fentanyl, is only 0.13 the potency of fentanyl, but 7 times more potent than morphine. Cross-reactivity of fentanyl and butyrfentanyl on immunoassay is very high (Cole et al, 2015).
    6) BROWN
    a) In Europe, an illicit synthetic opiate mixture, called "Brown", has been abused and has resulted in fatalities. The mixture contains codeine (as a precursor), hydrocodone (target substance), and dihydrocodeine (reaction byproduct) (Balikova & Maresova, 1998).
    7) CHINA WHITE
    a) China White was originally used as the name for highly purified heroin from Asia; it may now mean on the street a relatively new abuse drug - alpha methyl fentanyl.
    b) This drug has an extremely fast onset and duration of 30 to 60 minutes. Overdoses of the drug need immediate respiratory support and treatment with naloxone.
    c) Tested in animals this agent was 1000 to 2000 times as potent as morphine. It is very difficult to isolate in the urine and will not be detected on toxicology screens (Brittain, 1982-83).
    d) Other potent analogs of fentanyl (i.e., parafluorofentanyl or 3-methyl fentanyl), have been sold as China White on the street (LaBarbera & Wolfe, 1983).
    e) Street samples of 3-methylfentanyl have yielded about 100 micrograms of 3-MF in a typical 20-milligram packet (average purity 0.47%) (Esposito & Winek, 1991).
    8) DESOMORPHINE
    a) "Crocodile" (also known as Krokodil or Russian heroin) is a Russian slang term for homemade desomorphine, an opioid analogon with analgesic, muscle-relaxing, sedating, and euphoric properties. It is an inexpensive designer drug that originated in Russia and has now been found in Germany (among immigrants from Russia), Czech Republic, Ukraine, France, Belgium, Sweden and Norway (Gahr et al, 2012; Skowronek et al, 2012; Gahr et al, 2012a). Krokodil has also been found in the US.
    b) "Crocodile" (Krokodil) is mainly used by opiate drug addicts, as a cheaper alternative to heroin. Crocodile is prepared by boiling 5 to 10 codeine-containing tablets with a diluting agent (usually a paint thinner that may contain lead, zinc, ferric or ferrous agents and antimony), lighter fuel (gasoline), hydrochloric acids, iodine, and red phosphorus (as a phosphate source) which are scraped from the striking surfaces on matchboxes. Desomorphine is created from codeine (3-methylmorphine) by 2 intermediate steps (alpha-chlorocodide and desocodeine); the final product is a suspension that contains desomorphine and the other agents used in the process. It is administered by IV or IM. It purportedly is 8 to 15 times more potent than morphine with weaker convulsant, emetic and respiratory depressant effects. Onset of action is 2 to 3 minutes with a short duration of action (2 hours); onset and elimination half-life are faster than morphine. Individuals may show signs of addiction within 5 to 10 days. Severe toxicity is usually due to impurities. It may cause severe tissue damage, phlebitis, gangrene, and limb ischemia leading to amputation following injection. The slang term, "Krokodil" reportedly refers to the user's greenish and scaly skin at the site of injection (similar in appearance to a crocodile's skin). Deaths have occurred. Users of the drug may have a characteristic smell of iodine that is used during the production of the drug. As of June 2012, Russia has banned the sale of codeine-containing tablets as over-the-counter products in an attempt to reduce production of this drug (Gahr et al, 2012; Skowronek et al, 2012; Gahr et al, 2012a).
    9) ETORPHINE
    a) Etorphine is a derivative of the opium alkaloid thebaine used for immobilization of wildlife. Etorphine is highly potent and rapid acting.
    b) Minute amounts may exert serious effects leading to coma. It is absorbed via skin and mucous membranes. Accidental injection or needle scratches should be treated immediately with naloxone (JEF Reynolds , 1989).
    c) Diprenorphine hydrochloride is another reversal agent for etorphine. Due to side effects, it should not be used in humans.
    d) An injection of the contents of a needle occurred following a needle scratch in a 41-year-old man which produced dizziness, nausea and coma. Two 10-mg doses of nalorphine were required to reverse the coma (Firn, 1973).
    10) HEROIN
    a) A common drug of abuse, heroin is usually present in the street product at concentrations ranging from 21% to 60% (Gomez & Rodriguez, 1989).
    b) The remaining impurities and adulterants rarely cause significant clinical signs (Shesser et al, 1991); however, several cases of significant toxicity from adulterated heroin have been reported.
    1) An epidemic of heroin adulterated with scopolamine resulting in severe anticholinergic toxicity was reported (Hamilton et al, 2000).
    2) A case of amphetamine toxicity caused by use of adulterated heroin has been reported (Choudry & Doe, 1986).
    3) Clenbuterol has also been found as an adulterant in heroin (Hieger et al, 2015; U.S. Drug Enforcement Administration, 2009).
    c) Heroin exposure via inhalation of heated heroin vapors is referred to "chasing the dragon" (Hill et al, 2000).
    d) SUPERBUICK or HOMICIDE are two reported slang terms for heroin adulterated with any or all of the following: scopolamine, cocaine, caffeine, thiamine, or dextromethorphan. Overdoses may result in opiate and anticholinergic toxicities.
    e) Heroin substitutes have included a combination of pentazocine and the antihistamine tripelennamine. One case describes the fatal use of dextromethorphan with terfenadine as a heroin substitute (Kintz & Mangin, 1992).
    f) Metal contamination found in illicit heroin samples, that can increase the inherent toxicity of the opioid, has included cadmium, zinc, copper, and iron (Infante et al, 1999).
    11) MT-45 DESIGNER OPIOID
    a) MT-45 (DESIGNER OPIOID): An observational case series was conducted, involving 9 patients identified with a psychoactive substance intoxication, later identified as MT-45, a designer opioid, as either the sole drug or in combination with other agents (eg, alcohol, phencyclidine, oxycodone, benzodiazepines). Reported symptoms included paresthesia in the extremities, severe CNS depression, respiratory depression, cyanosis, miosis, and impaired vision and hearing. Routes of administration included oral, IV, nasal, and rectal. Naloxone was administered to 7 of the 9 patients, with 4 of the patients responding well to the treatment (Helander et al, 2014).
    12) OTHER
    a) MEPTAZINOL HYDROCHLORIDE is an analgesic with narcotic agonist activity.
    b) OPIUM: Deodorized tincture of opium contains 10 mg/mL of anhydrous morphine.
    c) PAREGORIC is camphorated tincture of opium. Each mL contains 0.4 mg of anhydrous morphine.
    d) REMIFENTANIL is available in the United States as a lyophilized powder in concentrations of 1 mg/3 mL vial, 2 mg/5 mL vial, and 5 mg/10 mL vial (Prod Info Ultiva(R), remifentanil hydrochloride, 1996).
    13) FOLK REMEDIES/ALTERNATIVE MEDICINE
    a) HMONG FOLK REMEDIES may contain combinations of opioids with other substances. The purported ingredients of the backache remedy include acetylated opioids and heroin, as well as, derivatives of aspirin and acetaminophen (Smith & Nelsen, 1991).
    b) GEE'S LINCTUS COUGH is a pharmacy only cough mixture found throughout New Zealand that contains anhydrous morphine, squill (obtained from the Urginea maritima plant and contains several cardiac glycosides), and ethanol. There was a case of cardiac toxicity (AV conduction block) and proximal myopathy (severe muscle weakness) in an adult that ingested 200 mL of the cough syrup daily for over 4 months. No permanent effects were reported (Griffiths et al, 2009).
    14) WITHDRAWAL FROM MARKET
    a) PALLADONE: The US FDA reports that the marketing of Palladone(R), Purdue Pharma, has been suspended because of the potential for severe adverse events when combined with alcohol. Palladone, a time-release formulation of hydromorphone hydrochloride, when coingested with alcohol has been shown to produce the rapid release of hydromorphone leading to increased blood concentrations approximately 6 times that observed with the coingestion of water. It is suggested that alcohol may accelerate the breakdown of the capsule resulting in a "dose dumping" phenomenon. This phenomenon may also occur with other slow-release opioids (eg, Roxanol SR (morphine sulfate), Oxycontin SRT (oxycodone), Hydromorph Contin (hydromorphone)) when combined with alcohol (Murray & Wooltorton, 2005; FDA MedWatch Safety Alert Palladone, 2005).
    C) USES
    1) Opioids are primarily used for the treatment of pain, less often for cough suppression. Opioids are commonly abused for euphoric effects by multiple routes (ie, injection, insufflation, smoking, ingestion and transdermal). The following agents and opioid withdrawal have their own specific managements, please refer to them as indicated: buprenorphine, butorphanol, codeine, dihydrocodeine, fentanyl, hydrocodone, hydromorphone, levorphanol, long-acting opioid antagonists (nalmefene, naltrexone, nalmexone), meperidine, methadone, methadone, oxycodone, pentazocine, propoxyphene, tramadol, and naloxegol.
    2) Alvimopan is indicated as short-term, in-hospital treatment for the acceleration of upper and lower gastrointestinal recovery following partial bowel resection surgery with primary anastomosis. Alvimopan effectively manages this condition without compromising analgesia (Prod Info ENTEREG(R) oral capsules, 2013).

Life Support

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

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: Opioids are primarily used for the treatment of pain, less often for cough suppression. Opioids are commonly abused for euphoric effects by multiple routes (ie, injection, insufflation, smoking, ingestion and transdermal). The following agents and opioid withdrawal have their own specific managements, please refer to them as indicated: buprenorphine, butorphanol, codeine, dihydrocodeine, fentanyl, hydrocodone, hydromorphone, levorphanol, long-acting opioid antagonists (nalmefene, naltrexone, nalmexone), meperidine, methadone, methadone, oxycodone, pentazocine, propoxyphene, tramadol, and naloxegol.
    B) PHARMACOLOGY: Opioids are a group of chemical substances, naturally occurring and synthetic, that bind at the opiate receptor. Opiates are a group of naturally occurring compounds derived from the poppy, Papaver somniferum.
    C) TOXICOLOGY: Therapeutic and toxic effects are mediated by different opioid receptors. Mu 1: Supraspinal and peripheral analgesia, sedation and euphoria. Mu 2: Spinal analgesia, respiratory depression, physical dependence, GI dysmotility, bradycardia and pruritus. Kappa 1: Spinal analgesia and miosis. Kappa 2: Dysphoria and psychotomimesis. Kappa 3: Supraspinal analgesia. Chronic opioid users develop tolerance to the analgesic and euphoric effects, but not to the respiratory depression effects.
    D) EPIDEMIOLOGY: Overdose is common, particularly in patients with chronic opioid abuse, and often life threatening (especially a heroin or methadone overdose).
    E) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE POISONING: Euphoria, drowsiness, constipation, nausea, vomiting and miosis. Mild bradycardia or hypotension may be present.
    2) SEVERE POISONING: Respiratory depression leading to apnea, hypoxia, coma, bradycardia, or acute lung injury. Rarely, seizures may develop from hypoxia. Acute tubular necrosis secondary to rhabdomyolysis and myoglobinuria may develop in patients with prolonged coma or seizures. Death may result from any of these complications. Scleroderma following heroin abuse has been reported and may be linked to talc mixed with heroin. Wound botulism resulting in flaccid paralysis may occur following black tar heroin abuse. Hypoglycemia and leukocytosis have been reported in heroin abusers. Intraarterial injection can cause ischemia.
    3) DESOMORPHINE: Severe tissue damage, phlebitis, gangrene, and limb ischemia leading to amputation, and death have been reported following IV or IM injections of "Crocodile" (also known as Krokodil or Russian heroin), a Russian slang term for homemade desomorphine.
    0.2.3) VITAL SIGNS
    A) WITH POISONING/EXPOSURE
    1) In severe overdose, hypotension, and respiratory depression are common.
    0.2.20) REPRODUCTIVE
    A) Most opioids and opioid antagonists are classified as FDA pregnancy category B or C. Fetal physical dependence, withdrawal symptoms and respiratory difficulties may occur in infants born to mothers physically dependent on opioids. Neonatal withdrawal may be seen in the infants of addicted mothers 12 to 72 hours after birth. Infants may be dehydrated, irritable, and experience tremors and cry continually and may have diarrhea. In addition, there have been reports of severe fetal bradycardia, including life-threatening cases, following maternal administration of nalbuphine during pregnancy and/or labor and delivery.
    0.2.21) CARCINOGENICITY
    A) Bladder carcinomas have been reported in a higher proportion of opium smokers than in cigarette smokers or non-smokers. A case-control study conducted in Iran of 309 cases of gastric adenocarcinoma and 613 matched controls found that opium use was associated with an increased risk of gastric adenocarcinoma.

Laboratory Monitoring

    A) Monitor vital signs frequently, pulse oximetry, and continuous cardiac monitoring.
    B) Monitor for CNS and respiratory depression.
    C) Opioid plasma levels are not clinically useful or readily available. Urine toxicology screens may confirm exposure, but are rarely useful in guiding therapy; urine toxicology immunoassays may also miss synthetic opioids (eg, fentanyl and methadone).
    D) Obtain acetaminophen and salicylate levels in patients with a suspected overdose that may include combination products.
    E) Routine lab work is usually not indicated, unless it is helpful to rule out other causes or if the diagnosis of opioid toxicity is uncertain.
    F) Obtain a chest x-ray for persistent hypoxia. Consider a head CT and/or lumbar puncture to rule out an intracranial mass, bleeding or infection, if the diagnosis is uncertain.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) DECONTAMINATION
    1) PREHOSPITAL: Opioid overdoses are life threatening. Activated charcoal should be considered early after a significant oral ingestion, if a patient can protect their airway and is without significant signs of toxicity. If a patient is displaying signs of moderate to severe toxicity, do NOT administer activated charcoal because of the risk of aspiration.
    2) HOSPITAL: Consider activated charcoal if a patient presents soon after an ingestion and is not manifesting signs and symptoms of toxicity. Activated charcoal is generally not recommended in patients with significant signs of toxicity because of the risk of aspiration. Gastric lavage is not recommended as patients usually do well with supportive care.
    B) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Patients may only need observation.
    C) 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). Orotracheal intubation for airway protection should be performed early in cases of obtundation and/or respiratory depression that do not respond to naloxone.
    D) AIRWAY MANAGEMENT
    1) Administer oxygen and assist ventilation for respiratory depression. Orotracheal intubation for airway protection should be performed early in cases of obtundation and/or respiratory depression that do not respond to naloxone, or in patients who develop severe acute lung injury.
    E) ANTIDOTE
    1) NALOXONE, an opioid antagonist, is the specific antidote. Naloxone can be given intravascularly, intramuscularly, subcutaneously, intranasally or endotracheally. The usual dose is 0.4 to 2.0 mg IV. In patients with suspected opioid dependence, 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, but may be necessary in overdoses of high potency opioids, like fentanyl.
    2) A CONTINUOUS infusion may be necessary in patients that have ingested a long-acting opioid. A suggested starting rate is two-thirds of the dose effective for initial reversal that is administered each hour; titrate as needed. DURATION of effect is usually 1-2 hours. Many opioids have a much longer duration of effect, so it may be necessary to observe the patient at least 3-4 hours after the last dose of naloxone to ensure that the patient does not have recurrent symptoms of toxicity. Naloxone can precipitate withdrawal in an opioid-dependent patients, which is usually not life-threatening; however it can be extremely uncomfortable for the patient.
    F) SEIZURE
    1) Seizures are rare, but may be a result of hypoxia or due to properties of certain agents (eg, meperidine, tramadol, propoxyphene). Treatment includes ensuring adequate oxygenation, and administering intravenous benzodiazepines; propofol or barbiturates may be indicated, if seizures persist.
    G) ACUTE LUNG INJURY
    1) Acute lung injury can develop in a small proportion of patients after an acute opioid overdose. The pathophysiology is unclear. Patients should be observed for 4 to 6 hours after overdose to evaluate for hypoxia and/or the development of acute lung injury.
    H) HYPOTENSION
    1) Hypotension is often reversed by naloxone. Initially, treat with a saline bolus, if patient can tolerate a fluid load, then adrenergic vasopressors to raise mean arterial pressure.
    I) ENHANCED ELIMINATION
    1) Hemodialysis and hemoperfusion are not of value because of the large volume of distribution for opioids.
    J) INTRATHECAL INJECTION
    1) Limited experience. Treat seizures (eg, benzodiazepines, barbiturates, propofol) and support blood pressure with fluids and pressors as needed. Naloxone infusion may be useful. Intubate and ventilate as needed. Cerebrospinal fluid drainage may accelerate recovery.
    K) PATIENT DISPOSITION
    1) HOME CRITERIA: Respiratory depression may occur at doses just above the therapeutic dose. Children should be observed and evaluated in the hospital as they are generally opioid naive and may develop respiratory depression. Adults should be evaluated by a health care professional if they have received a higher than recommended (therapeutic) dose, especially if opioid naive.
    2) OBSERVATION CRITERIA: Patients with deliberate ingestions or a pediatric ingestion should be sent to a health care facility for observation for at least 4 hours, to ensure that peak plasma levels have been reached and there has been sufficient time for symptoms to develop. Patients that have ingested a sustained release or long acting product have the potential to manifest symptoms in a delayed fashion and should be observed for 24 hours. Patients who are treated with naloxone should be observed for 4 to 6 hours after the last dose, for recurrent CNS depression or acute lung injury.
    3) ADMISSION CRITERIA: Patients with significant, persistent central nervous system depression should be admitted to the hospital. A patient needing more than 2 doses of naloxone should be admitted as a longer-acting opioid has likely been taken; additional doses may be needed. Patients with coma, seizures, dysrhythmias, delirium, and those needing a naloxone infusion or who are intubated should be admitted to an intensive care setting.
    4) CONSULT CRITERIA: Consult a poison center or medical toxicologist for assistance in managing patients with severe toxicity or in whom the diagnosis is not clear.
    L) PITFALLS
    1) Patients may be discharged prematurely after mental status clears with a dose of naloxone. Naloxone's duration of effect may be much shorter than the duration of effect for many opioids. Other causes of altered mental status must be ruled out, such as hypoxia or hypoglycemia.
    M) PHARMACOKINETICS
    1) Opioids may be absorbed via many routes, most commonly oral or transdermal for pain control. Abusers may inject, snort or smoke an opioid (these routes rapidly achieve high serum levels), which can produce euphoria quickly and place the individual at risk for severe toxicity. Opioids slow GI motility, which may lead to prolonged absorption.
    N) DIFFERENTIAL DIAGNOSIS
    1) Overdose with other sedating agents (e.g., ethanol, benzodiazepine/barbiturate, antipsychotics); overdose with central alpha 2 agonists (e.g., clonidine, tizanidine, imidazoline decongestants); CNS infection; intracranial hemorrhage; hypoglycemia or hypoxia.
    0.4.3) INHALATION EXPOSURE
    A) Inhalation of opioids may result from intentional crushing and "snorting" of tablets or smoking heroin. Refer to ORAL exposure for further treatment guidelines.
    0.4.6) PARENTERAL EXPOSURE
    A) INTRATHECAL INJECTION: Limited experience. Treat seizures (ie, benzodiazepines, barbiturates, propofol) and support blood pressure with fluids and pressors as needed. Naloxone infusion may be useful. Intubate and ventilate as needed. Cerebrospinal fluid drainage may accelerate recovery. Refer to ORAL exposure for further treatment guidelines.

Range Of Toxicity

    A) TOXICITY: The toxicity of an opioid varies with the agent, as well as with tolerance developed from habitual use. Infants and children have unusual sensitivity to opioid agents.

Vital Signs

    3.3.1) SUMMARY
    A) WITH POISONING/EXPOSURE
    1) In severe overdose, hypotension, and respiratory depression are common.
    3.3.3) TEMPERATURE
    A) WITH POISONING/EXPOSURE
    1) Cotton fever is a febrile reaction that develops in patients who inject intravenous drugs that have been filtered through cotton balls (Harrison & Walls, 1990).
    a) Fever in a patient with a history of injection drug use should be presumed to be secondary to infection until proven otherwise.
    2) HYPOTHERMIA: Hypothermia has been reported following overdoses of opiates, likely secondary to prolonged coma in a cold environment (Gober et al, 1979; Lawrenson et al, 1993; Arvanitis & Satonik, 2002).
    a) Hypothermia has been reported in the setting of opioid overdose secondary to attempts by bystanders to revive patients by immersion in ice water (Osterhoudt & Perrone, 2002).
    3.3.4) BLOOD PRESSURE
    A) WITH POISONING/EXPOSURE
    1) In severe overdose, hypotension is common (Prod Info hydromorphone hcl oral tablets, 2003; Prod Info AVINZA(R) extended-release oral capsules, 2005).

Heent

    3.4.3) EYES
    A) WITH POISONING/EXPOSURE
    1) MIOSIS: Pupils are usually pinpoint although may be dilated in severe acidemia, hypoxia, hypotension, bradycardia, respiratory depression in mixed overdoses with anticholinergic or sympathomimetic drugs.
    a) INCIDENCE: In a retrospective study of 726 patients, 94% of opioid overdose patients had pinpoint pupils (Sporer et al, 1996).
    2) MYDRIASIS: Pupils are normally pinpoint following an opioid overdose. However, in the presence of a mixed ingestion or severe and prolonged hypoxia, which may precipitate severe hypotension and shock, mydriasis may occur (Jaffe & Martin, 1990).
    3) CORTICAL BLINDNESS has been reported as a consequence of heroin-induced anoxic encephalopathy (McDonald et al, 1995).
    4) MT-45 (DESIGNER OPIOID): An observational case series was conducted, involving 9 patients identified with a psychoactive substance intoxication, later identified as MT-45, a designer opioid, as either the sole drug or in combination with other agents (eg, alcohol, phencyclidine, oxycodone, benzodiazepines). Miosis and impaired vision were two of the reported symptoms (Helander et al, 2014).
    3.4.4) EARS
    A) WITH POISONING/EXPOSURE
    1) HEARING LOSS
    a) CHRONIC ABUSE: Rapidly developing profound bilateral sensorineural hearing loss, developing over several weeks, has occurred following chronic abuse of high dose acetaminophen with opioids. In 2 patients, vestibular function appeared to be spared, despite profound hearing loss. Audiometric testing suggested that the sensory end organ was the site of damage. Cochlear implantation was required in one patient to restore functional hearing (Oh et al, 2000).
    b) HEROIN: A 27-year-old man with a history of hepatitis C virus infection and heroin abuse in abstinence for 3 years, presented with coma, depressed respiration, increased pulse rate, contracted pupils and vomiting after drinking 4 bottles of vodka and sniffing heroin. He also had hypoxia and hypercapnia, requiring oxygen administration. Following supportive care, including antibiotics for aspiration pneumonia, he regained consciousness 16 hours later and complained of sudden hearing loss on both ears. Standard pure-tone audiometry revealed symmetrical moderate bilateral sensorineural hearing loss. Both otoacoustic emissions and auditory brainstem response waves were absent, indicating both cochlear and retrocochlear involvement. All other physical, neurological and laboratory tests were normal. Following treatment with prednisolone (for 15 days) and magnesium (for 30 days), his symptoms gradually improved. On one month follow-up, audiometry, auditory brainstem responses and otoacoustic emissions were normal (Antonopoulos et al, 2012).
    c) HEROIN: A 29-year-old woman with a history of heroin and cocaine abuse, presented with bilateral hearing loss and severe tinnitus after a heroin overdose. On presentation, she was agitated and distractible. She was treated with a course of prednisone and fitted with hearing aids. Her hearing loss gradually improved (Nair et al, 2010).
    d) COMBINATION EXPOSURE: An 18-year-old woman developed moderately severe bilateral sensorineural hearing loss after abusing heroin, benzodiazepine, alcohol, and crack (smoked cocaine) for 2 days. Following supportive therapy, including high-dose prednisone for a month and pentoxifylline for 10 months, her hearing sensitivity gradually improved, except for residual high-frequency sensorineural hearing loss (Schweitzer et al, 2011).
    e) MT-45 (DESIGNER OPIOID): An observational case series was conducted, involving 9 patients identified with a psychoactive substance intoxication, later identified as MT-45, a designer opioid, as either the sole drug or in combination with other agents (eg, alcohol, phencyclidine, oxycodone, benzodiazepines). Impaired hearing was one of the reported symptoms (Helander et al, 2014).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) HYPOTENSIVE EPISODE
    1) WITH THERAPEUTIC USE
    a) Hypotension from opioids may be due to opioid-induced arteriolar and venous dilation (Nelson, 1998).
    2) WITH POISONING/EXPOSURE
    a) Hypotension and shock may occur, especially in the presence of prolonged and severe hypoxia (Miller, 1980; Whipple et al, 1994; Lawrenson et al, 1993).
    b) Hypotension and a decrease in mental status has occurred following overdose (Whipple et al, 1994). Severe hypotension (70/40 mmHg) and ventricular fibrillation has been reported at 12 hours following a mixed intravenous overdose of cocaine and heroin (McCann et al, 2002).
    c) EPIDURAL: Accidental high thoracic epidural doses of sufentanil and bupivacaine resulted in severe hypotension and increased sensory and motor blockade (Wolff et al, 1992).
    d) CASE REPORT: Profound circulatory shock was reported in a 17-year-old girl several hours following an intravenous heroin overdose. Systolic central venous pressure of 21 mmHg was recorded with heart rate of 156 beats per minute. Severe depression of myocardial contractility was noted. Volume loading and high dose dobutamine successfully reversed the shock, and recovery was then uneventful (Remskar et al, 1998).
    B) ATRIAL FIBRILLATION
    1) WITH POISONING/EXPOSURE
    a) Atrial fibrillation has been reported following abuse of crude heroin in an adult man (Lawrenson et al, 1993)
    C) CONDUCTION DISORDER OF THE HEART
    1) WITH POISONING/EXPOSURE
    a) VENTRICULAR DYSRHYTHMIAS
    1) COCAINE/HEROIN: A mixed intravenous overdose of cocaine and heroin resulted in ventricular fibrillation and asystole (attributed to hyperkalemia and rhabdomyolysis). Administration of naloxone may have been contributory to the dysrhythmic effects (McCann et al, 2002).
    D) INTRA-ARTERIAL INJECTION
    1) Intraarterial injection can cause severe pain, edema, petechiae and arterial occlusion. In severe cases, compartment syndrome and gangrene have been reported (Gaspar & Hare, 1972; Funk et al, 1999; DelGiudice et al, 2005).
    E) BRADYCARDIA
    1) WITH THERAPEUTIC USE
    a) Bradycardia may be seen and is due to an associated reduction in central nervous system stimulation (Nelson, 1998).
    2) WITH POISONING/EXPOSURE
    a) Bradycardia may develop in patients with severe respiratory depression (Prod Info AVINZA(R) extended-release oral capsules, 2005).
    b) INCIDENCE: Only 2% of 726 patients presented with bradycardia following opioid overdoses as reported in a retrospective study of opioid overdoses in an urban setting (Sporer et al, 1996).
    F) TACHYARRHYTHMIA
    1) WITH POISONING/EXPOSURE
    a) Tachycardia (heart rate, 156 beats per minute) and ECG readings of sinus tachycardia and nonspecific ST-T segment changes were reported in a previously healthy 17-year-old girl following an IV heroin overdose (Remskar et al, 1998).
    G) MYOCARDIAL NECROSIS
    1) WITH POISONING/EXPOSURE
    a) Myocardial damage, with focal lesions formed by small mononuclear inflammatory cells and with degenerated, necrotic myocardial fibers and congestion, has been shown to occur as a result of prolonged hypoxic coma following opiate intoxication (Melandri et al, 1996).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) ACUTE RESPIRATORY INSUFFICIENCY
    1) WITH POISONING/EXPOSURE
    a) SUMMARY
    1) Respiratory depression and apnea are characteristic effects of opioid overdose and when severe may result in severe hypoxia, leading to hypotension and shock, acute lung injury and respiratory arrest (Wilkes et al, 1981; Jaffe & Martin, 1990; McCann et al, 2002a).
    2) INCIDENCE: Respiratory rates of less than 8 beat per minute were reported in more than 60% of opioid overdose patients in 2 large studies (Sporer et al, 1996; Cook et al, 1998).
    b) SPECIFIC AGENTS
    1) OTHER
    a) ALFENTANIL: Following parenteral misuse of alfentanil/midazolam (2:1 mixture), a 27-year-old died due to asphyxia from respiratory depression (Bralthwaite et al, 2000).
    b) MEPTAZINOL: Respiratory depression that was NOT reversible by naloxone 10 mg was reported in a 61-year-old woman who ingested 50 meptazinol 200 mg tablets (Davison et al, 1987).
    c) MT-45 (DESIGNER OPIOID): An observational case series was conducted, involving 9 patients identified with a psychoactive substance intoxication, later identified as MT-45, a designer opioid, as either the sole drug or in combination with other agents (eg, alcohol, phencyclidine, oxycodone, benzodiazepines). Reported symptoms included paresthesia in the extremities, severe CNS depression, respiratory depression, cyanosis, miosis, and impaired vision and hearing. Routes of administration included oral, IV, nasal, and rectal. Naloxone was administered to 7 of the 9 patients, with 4 of the patients responding well to the treatment (Helander et al, 2014).
    d) REMIFENTANIL: A primiparous woman who had received epidural fentanyl with bupivacaine (three 10-mL dose of bupivacaine 0.1% with fentanyl 2 mcg/mL) for labor pain, became unconscious and developed respiratory arrest within 5 minutes of self-administering the first dose of remifentanil (programmed regimen: 40 mcg/mL, 1-mL bolus [40 mcg], 2-min lockout) patient-controlled analgesia (PCA). She recovered within 30 to 60 seconds of supportive care. The PCA machine was examined and it was determined that 160 mcg was inadvertently delivered over 4 attempts (Pruefer & Bewlay, 2012).
    e) OPIOID TOLERANT INDIVIDUALS: One study evaluated the time course and severity of respiratory depression after the use of injected opioids (diamorphine or methadone) and oral opioids (methadone or sustained release oral morphine) in 10 opioid-tolerant addicts (including 9 patients with chronic obstructive pulmonary disease [COPD]). Pulse oximetry (SpO2%), end-tidal CO2 (ETCO2%), and parasternal intercostal muscle electromyogram recordings were used to detect acute reduction in neural respiratory drive (NRD) and to determine the value of advanced physiological monitoring over pulse oximetry alone. Overall, opioid drugs administered to treat opioid addiction caused significant acute respiratory depression. After the injectable opioids, increases in ETCO2% revealed significant respiratory depression at 30 minutes in 8 of the 10 patients. However, SpO2% revealed that only 4 of the 10 had significant respiratory depression, with small absolute changes in SpO2% at 30 minutes. After the use of injectable opioids, a non-statistically significant reduction in NRD from baseline to 30 minutes was also noted. There was a significant inverse relationship between baseline NRD and opioid-induced decrease in SpO2% (Jolley et al, 2015).
    B) ACUTE LUNG INJURY
    1) WITH POISONING/EXPOSURE
    a) Acute lung injury (pulmonary edema) may occur after severe opioid overdose (Glassroth et al, 1987; Jaffe & Martin, 1990; Dettmeyer et al, 2000; Warner-Smith et al, 2001; Sporer & Dorn, 2001) and may be a contributing factor in fatal overdoses.
    b) INCIDENCE: In a retrospective study of 125 patients admitted to an urban ED with heroin overdose, 13 (10.4%) developed acute lung injury. Patients who developed acute lung injury had a lower respiratory rate on paramedic arrival, and had a shorter duration of heroin use than patients who did not develop acute lung injury. All patients who developed acute lung injury received naloxone in the prehospital setting (Sterrett et al, 2003).
    c) BUTYRFENTANYL: CASE REPORT: An 18-year-old man with a past medical history of IV heroin abuse, developed hemoptysis, acute lung injury, hypoxic respiratory failure, and diffuse alveolar hemorrhage, after snorting an unknown amount of butyrfentanyl instead of acetyl fentanyl. Initially, he was found unconscious by his mother. His mental status improved after receiving 0.4 mg IV naloxone; however, he developed dyspnea and hemoptysis. A chest radiograph revealed bilateral "batwing"-shaped perihilar predominant airspace opacities with diffuse increased interstitial markings. Fentanyl and opiates immunoassays were both positive, but fentanyl was not present in high-performance liquid chromatography and mass spectrometry. Butyrfentanyl was detected using a high-performance gas chromatography obtained from the Bureau of Criminal Apprehension. At this time, his condition deteriorated, requiring intubation. Results from the cytology of the fluid from bronchoalveolar lavage obtained during bronchoscopy were consistent with diffuse alveolar hemorrhage. Echocardiography showed an ejection fraction of 55% to 60% with no wall-motion abnormalities. He was treated with fentanyl, midazolam, and dexmedetomidine to maintain adequate sedation. Following further supportive care, his condition improved gradually and he was extubated on day 4 and discharged 3 days later after receiving treatment for ventilator-associated pneumonia (Cole et al, 2015).
    d) HEROIN: Non-cardiogenic pulmonary edema ("heroin-lung") is an infrequent, but severe, complication of heroin overdose and is generally abrupt in onset (immediate-2 hours) following intravenous heroin overdose (Duberstein & Kaufman, 1971). Delayed onset has been described 4 hours following intranasal heroin use (Steinberg & Karlinger, 1968) and 3, 9, 14 and 24 hours following oral methadone ingestions (Wilen et al, 1975; Sey et al, 1971).
    1) Manifestations of heroin-induced pulmonary edema include rales, pink frothy sputum, significant hypoxia and bilateral fluffy infiltrates on chest x-ray. Some patients require mechanical ventilation.
    2) Resolution of symptoms usually occurs rapidly with supportive care alone, within hours to 1 to 2 days (Sporer & Dorn, 2001; Sporer, 1999). Heroin mixed with diphenhydramine to prevent anaphylaxis, still caused pulmonary edema in a significant percentage of patients in a Taiwan study (Chan et al, 1995). The most common complication of severe heroin overdose is pulmonary edema (Warner-Smith et al, 2001).
    e) CASE REPORT (HEROIN): An man presented with acute lung injury following an opiate overdose from smoking crude heroin. Pulmonary arterial wedge pressure was 12 mmHg with a cardiac output of 6.8 L/min (Lawrenson et al, 1993).
    f) CASE REPORT (HEROIN): Symmetric alveolar pulmonary edema was shown on chest x-ray in a 17-year-old girl several hours after an intravenous heroin overdose (Remskar et al, 1998).
    g) CASE REPORT (PAREGORIC): A 3-week-old infant who received 4 drops 6 times a day of paregoric developed pulmonary edema. With normal recovery, the majority of x-ray changes resolved within 24 to 48 hours (Rice, 1984).
    C) RESPIRATORY TRACT HEMORRHAGE
    1) WITH POISONING/EXPOSURE
    a) BUTYRFENTANYL: CASE REPORT: An 18-year-old man with a past medical history of IV heroin abuse, developed hemoptysis, acute lung injury, hypoxic respiratory failure, and diffuse alveolar hemorrhage, after snorting an unknown amount of butyrfentanyl instead of acetyl fentanyl. Initially, he was found unconscious by his mother. His mental status improved after receiving 0.4 mg IV naloxone; however, he developed dyspnea and hemoptysis. A chest radiograph revealed bilateral "batwing"-shaped perihilar predominant airspace opacities with diffuse increased interstitial markings. Fentanyl and opiates immunoassays were both positive, but fentanyl was not present in high-performance liquid chromatography and mass spectrometry. Butyrfentanyl was detected using a high-performance gas chromatography obtained from the Bureau of Criminal Apprehension. At this time, his condition deteriorated, requiring intubation. Results from the cytology of the fluid from bronchoalveolar lavage obtained during bronchoscopy were consistent with diffuse alveolar hemorrhage. Echocardiography showed an ejection fraction of 55% to 60% with no wall-motion abnormalities. He was treated with fentanyl, midazolam, and dexmedetomidine to maintain adequate sedation. Following further supportive care, his condition improved gradually and he was extubated on day 4 and discharged 3 days later after receiving treatment for ventilator-associated pneumonia (Cole et al, 2015).
    D) PNEUMONIA
    1) WITH POISONING/EXPOSURE
    a) Persistence of abnormalities beyond 24 to 48 hours including atelectasis, fibrosis, bronchiectasis, granulomatous disease, and pneumomediastinum suggest the presence of aspiration or bacterial pneumonitis (Saba et al, 1974; Wilen et al, 1975; Glassroth et al, 1987). A frequent autopsy finding in heroin fatalities is undiagnosed pneumonia (Warner-Smith et al, 2001).
    E) LUNG FINDING
    1) WITH POISONING/EXPOSURE
    a) BULLOUS PULMONARY DAMAGE: Bullous pulmonary damage was found in 2 percent of a group of 387 illicit intravenous drug users. The drugs used by these patients were not specified (Goldstein et al, 1986).
    F) BRONCHOSPASM
    1) WITH POISONING/EXPOSURE
    a) Airway obstruction and bronchospasm, or status asthmaticus, have been associated with inhalation of heated heroin. Eosinophils have been detected in the peripheral blood and/or respiratory secretions in these patients. More investigation is needed to determine the factors and the mechanisms involved (Del Los Santos-Sastre et al, 1986; Cygan et al, 2000).
    b) HEROIN
    1) Bronchospasm and wheezing have been reported in both intravenous and inhalational abusers of heroin (Anderson, 1986; Oliver, 1986; Cygan et al, 2000).
    2) A temporal relationship between heroin insufflation and precipitation or exacerbation of acute bronchospasm in patients with previous histories of asthma was reported (Prachand et al, 1999).
    3) Three cases of severe acute asthma following inhalation of heroin vapor were reported. Fatal respiratory arrest occurred in two of these patients (Hughes & Calverley, 1988).
    G) HYPOXEMIA
    1) WITH POISONING/EXPOSURE
    a) A marked increase in cerebral deoxygenated hemoglobin following heroin IV injections in 7 heroin-dependent patients was reported (Hock et al, 2002).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) COMA
    1) WITH POISONING/EXPOSURE
    a) Acute overdosage of any of the opioid drugs may result in coma, pulmonary edema and cessation of respiration, depending on the dose and degree of tolerance. Prolonged coma may result due to delayed gastric emptying (Melandri et al, 1996a). .
    b) INCIDENCE: 64% of 52 patients in a case series of acute opiate overdoses admitted to an ED were reported to have a rating of <8 on the Glasgow Coma Scale (Cook et al, 1998).
    c) REMIFENTANIL: A primiparous woman who had received epidural fentanyl with bupivacaine (three 10-mL dose of bupivacaine 0.1% with fentanyl 2 mcg/mL) for labor pain, became unconscious and developed respiratory arrest within 5 minutes of self-administering the first dose of remifentanil (programmed regimen: 40 mcg/mL, 1-mL bolus [40 mcg], 2-min lockout) patient-controlled analgesia (PCA). She recovered within 30 to 60 seconds of supportive care. The PCA machine was examined and it was determined that 160 mcg was inadvertently delivered over 4 attempts (Pruefer & Bewlay, 2012).
    d) CASE REPORT: A 25-year-old woman with a 2-year history of heroin inhalation was found comatose in her car. On presentation to the ED, she had a Glasgow Coma Scale score (GCS) of 6, a heart rate of 109 beats/min, and a blood pressure of 168/44 mmHg. Neuroimaging studies revealed hydrocephalus with enlarged ventricles, communicating ventriculomegaly, and cerebellar enhancement. All laboratory results, serial CT scans, and a second MRI were normal. Her cerebellar radiologic changes were improved after an external ventricular drain (EVD) was placed; however, she developed tremor and cogwheeling. She received acetazolamide 500 mg every 12 hours and dexamethasone to treat hydrocephalus. During multiple EVD clamping trials, her intracranial pressures increased and she became obtunded. On day 13 of hospitalization, she received a ventriculoperitoneal shunt (VPS) and she was discharged to a rehabilitation facility with a GCS of 15. On a 3-month follow-up, her symptoms had improved, but she sustained cognitive deficits with a low-pressure headache. Large improvement and increased pachymeningeal enhancement in postgadolinium T1 sequences were observed in a brain MRI (Bui et al, 2015).
    B) DROWSY
    1) WITH POISONING/EXPOSURE
    a) Decreased mental status is one of the most prominent symptoms in a narcotic overdose, which may progress to coma (Whipple et al, 1994).
    b) CASE REPORT: A 52-year-old woman developed only nausea and mild drowsiness after inadvertently receiving 3 mL (3 mg) of diamorphine intrathecally instead of 0.5% bupivacaine for spinal anesthesia for a total knee replacement surgery. She recovered after 48 hours of observation (Jafar & Chowdhry, 2014).
    C) SEIZURE
    1) WITH THERAPEUTIC USE
    a) ALFENTANIL: A case of jerking movements progressing to seizures and unconsciousness was reported in a 60-year-old man given 25 mcg/kg alfentanil over 30 seconds (Strong & Matson, 1989).
    2) WITH POISONING/EXPOSURE
    a) Seizures may occur following overdoses, but are unusual following therapeutic dosing. Seizures may be related to hypoxia (Nelson, 1998). Rarely, use of naloxone to reverse opioid overdose has been reported to provoke seizures (Remskar et al, 1998).
    D) DRUG-INDUCED MYELOPATHY
    1) WITH POISONING/EXPOSURE
    a) Toxic Myelopathy
    1) CASE REPORT: Following an overdose of intranasal insufflated heroin and amphetamines (approximately 1 gram) a 17-year-old woman was admitted with an inability to walk and decreased level of consciousness. Weakness progressed to all four limbs over several hours, along with severe rhabdomyolysis (creatine phosphokinase 36,880 mg/dL) with acute renal failure and hepatic failure. Following supportive therapy, the patient was alert and cooperative with complaints of muscle pain, tenderness and weakness that progressed to flaccid, areflexic paralysis. An initial MRI was normal, but one month later the imaging showed selective T2 hyperintensity and intense enhancement confined to the spinal anterior horns and lumbar nerve roots and plexus. Based on physical and MRI findings the authors suggested that the initial process was likely motor neuron cell death in the spinal anterior horns, with subsequent involvement of the nerve roots and lumbar plexus due to wallerian degeneration. Four months after the initial admission, the patient remained a paraplegic(Riva et al, 2007).
    E) MYOCLONUS
    1) WITH THERAPEUTIC USE
    a) Myoclonic activity has been reported in two patients on high-dose spinal opioid therapy for pain (Parkinson et al, 1990).
    2) WITH POISONING/EXPOSURE
    a) A man developed leukoencephalopathy after heroin overdose presented with axial myoclonus involving the neck flexors, pectoral muscles and abdominal musculature (Hill et al, 2000).
    F) CENTRAL NERVOUS SYSTEM DEPRESSION
    1) WITH POISONING/EXPOSURE
    a) MT-45 (DESIGNER OPIOID): An observational case series was conducted, involving 9 patients identified with a psychoactive substance intoxication, later identified as MT-45, a designer opioid, as either the sole drug or in combination with other agents (eg, alcohol, phencyclidine, oxycodone, benzodiazepines). Reported symptoms included paresthesia in the extremities, severe CNS depression, respiratory depression, cyanosis, miosis, and impaired vision and hearing. Routes of administration included oral, IV, nasal, and rectal. Naloxone was administered to 7 of the 9 patients, with 4 of the patients responding well to the treatment (Helander et al, 2014).
    G) CEREBROVASCULAR DISEASE
    1) WITH POISONING/EXPOSURE
    a) Stroke may occur as an infrequent complication of heroin abuse (Vila & Chamorro, 1997).
    b) CASE REPORT: Ballistic movements after IV heroin overdose were reported in 2 adult patients. In one patient, an MRI revealed bilateral ischemic lesions of the globus pallidus, indicating generalized cerebral hypoxia during a comatose state. The second patient, with an acute left hemiballismus, had an MRI which showed an ischemic infarct in the right striatum. This patient was suspected to have an embolic stroke (Vila & Chamorro, 1997).
    c) CASE REPORT: A 28-year-old woman presented with altered mental status after using heroin. Physical examination revealed negative clonus, pinpoint and sluggish pupils, disorientation, persistent confusion, and expressive aphasia. A computed tomographic scan of the head revealed a large 5.1 x 5-cm intraparenchymal hemorrhage in the left frontal lobe, vasogenic edema, and a 5-mm midline shift. Surgical intervention was not considered because of the stable appearance of a repeat imaging. Despite a mild improvement in her cognitive function, she continued to be confused with significant memory loss during her hospitalization. She was discharged to a long-term facility with a diagnosis of hemorrhagic stroke secondary to heroin abuse after other etiologies were ruled-out (Kumar et al, 2015).
    H) LEUKOENCEPHALOPATHY
    1) WITH POISONING/EXPOSURE
    a) Heroin-induced spongiform leukoencephalopathy, a result of fluid accumulation within the myelin sheaths, has been reported following heroin intoxication, primarily after inhaling the pyrolysate generated by heating heroin base on aluminum foil ("chasing the dragon").
    b) In the initial stage of illness patients demonstrate soft speech, cerebellar ataxia, restlessness and apathy. During the intermediate stage pseudobulbar reflexes, spastic paresis, tremors, myoclonic jerking, hyperreflexia, worsening gait disturbances and choreoathetoid movements may develop. In the terminal stages central fevers, stretching spasms, hypotonia, areflexia and akinetic mutism may develop followed by brain death (Wolters et al, 1982). Progression occurs over weeks or months and not all patients progress through all stages. Mortality is approximately 25%.
    c) MRI is the diagnostic modality of choice. Diffusion-weighted (DW) magnetic resonance imaging (MRI) findings may show restriction of water diffusion in white matter at the intermediate stage of disease (Chen et al, 2000). A characteristic pattern may be seen on MRI (Kriegstein et al, 1997) consisting of spongiform demyelination as the morphological substrate of lesions which are hyperintense on T2-weighted MRI and hypodense on CT (Weber et al, 1998).
    d) Blood and CSF are typically normal, and cranial CT may be normal early in the course of illness (Long et al, 2003).
    e) Autopsy findings include spongiform degeneration of white matter, vacuolization and fluid accumulation in myelin sheaths (Long et al, 2003).
    f) CASE REPORTS
    1) Spongiform leukoencephalopathy was reported in two patients who were regular heroin smokers. Both had neurological signs (cerebellar ataxia, bilateral pyramidal signs, dysarthria); CT scan and autopsy showed extensive white-matter spongiosis and vacuolization (Sempere et al, 1991).
    2) Two more cases of heroin-induced progressive spongiform leukoencephalopathy following 6 month histories of inhaling heroin vapor were reported (Kriegstein et al, 1997). Heroin overdose via inhalation of heated heroin vapors has resulted in leukoencephalopathy with coma (Glasgow Coma Scale 6), axial myoclonus, diffuse hypotonia, rhabdomyolysis and death (Hill et al, 2000).
    3) Other cases of severe irreversible neurological illness characterized by a cerebellar syndrome and postmortem findings of spongiform encephalopathy after inhaling heroin pyrolysate or smoking heroin have been reported (Long et al, 2002; Weber et al, 1998; Schiffer et al, 1985; Wolters et al, 1982).
    4) A 40-year-old man presented with severe ataxia and dysarthria, without nystagmus or extremity tremor, after smoking heroin. MRI showed symmetric C shaped lesions in the deep cerebellar hemispheres. The patient's neurologic exam did not improve over the next 6 months (Ropper & Blair, 2003). This case is unusual in that there were no cerebral lesions on MRI.
    5) A 50-year-old man developed delayed post-hypoxic leukoencephalopathy after heroin abuse. On day 24, an MRI revealed diffuse abnormal increased T2 signal in the supratentorial white matter with diffuse restricted diffusion. Following supportive care, his condition slowly improved. On day 70, neuropsychometric testing (NPT) showed moderate to severe bihemispheric disturbance with impairments reflecting a decline in higher cognitive functioning. A second NPT on day 92 revealed significant improvement (Snow et al, 2015).
    I) PARESTHESIA
    1) WITH POISONING/EXPOSURE
    a) MT-45 (DESIGNER OPIOID): An observational case series was conducted, involving 9 patients identified with a psychoactive substance intoxication, later identified as MT-45, a designer opioid, as either the sole drug or in combination with other agents (eg, alcohol, phencyclidine, oxycodone, benzodiazepines). Paresthesia in the extremities was one of the reported symptoms (Helander et al, 2014).
    J) ANOXIC ENCEPHALOPATHY
    1) WITH POISONING/EXPOSURE
    a) Delayed postanoxic encephalopathy with cortical blindness was reported following a heroin overdose in a 43-year-old man. Two weeks following the overdose, a MRI revealed bilateral infarcts in the putamen, caudate and parietal and occipital lobes (McDonald et al, 1995) .
    b) Hypoxic encephalopathy was reported in a 17-year-old man who 4 days after hospital admission developed encephalopathy as a result of inhaling an unknown quantity of heroin (Zuckerman et al, 1996).
    c) An atypical course following acute heroin intoxication has included encephomyelo-polyradiculoneuropathy with diffuse impairment of the central and peripheral nervous system in one patient (Batora et al, 2002).
    K) MYELITIS
    1) WITH POISONING/EXPOSURE
    a) Acute transverse myelitis has been associated with intravenous or intranasal insufflation of heroin. A period of heroin abstinence followed by a single use of heroin has led to paralysis. Concurrent symptoms have included urinary retention, heroin-associated rhabdomyolysis, and onset of paraplegia after awakening from sleep. Normal CSF findings on routine analysis are reported in most cases, although pleocytosis and increased total protein may occur. MRI findings may, in some cases, be consistent with acute transverse myelitis. Following rehabilitation, patients often recover after several weeks (McCreary et al, 2000).
    L) SECONDARY PERIPHERAL NEUROPATHY
    1) WITH POISONING/EXPOSURE
    a) HEROIN
    1) CASE REPORT: A man injected the drug once into his right brachial plexus area presented in a coma with acute pulmonary edema, flaccid paralysis of the ipsilateral arm and leg, and severe edema of the arm. Severe brachial plexitis was present, which the authors speculated was a hypersensitivity response (Stamboulis et al, 1988).
    2) Rhabdomyolysis and neuropathy of a peripheral nerve or nerve plexus were reported in 7 patients after heroin abuse without evidence of muscle or nerve compression (deGans et al, 1985).
    3) CASE SERIES: In a case series of 136 subjects with chronic heroin abuse and a history of heroin overdose, 49% reported symptoms of peripheral neuropathy (numbness and tingling of hands or feet); in 16% these symptoms had lasted more than 2 hours (Warner-Smith et al, 2002). In this same group 26% reported having suffered paralysis of the limbs from extended pressure while unconscious (Warner-Smith et al, 2002).
    M) HYDROCEPHALUS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 25-year-old woman with a 2-year history of heroin inhalation was found comatose in her car. On presentation to the ED, she had a Glasgow Coma Scale score (GCS) of 6, a heart rate of 109 beats/min, and a blood pressure of 168/44 mmHg. Neuroimaging studies revealed hydrocephalus with enlarged ventricles, communicating ventriculomegaly, and cerebellar enhancement. All laboratory results, serial CT scans, and a second MRI were normal. Her cerebellar radiologic changes were improved after an external ventricular drain (EVD) was placed; however, she developed tremor and cogwheeling. She received acetazolamide 500 mg every 12 hours and dexamethasone to treat hydrocephalus. During multiple EVD clamping trials, her intracranial pressures increased and she became obtunded. On day 13 of hospitalization, she received a ventriculoperitoneal shunt (VPS) and she was discharged to a rehabilitation facility with a GCS of 15. On a 3-month follow-up, her symptoms had improved, but she sustained cognitive deficits with a low-pressure headache. Large improvement and increased pachymeningeal enhancement in postgadolinium T1 sequences were observed in a brain MRI (Bui et al, 2015).
    b) CASE REPORT: A 28-year-old man who became unconscious after using heroin inhalation, developed progressive parkinsonism over the next 4 weeks. He also complained about worsening headaches that started a few days before presentation. Physical examination showed sluggish pupils, downward ocular deviation, and intermittent tonic spasms. A brain MRI showed supratentorial symmetric white matter lesions typical of heroin-induced leukoencephalopathy, ventriculomegaly with sulcal effacement and tonsillar herniation. Brainstem compression from cerebellar swelling obliterated the 4th ventricle. He regained consciousness after the placement of ventriculostomy and treatment with dexamethasone; however, his conditioned worsened and despite further supportive care, including mannitol therapy, he died about 2 months after the onset of his illness (Dastur & Chang, 2015).
    N) PAIN
    1) WITH THERAPEUTIC USE
    a) High doses of opioids or combination opioid agonists/antagonists have been reported to cause a paradoxical pain reaction which resolves upon cessation of the drug.
    1) A trauma victim given high doses of nalbuphine (up to 300 mg/day) experienced nightmares and extreme pain until the doses were decreased (Reents, 1986).
    O) BOTULISM
    1) WITH POISONING/EXPOSURE
    a) Wound botulism, which has been associated with subcutaneous or intramuscular black tar heroin injections, has caused potentially lethal, descending, flaccid paralysis when Clostridium botulinum spores germinated in the wound which released neurotoxins (Passaro et al, 1998).
    P) PARAPLEGIA
    1) WITH POISONING/EXPOSURE
    a) A case of paraplegia below T12, compartment syndrome in one leg and eventual limb amputation was reported in a 29-year-old man with a known history of heroin abuse following a prolonged period of immobility (prolonged 'lotus' posture) as a result of acute heroin overdose (Kumar et al, 1999).
    3.7.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) NEUROPATHY
    a) Fentanyl, sufentanil, and alfentanil were administered as toxic doses to rats in a study of neurotoxicity. All three drugs produced histologic evidence of limbic system injury (Kofke et al, 1996).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) DRUG-INDUCED ILEUS
    1) WITH THERAPEUTIC USE
    a) DELAYED GASTRIC EMPTYING: Most members of the opioid group tend to delay gastric emptying so that reversal with an opioid antagonist may result in a return of peristalsis and further absorption of drug which results in prolonged coma (Jaffe & Martin, 1990).
    B) NAUSEA
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A 52-year-old woman developed only nausea and mild drowsiness after inadvertently receiving 3 mL (3 mg) of diamorphine intrathecally instead of 0.5% bupivacaine for spinal anesthesia for a total knee replacement surgery. She recovered after 48 hours of observation (Jafar & Chowdhry, 2014).
    C) CONSTIPATION
    1) WITH THERAPEUTIC USE
    a) Cramping and constipation may occur (Prod Info Infumorph(R), 1993; Prod Info Duragesic(R), 2001).
    D) SERUM AMYLASE RAISED
    1) WITH POISONING/EXPOSURE
    a) Hyperamylasemia has been reported (Jaffe & Martin, 1990). Theoretically, pancreatitis is a possible adverse effect of opioid overdoses.

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) CRUSH SYNDROME
    1) WITH POISONING/EXPOSURE
    a) Acute tubular necrosis secondary to rhabdomyolysis and myoglobinuria have been associated with impure heroin and the complications of seizures associated with opioid overdose (Nicholls et al, 1982; Krige et al, 1983; Chan et al, 1990; Rice et al, 2000; McCann et al, 2002; Kumar et al, 1999).
    B) GLOMERULONEPHRITIS
    1) WITH POISONING/EXPOSURE
    a) Focal glomerulosclerosis, membranous glomerulonephritis (associated with chronic hepatitis B), and immune complex proliferative glomerulonephritis (associated with endocarditis) have been described with heroin abuse (Cunningham et al, 1983; Dubrow et al, 1985).
    C) RENAL FAILURE SYNDROME
    1) WITH POISONING/EXPOSURE
    a) Heroin-associated nephropathy is a poorly explained cause of end stage renal disease (Cunningham et al, 1983; doSameiroFaria et al, 2003).
    D) AMYLOIDOSIS
    1) WITH POISONING/EXPOSURE
    a) Renal amyloidosis is associated with long term parenteral heroin abuse (Dubrow et al, 1985; Neugarten et al, 1986).
    E) PRIAPISM
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: A case of priapism, present for 12 hr prior to ED admission, was reported in a 41-year-old man which began 4 hours following a dose of IV heroin. Similar episodes had occurred in the past, all following heroin use. When medical therapy failed, both corpora cavernosa were drained and irrigated with lidocaine-epinephrine solution. Four days later the patient had another episode of heroin-induced priapism requiring surgical intervention (Nocchi & D'az, 1999).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) LEUKOCYTOSIS
    1) WITH POISONING/EXPOSURE
    a) Leukocytosis was reported in 90% of 18 hospital admissions in Taiwan due to heroin overdoses (Chan et al, 1995).
    b) Extreme leukocytosis (31.0 x 10(9)/L) without leftward shift in a 17-year-old girl was reported after an intravenous heroin overdose (Remskar et al, 1998).
    B) DISSEMINATED INTRAVASCULAR COAGULATION
    1) WITH POISONING/EXPOSURE
    a) Following a mixed intravenous heroin/cocaine overdose, an adult developed disseminated intravascular coagulation and acute renal failure and hyperkalemia subsequent to rhabdomyolysis . The patient died 2 months later with multiorgan failure (McCann et al, 2002).

Dermatologic

    3.14.2) CLINICAL EFFECTS
    A) ERUPTION
    1) WITH THERAPEUTIC USE
    a) Pruritus is a common adverse event following the administration of opiates, particularly morphine sulfate (Larijani et al, 1996).
    2) WITH POISONING/EXPOSURE
    a) CASE REPORT/INTRATHECAL: A 33-year-old woman in active labor was inadvertently given 45 mcg of sufentanil (a 9-fold increase; intended does 5 mcg) into the intrathecal space and complained of pruritus within 2 minutes of receiving the dose along with some difficulty swallowing (Coleman et al, 2009). The infant was delivered within 12 minutes of the event and had apgar scores of 8 and 9 at 1 and 5 minutes. The woman received naloxone shortly after delivery for complaints of severe pruritus, which the authors suggested may have limited the development of other adverse events. The patient and infant were discharged the following day.
    B) PETECHIAE OF SKIN
    1) WITH POISONING/EXPOSURE
    a) BLACK TAR HEROIN ADULTERATED WITH CINNAMON: A 38-year-old heroin user (500 mg IV twice daily) developed a painful and petechial rash on the palmar aspect of the left forearm soon after injecting (IV) black tar heroin adulterated with cinnamon. She did not experience any euphoria or opioid-like symptoms; however, she developed mild heroin withdrawal symptoms, including myalgia, palpitation, and anxiety. On presentation, she was mildly tachycardic (101 beats/min) and hypertensive (BP 142/86 mmHg). Following supportive care, including warm compresses and NSAIDs, her symptoms resolved (Hendrickson, 2015).
    C) SYSTEMIC SCLEROSIS
    1) WITH POISONING/EXPOSURE
    a) CASE REPORT: Diffuse cutaneous scleroderma of the hands, arms and face was reported in a 38-year-old man 3 years after successful detoxification from a 10 year history of IV heroin abuse. Subsequently, multiple telangiectases and painful ulcers of the fingers appeared. Four years earlier, while still using IV heroin, he had begun experiencing Raynaud's phenomenon (Biasi et al, 1998).
    D) PERIPHERAL ISCHEMIA
    1) WITH POISONING/EXPOSURE
    a) DESOMORPHINE
    1) Severe tissue damage, phlebitis, gangrene, and limb ischemia leading to amputation, and death have been reported following IV or IM injections of "Crocodile" (also known as Krokodil or Russian heroin), a Russian slang term for homemade desomorphine, an opioid analogon with analgesic, muscle-relaxing, sedating, and euphoric properties. Users of the drug may have a characteristic smell of iodine that is used during the production of the drug. Crocodile is manufactured from codeine, iodine, and red phosphorus, and may be contaminated with various toxic and corrosive substances. It purportedly is 8 to 15 times more potent than morphine with weaker convulsant, emetic and respiratory depressant effects (Gahr et al, 2012; Skowronek et al, 2012; Gahr et al, 2012a).
    E) EXTRAVASATION INJURY
    1) WITH POISONING/EXPOSURE
    a) HEROIN: CASE REPORT: A 57-year-old man developed extravasation injury following heroin injections, and presented with a skin lesion on the posterior region of the left forearm with extensive necrosis of skin and subcutaneous layer, involving the underlying muscle planes. The lesion had a sanious, fibrinous, secreting and smelly bottom. He received ciprofloxacin for 2 weeks after the lesion culture was positive for Pseudomonas aeruginosa. He was treated with daily dressing before the chemical debridement of the lesion using a topical ointment containing a collagenase plus hyaluronic acid (Bionect Start(R)). Each dressing contained 4 phases: disinfection with sodium hypochlorite 0.05% and povidone-iodine solution 10%, cleansing with saline solution, the application of a layer of 2 mm Binect Start(R) and covering with a pre-medicated patch. His symptoms gradually improved and a complete wound healing was observed 3 months after presentation (Onesti et al, 2014).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) RHABDOMYOLYSIS
    1) WITH POISONING/EXPOSURE
    a) Rhabdomyolysis, myoglobinuria, and subsequent acute tubular necrosis (ATN) have been associated with impure heroin injections, prolonged coma, or as a complication of seizures associated with opioid overdose (Nicholls et al, 1982; Krige et al, 1983; Sporer, 1999; Rice et al, 2000). In a mixed overdose of cocaine and heroin, rhabdomyolysis and hyperkalemia led to cardiac arrest (McCann et al, 2002).
    b) INCIDENCE: A 22% (4 of 18 patients) incidence of rhabdomyolysis occurred in heroin overdoses in one case series (Chan et al, 1995a).
    c) CASE REPORTS
    1) Intravenous heroin abuse led to rhabdomyolysis in 7 cases. These patients had CPK levels of 7 to 100 thousand units/liter (De Gans et al, 1985).
    2) An adult man died 3 days after heroin injection following the development of rhabdomyolysis and myoglobinuria (Chan et al, 1990).
    3) A 47-year-old man presented to the ED with a one day history of severe myalgia, vomiting, confusion and hallucinations. Lab values included CPK, 62,000 IU/L; LDH, 3,530 IU/L; ALT, 3,660 IU/L; and AST, 775 IU/L. A diagnosis of rhabdomyolysis, most likely due to opiates and benzodiazepines, was made. Acute renal failure, secondary to rhabdomyolysis also occurred (Larbi, 1997).
    4) A 26-year-old man was admitted to the ED with prolonged hypoxic coma with rhabdomyolysis and myocardial damage following acute intranasal heroin use 15 hours earlier. A serum CPK of 35,286 U/L was reported. The patient recovered following supportive care (Melandri et al, 1996a).
    5) A 29-year-old man developed rhabdomyolysis (serum myoglobin >400,000 mcg/L and serum creatine kinase (CK) of 122,000 IU) and subsequent acute renal failure following an overdose of heroin and cocaine. The patient recovered following symptomatic care (Kumar et al, 1999).
    B) COMPARTMENT SYNDROME
    1) WITH POISONING/EXPOSURE
    a) Compartment syndrome may occur in a comatose patient following abuse of narcotic injections, such as heroin (Franc-Law et al, 2000; Sporer, 1999; Kumar et al, 1999).
    b) CASE REPORT: A mixed intravenous overdose of cocaine and heroin has resulted in lower limb compartment syndrome requiring fasciotomy (McCann et al, 2002).
    c) CASE REPORT: A case of bilateral tibial compartment syndrome, severe hyperkalemia (9.2 mmol/L) and rhabdomyolysis (CPK 226,311 U/L), after abusing an unknown amount of opiates, benzodiazepines and cannabinoids with death resulting, was reported in a 35-year-old man(Ochoa-Gomez et al, 2002).
    d) CASE SERIES: The most frequent signs/symptoms in 4 cases of opiate (heroin) induced acute atraumatic compartment syndrome (AACS) were edema, pain, tension, and skin changes. The implicated mechanism of opiate-induced AACS was limb compression due to prolonged comatose state (Crush Syndrome). Prolonged limb compression tended to progress to complications such as cardiac dysrhythmias, hypotension, azotemia and renal failure. Long-term sequelae of motor loss, sensory disruption and development of contracture were reported (Franc-Law et al, 2000).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) HYPOGLYCEMIA
    1) WITH POISONING/EXPOSURE
    a) Hypoglycemia has been reported in a young adult man following heroin and amphetamine abuse. The patient presented in a coma and was restored to consciousness with a dextrose infusion. After losing consciousness again, he responded to naloxone (Kao et al, 1994).
    B) DISORDER OF ENDOCRINE SYSTEM
    1) Therapeutic and toxic doses of opioids may partially inhibit the release of cortisol in response to ACTH (Pullan et al, 1983; McDonald et al, 1959).
    2) CALCITONIN: Basal levels of immunoreactive calcitonin-like material were significantly higher in heroin addicts than in controls in one study (Tagliaro et al, 1985). Withdrawal of heroin decreased the levels in all subjects.

Reproductive

    3.20.1) SUMMARY
    A) Most opioids and opioid antagonists are classified as FDA pregnancy category B or C. Fetal physical dependence, withdrawal symptoms and respiratory difficulties may occur in infants born to mothers physically dependent on opioids. Neonatal withdrawal may be seen in the infants of addicted mothers 12 to 72 hours after birth. Infants may be dehydrated, irritable, and experience tremors and cry continually and may have diarrhea. In addition, there have been reports of severe fetal bradycardia, including life-threatening cases, following maternal administration of nalbuphine during pregnancy and/or labor and delivery.
    3.20.2) TERATOGENICITY
    A) ANIMAL STUDIES
    1) ELUXADOLINE
    a) No teratogenic effects or adverse effects on embryofetal development were observed in animals at doses up to 115 times the human exposure (Prod Info VIBERZI oral tablets, 2015).
    2) NALBUPHINE
    a) There was no evidence of teratogenicity or fetal harm when rats were given subQ nalbuphine at doses up to 100 mg/kg/day (approximately 6 times the maximum recommended human dose (MRHD)) and when rabbits were given IV nalbuphine at doses up to 32 mg/kg/day (approximately 4 times the MRHD) (Prod Info nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution, 2007).
    3) REMIFENTANIL
    a) In animal studies, there were no teratogenic effects observed when pregnant rats (5 mg/kg) and rabbits (0.8 mg/kg) were given remifentanil at doses up to 400 times and 125 times, respectively, the maximum recommended human dose on a mg/m(2) basis. Rats administered remifentanil throughout late gestation and lactation at IV doses up to 5 mg/kg (400 times the maximum recommended human dose on a mg/m2 basis) showed no significant effect (survival, development, or reproductive performance) on the F1 generation (Prod Info ULTIVA(R) IV injection, 2007).
    3.20.3) EFFECTS IN PREGNANCY
    A) RISK SUMMARY
    1) ELUXADOLINE: Exercise caution when administering to a pregnant woman (Prod Info VIBERZI oral tablets, 2015)
    B) FETAL BRADYCARDIA
    1) NALBUPHINE: Severe fetal bradycardia has occurred with nalbuphine administration during labor, but not earlier in pregnancy. However, the possibility of fetal bradycardia earlier in pregnancy cannot be ruled out. Following maternal administration of nalbuphine during labor, fetal and neonatal adverse events, some of which were life-threatening, have been reported. These effects included fetal bradycardia (some cases severe and prolonged). In some cases, these adverse events have resolved normal when the mother was administered naloxone during labor. There have been cases of permanent neurological damage associated with fetal bradycardia (Prod Info nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution, 2007). The use of nalbuphine during labor was also reported to cause fetal bradycardia, both after intravenous and intramuscular administration (Van Nesselrooij et al, 1992).
    C) PLACENTAL BARRIER
    1) NALBUPHINE: Transfers across the placental barrier at a rate that is high, rapid, and variable. The maternal to fetal ratio ranges from 1:0.37 to 1:6 (Prod Info nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution, 2007).
    D) RESPIRATORY DEPRESSION
    1) NALBUPHINE: Following maternal administration of nalbuphine during labor, fetal and neonatal adverse events, some of which were life-threatening, have been reported. These effects included respiratory depression at birth, apnea, cyanosis, and hypotonia. In some cases, these adverse events have resolved when the mother was administered naloxone during labor (Prod Info nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution, 2007). Transient neonatal respiratory depression was reported among infants born to women treated with nalbuphine during labor (Sgro et al, 1990; Guillonneau et al, 1990; Dan et al, 1991).
    E) OTHER FETAL/NEONATAL EFFECTS
    1) NALBUPHINE: Following maternal administration of nalbuphine during labor, fetal and neonatal adverse events, some of which were life-threatening, have been reported. These effects included apnea, cyanosis, and hypotonia. In some cases, these adverse events have resolved when the mother was administered naloxone during labor. Nalbuphine administration has also been associated with a sinusoidal fetal heart rate pattern (Prod Info nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution, 2007).
    F) WITHDRAWAL SYMPTOMS
    1) Fetal physical dependence, withdrawal symptoms and respiratory difficulties may occur in infants born to mothers physically dependent to opioids (Prod Info EXALGO(R) extended release oral tablets, 2010; Prod Info EMBEDA(R) oral extended-release capsules, 2009; Prod Info tapentadol immediate release oral tablets, 2008).
    2) Withdrawal symptoms have been observed in neonates whose mothers were taking narcotic analgesics during pregnancy. Symptoms of withdrawal may include irritability, hyperreflexia, hyperactivity, tremors, abnormal crying, diaphoresis, fever, vomiting, sneezing, yawning, and tachypnea (Blinick et al, 1973).
    3) Complications occur more frequently in infants with neonatal abstinence syndrome (NAS), a postnatal opioid withdrawal syndrome, than in other hospital births (Patrick et al, 2015).
    a) In a retrospective serial, cross-sectional analysis that estimated 21,732 infants with NAS and 3,716,916 other hospital births in 2012, reported the following complications in NAS patients compared to other hospital births: low birthweight (24.4% vs 7.2%), transient tachypnea of the newborn (11.7% vs 3.1%), meconium aspiration syndrome (2.8% vs 0.4%), respiratory distress syndrome (4.5% vs 2%), jaundice (32.8% vs 19.1%), feeding difficulty (17.3% vs 3%), seizures (1.4% vs 0.1%) and possible sepsis (14.8% vs 2.2%; p less than 0.001) (Patrick et al, 2015).
    G) PREGNANCY CATEGORY
    1) The following opioids have been classified as FDA pregnancy category D:
    1) NALORPHINE (Briggs et al, 1998)
    2) The following opioids have been classified as FDA pregnancy category C:
    1) ALFENTANIL HYDROCHLORIDE* (Prod Info ALFENTA(R) IV injection, 2008)
    2) ALPHAPRODINE* (Briggs et al, 1998)
    3) ETHOHEPTAZINE* (Briggs et al, 1998)
    4) OPIUM (Prod Info opium tincture (DEODORIZED) oral solution, 2008)
    5) OPIUM ALKALOIDS/BELLADONNA EXTRACT (Prod Info Belladonna & Opium rectal suppositories, 2005)
    6) PAREGORIC SYRUP (Prod Info paregoric oral solution, 1996)
    7) PENTAZOCINE* (Briggs et al, 1998)
    a) *Risk Factor D if used for prolonged periods or in high doses at term (Briggs et al, 1998).
    3) The following opioids have been classified as FDA pregnancy category B:
    1) ANILERIDINE* (Briggs et al, 1998)
    2) HEROIN* (Briggs et al, 1998)
    3) NALBUPHINE (Prod Info nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution, 2007)
    a) *Risk Factor D if used for prolonged periods or in high doses at term (Briggs et al, 1998).
    H) PLACENTAL BARRIER
    1) If delivery occurs quickly following an opioid dose to the mother, or after adequate time has passed to allow for maternal clearance, it is unlikely that the fetus would be affected. When maternal-fetal pH difference is minimal, fetal drug can cross back to the maternal side and be eliminated. Fetuses demonstrating significant distress and acidosis and whose mothers received opioids 1 to 3 hours prior to delivery, or multiple doses, may be at increased risk for respiratory depression, which would most likely be multifactorial in origin (Herschel et al, 2000).
    2) A heroin overdose in week 38 of pregnancy has been reported. The patient was admitted 4 hours after taking heroin (unknown amount) with hypothermia, grand mal seizures, and followed by coma (Glasgow 8). A C-section was performed. The newborn required ventilation. The mother developed SIADH, acute renal failure and acute pancreatitis secondary to heroin overdose. She recovered following symptomatic therapy (Cooley et al, 2002).
    I) ANIMAL STUDIES
    1) ELUXADOLINE
    a) No adverse effects on prenatal and postnatal development were reported in the offspring of animals at doses approximately 10 times the human exposure (Prod Info VIBERZI oral tablets, 2015).
    2) NALBUPHINE
    a) There were decreases in neonatal body weight and survival rates at birth and during lactation when subQ nalbuphine was given at doses approximately 4 times the maximum recommended human dose to female and male rats prior to mating and throughout gestation and lactation or to pregnant rats during the last third of gestation and throughout lactation (Prod Info nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution, 2007).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) BREAST MILK
    1) No reports describing the use of remifentanil during human lactation or measuring the amount, if any, of the drug excreted into milk have been located (Prod Info ULTIVA(R) IV injection, 2007).
    2) The American Academy of Pediatrics considers codeine, meperidine, methadone, and morphine compatible with breastfeeding (Anon, 2001).
    3) Heroin crosses into breast milk in sufficient quantities to cause addiction in the infant. The American Academy of Pediatrics classifies heroin abuse as a contraindication to breast feeding (Anon, 2001).
    4) ELUXADOLINE: No human lactation data are available; however, eluxadoline is present in rat milk. Exercise caution when administering to a nursing woman and weigh the benefits of breastfeeding with the risk to the infant (Prod Info VIBERZI oral tablets, 2015).
    5) NALBUPHINE: Limited data have shown that nalbuphine is excreted in the breast milk of humans in small concentrations (less than 1% of the administered dose) which is unlikely to have a significant effect on the infant (Prod Info nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution, 2007).
    B) ANIMAL STUDIES
    1) ELUXADOLINE
    a) Eluxadoline was detected in the milk of lactating animals after administration of doses 1.8, 3, and 10 times the human exposure (Prod Info VIBERZI oral tablets, 2015).
    3.20.5) FERTILITY
    A) ANIMAL STUDIES
    1) ELUXADOLINE: There were no reports of adverse effects on fertility or reproductive performance in animals administered eluxadoline at approximately 10 times the human exposure (Prod Info VIBERZI oral tablets, 2015).
    2) NALBUPHINE: There was no effect on male or female fertility when male and female rats were given subQ nalbuphine doses up to 56 mg/kg/day (Prod Info nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution, 2007).
    3) REMIFENTANIL: Male rats showed reduced fertility following daily intravenous remifentanil doses of 0.5 mg/kg (40 times the maximum recommended human dose on a mg/m(2) basis) after 70 days. Female rats showed no fertility issues at IV doses as great as 1 mg/kg (administered at least 15 days before mating) (Prod Info ULTIVA(R) IV injection, 2007).

Carcinogenicity

    3.21.2) SUMMARY/HUMAN
    A) Bladder carcinomas have been reported in a higher proportion of opium smokers than in cigarette smokers or non-smokers. A case-control study conducted in Iran of 309 cases of gastric adenocarcinoma and 613 matched controls found that opium use was associated with an increased risk of gastric adenocarcinoma.
    3.21.3) HUMAN STUDIES
    A) BLADDER CARCINOMA
    1) A high incidence of bladder carcinomas was observed in a population of opium smoking addicts, as opposed to cigarette smokers only or non-smokers in Iran. The authors conclude that the pyrolysis derived fractions of opium smoking may be potential bladder carcinogens in humans (Sadeghi et al, 1979).
    B) GASTRIC ADENOCARCINOMA
    1) A case-control study conducted in Iran of 309 cases of gastric adenocarcinoma and 613 matched controls found that opium use was associated with an increased risk of gastric adenocarcinoma (adjusted odds ratio [OR], 3.1; 95% CI, 1.9 to 5.1). Odds ratios were adjusted for age, ethnicity, education, socioeconomic status, total daily fruit intake, and total daily intake of vegetables. In addition, opium exposure was adjusted for the other main exposures in the study (ie, hookah, nass, and cigarettes). This increased risk was apparent for both anatomic subsites of gastric adenocarcinomas, cardia (adjusted OR, 2.8; 95% CI, 1.4 to 5.7) and noncardia (adjusted OR, 3.9; 95% CI, 1.6 to 9.4). The effect was dose-related, with individuals who had the highest cumulative opium use (greater than the median of 29 unit-years) having the highest risk for gastric adenocarcinoma (adjusted OR, 4.5; 95% CI, 2.3 to 8.5). People in this study who used any of the tobacco products (hookah, nass, and cigarettes) did not have a statistically significant risk of gastric adenocarcinoma (overall or by anatomic subsite). Further studies are needed to confirm this study's findings (Shakeri et al, 2013).
    3.21.4) ANIMAL STUDIES
    A) LACK OF EFFECT
    1) ELUXADOLINE: No tumorigenic effects were observed in mice and rats administered eluxadoline at doses up to 14 and 36 times the human exposure, respectively (Prod Info VIBERZI oral tablets, 2015).

Summary Of Exposure

    A) USES: Opioids are primarily used for the treatment of pain, less often for cough suppression. Opioids are commonly abused for euphoric effects by multiple routes (ie, injection, insufflation, smoking, ingestion and transdermal). The following agents and opioid withdrawal have their own specific managements, please refer to them as indicated: buprenorphine, butorphanol, codeine, dihydrocodeine, fentanyl, hydrocodone, hydromorphone, levorphanol, long-acting opioid antagonists (nalmefene, naltrexone, nalmexone), meperidine, methadone, methadone, oxycodone, pentazocine, propoxyphene, tramadol, and naloxegol.
    B) PHARMACOLOGY: Opioids are a group of chemical substances, naturally occurring and synthetic, that bind at the opiate receptor. Opiates are a group of naturally occurring compounds derived from the poppy, Papaver somniferum.
    C) TOXICOLOGY: Therapeutic and toxic effects are mediated by different opioid receptors. Mu 1: Supraspinal and peripheral analgesia, sedation and euphoria. Mu 2: Spinal analgesia, respiratory depression, physical dependence, GI dysmotility, bradycardia and pruritus. Kappa 1: Spinal analgesia and miosis. Kappa 2: Dysphoria and psychotomimesis. Kappa 3: Supraspinal analgesia. Chronic opioid users develop tolerance to the analgesic and euphoric effects, but not to the respiratory depression effects.
    D) EPIDEMIOLOGY: Overdose is common, particularly in patients with chronic opioid abuse, and often life threatening (especially a heroin or methadone overdose).
    E) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE POISONING: Euphoria, drowsiness, constipation, nausea, vomiting and miosis. Mild bradycardia or hypotension may be present.
    2) SEVERE POISONING: Respiratory depression leading to apnea, hypoxia, coma, bradycardia, or acute lung injury. Rarely, seizures may develop from hypoxia. Acute tubular necrosis secondary to rhabdomyolysis and myoglobinuria may develop in patients with prolonged coma or seizures. Death may result from any of these complications. Scleroderma following heroin abuse has been reported and may be linked to talc mixed with heroin. Wound botulism resulting in flaccid paralysis may occur following black tar heroin abuse. Hypoglycemia and leukocytosis have been reported in heroin abusers. Intraarterial injection can cause ischemia.
    3) DESOMORPHINE: Severe tissue damage, phlebitis, gangrene, and limb ischemia leading to amputation, and death have been reported following IV or IM injections of "Crocodile" (also known as Krokodil or Russian heroin), a Russian slang term for homemade desomorphine.

Genotoxicity

    A) ELUXADOLINE: There was no evidence of genotoxicity or mutagenicity in the following tests: Ames test, chromosome aberration test in human lymphocytes, mouse lymphoma cell forward mutation test, and in vivo rat bone marrow micronucleus test (Prod Info VIBERZI oral tablets, 2015).
    B) OPIUM CHEWING/TOBACCO: Prolysed opium, from Iran and Transkei, showed mutagenic activity in the Salmonella typhimurium assay in rat liver microsomes (Hewer et al, 1978).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor vital signs frequently, pulse oximetry, and continuous cardiac monitoring.
    B) Monitor for CNS and respiratory depression.
    C) Opioid plasma levels are not clinically useful or readily available. Urine toxicology screens may confirm exposure, but are rarely useful in guiding therapy; urine toxicology immunoassays may also miss synthetic opioids (eg, fentanyl and methadone).
    D) Obtain acetaminophen and salicylate levels in patients with a suspected overdose that may include combination products.
    E) Routine lab work is usually not indicated, unless it is helpful to rule out other causes or if the diagnosis of opioid toxicity is uncertain.
    F) Obtain a chest x-ray for persistent hypoxia. Consider a head CT and/or lumbar puncture to rule out an intracranial mass, bleeding or infection, if the diagnosis is uncertain.
    4.1.2) SERUM/BLOOD
    A) BLOOD/SERUM CHEMISTRY
    1) CPK with enzyme fractionation may be useful in severe opioid poisoning cases or when the patient experiences chest pain, seizure or coma. Severely poisoned patients (seizures, persistent mental status changes, hypotension, ventricular dysrhythmias) should have monitoring of electrolytes, BUN, and creatinine and cardiac markers.
    B) LABORATORY INTERFERENCE
    1) POPPY SEEDS - Several studies have shown that ingestion of poppy seeds or poppy seed containing bakery goods may yield measurable urine levels of both codeine and morphine up to 22 hours post-ingestion (Zebelman et al, 1987; Struempler, 1987; Selavka, 1991; Abelson, 1991).
    a) After ingestion of poppy seeds, opioids show up in the urine within 5 hours (Beck et al, 1990).
    b) Soaking the seeds in water removed 45.6% of free morphine and 48.4% of free codeine (Lo & Chua, 1992). Soaked seeds have less potential for producing elevated urine levels.
    4.1.3) URINE
    A) URINARY LEVELS
    1) CODEINE/MORPHINE RATIO - In an attempt to decide whether urinary opioid levels are due to codeine, morphine, or heroin, urine tests may try to differentiate between the two. Cone et al (1991), using volunteers, found that the urinary codeine/morphine ratio was not a reliable indicator of the type of opioid that had been taken (Cone et al, 1991).
    a) The Department of Defense (DOD) and Health and Human Services (HHS) have set an opiate urine screening cutoff concentration of 2000 ng/mL and a confirmation cutoff concentration for total morphine of either 2000 ng/mL (HHS) or 4000 ng/mL (DOD). A specific heroin marker, 6-acetylmorphine (6-AM), was added to the testing if a urine tested positive for morphine. A cutoff for 6-AM is 10 ng/mL. A test coming back above the cutoffs for both morphine and heroin are considered positive for heroin. A test above the cutoff for morphine but not for heroin is considered positive for morphine (Moore et al, 2001).
    b) A urinary codeine concentration (following multiple doses of anti-cough syrup) larger than 300 ng/mL and the ratio of morphine to codeine to be less than 3 has been proposed as criteria for a positive codeine intake (as opposed to morphine) (Hsu et al, 2000).
    2) A urine screening cutoff of 50 ng/mL instead of the usual 300 ng/mL for suspected opiate intoxications, with follow-up blood screens was suggested. In a case series, it was reported that 23 of 67 cases of narcotic overdoses resulting in death had urine opiate concentrations less than 300 ng/mL (Levine & Smialek, 1998).
    B) URINALYSIS
    1) Monitor for the presence of urinary myoglobin in all cases of suspected or potential rhabdomyolysis.
    C) LABORATORY INTERFERENCE
    1) Quinolones may cause false-positive results for opiate urine screens (Baden et al, 2001).
    4.1.4) OTHER
    A) OTHER
    1) HAIR
    a) Past opiate usage can be detected by means of hair analysis as well as the diagnosis of poisoning associated with opiate addiction. Hair analysis may be accomplished by means of RIA, HPLC, or GC/MS. Hair analysis may provide both hair drug content and hair profile. A single opiate ingestion may not be detectable through hair analysis, which may be a drawback of this method (Staub, 1993). GC/MS method has been used for analyzing cocaine and its metabolites, opiates, and cannabinoids in human hair (Jurado et al, 1995).
    1) HEROIN - RIA method may be used for hair analysis of cocaine and heroin (Bermejo-Barrera & Strano-Rossi, 1995). A cut-off of 0.5 ng/mg of 6-monoacetylmorphine (6-MAM), a heroin metabolite, is recommended for hair analysis to establish heroin abuse using a GC/MS method (Kintz & Mangin, 1995). A mean morphine content of 1.15 ng/mg (range, 0-12.25 ng/mg) was found in the hair of heroin addicts who had died. A mean morphine content of 6.07 ng/mg was found in the hair of active surviving heroin addicts (Tagliaro et al, 1998).
    2) SALIVA
    a) On site saliva drug testing (Cozart RapiScan), which detects the parent drug for opiates, is based on the principle of competitive lateral flow immunoassay using digital photography to provide a semi-quantitative end point. Sensitivity and specificity were both 100% for codeine for 9 hours after ingestion. Tests are positive as long as codeine saliva concentrations are above 5 ng/mL by GC/MS (Jehanli et al, 2001).

Radiographic Studies

    A) RADIOGRAPHIC-OTHER
    1) Chest x-ray is recommended in patients with pulmonary symptoms.
    2) Abdominal x-rays may be of value in locating opioid packets in opioid smuggler "body-packers".
    3) Abdominal flat plate x-ray should be performed to establish diagnosis and location. Not all package types can be visualized on x-ray (McCarron & Wood, 1983) or may disappear from view following treatment with activated charcoal (Harchelroad, 1992).
    a) Upper abdominal CT scan and abdominal x-ray with Gastrografin(R) have been reported to result in better visualization (Diamant-Berger et al, 1988; Marc et al, 1990; Hartoko et al, 1988).
    4) Magnetic resonance does not visualize packets because of the lack of protons (Kersschot et al, 1985).
    5) X-rays must be repeated after each procedure until all packages have been removed.
    6) In 5 volunteers, non-contrast spiral CT scans were performed 2 to 4 hours after ingestion of a placebo wrapped in plastic bags, to determine the effectiveness of the CT scans in visualizing the placebo. Foreign bodies, consistent with placebo, were seen in 3 out of 5 CT scans (60%). No definite foreign body was visible in the other CT scans (Hibbard et al, 1999).
    7) CASE SERIES - In a series of 14 patients, 9 of the patients swallowed heroin-containing packets and 5 inserted the packets rectally. Thirteen had evidence of packets on KUB. Bisacodyl suppositories were used to evacuate packets from the rectum. No patient received ipecac or gastric lavage (Utecht et al, 1993).
    B) MRI
    1) Diffusion-weighted (DW) magnetic resonance (MR) imaging has been reported to show diffuse isotropic restriction of water diffusion in the myelin sheath at the intermediate stage of disease in heroin-induced spongiform leukoencephalopathy. In less severe heroin-induced leukoencephalopathy, reversible change of the DW MR signal may be expected (Chen et al, 2000).

Methods

    A) SAMPLING
    1) These drugs are qualitatively identified in the urine and are detected in blood for about 12 hours after therapeutic doses.
    a) In overdose, heroin may be detected for as long as 36 hours. Adulterants are often present and may be of limited interest.
    2) The laboratory must be notified if naloxone was administered prior to obtaining blood or urine for analysis so that a methodology will be used that will not falsely identify naloxone as the opioid.
    3) In assessing atypical symptoms consider designer agents such as fentanyl analogues. Send samples to an appropriate laboratory with necessary procedures to study these agents. These tests may not be available in routine laboratories. Use state or DEA laboratories if in doubt.
    a) Scopolamine was detected via gas chromatography-mass spectrometry in a sample of heroin and the urine of a patient presenting with an anticholinergic toxidrome following the use of heroin. It is not clear if this was an adulterant or contaminant of heroin. Atropine-like compounds should be suspected as co-intoxicants when anticholinergic toxidromes are seen (Perrone et al, 1999; Hamilton et al, 2000).
    B) IMMUNOASSAY
    1) Semiquantitative and qualitative EMIT(R) homogeneous enzyme immunoassays are available for measurement of the class of opioids in urine.
    a) The assays detect morphine, methadone, morphine glucuronide, codeine, and hydromorphone, and higher concentrations of nalorphine and meperidine.
    1) The detection limit (sensitivity) is 0.5 mcg/mL for morphine or its equivalent.
    2) The assays do not detect long-acting methadone, L-alpha-acetyl-methadol (LAAM), or its metabolites.
    3) CDC proficiency testing and clinical studies show this method to correlate well with GC, GLC, HPLC, RIA, and TLC.
    4) RELIABILITY
    a) There is a risk of false negatives for "designer" agents or exceedingly potent fentanyl varieties. Specific requests may be necessary. 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.
    b) In a study of 183 cases of fatal opioid overdose (primarily heroin), fluorescent polarization immunoassay (FPIA) detected opioids in 85% of the cases in which opioids were detected by gas chromatography/mass spectrophotometry analysis of blood, giving a false-negative rate of 14.75% was found using an immunoassay for urine sampling as compared with blood analysis postmortem (Molina & Dimaio, 2005).
    c) False positive EMIT II results for urine opiates from therapeutic dosing with ofloxacin have been reported. Results could not be confirmed by gas chromatography/mass spectrometry. Other quinolone antibiotics (norfloxacin, ciprofloxacin, nalidixic acid) did not show potential for false positive opiate results (Meatherall & Dai, 1997).
    1) Levofloxacin, ofloxacin and pefloxacin were shown to cause false positive opiate results with five different immunoassay testing techniques. Nine of 13 quinolones tested caused assay results above the threshold for a positive result (Baden et al, 2001).
    b) Quantifying 6-monoacetylmorphine in serum, a good indicator of heroin intake, is possible using GC/MS and RIA methods (Moeller & Mueller, 1995).
    c) BUTYRFENTANYL: Butyrfentanyl, a potent short-acting opioid and an analog of fentanyl, is only 0.13 the potency of fentanyl, but 7 times more potent than morphine. Cross-reactivity of fentanyl and butyrfentanyl on immunoassay is very high (Cole et al, 2015).
    1) CASE REPORT: An 18-year-old boy with a past medical history of IV heroin abuse, developed hemoptysis, acute lung injury, hypoxic respiratory failure, and diffuse alveolar hemorrhage, after snorting an unknown amount of butyrfentanyl instead of acetyl fentanyl. Fentanyl and opiates immunoassays were both positive, but fentanyl was not present in high-performance liquid chromatography and mass spectrometry. Butyrfentanyl was detected using a high-performance gas chromatography obtained from the Bureau of Criminal Apprehension. Following further supportive care, his condition improved gradually and he was extubated on day 4 and discharged 3 days later after receiving treatment for ventilator-associated pneumonia (Cole et al, 2015).
    C) CHROMATOGRAPHY
    1) Various metabolites were identified and quantified in blood from a fatal overdose involving a combination of ethylmorphine, hydrocodone, dihydrocodeine and codeine. GC/MS was used after silylation to detect these substances (Balikova & Maresova, 1998).
    2) AH-7921: Nine patients were found dead after using AH-7921, a designer opioid with about 80% of morphine's mu-agonist activity. Six patients also used other drugs, such as benzodiazepines, antidepressants, and analgesics; however, it was concluded that poly-drug use (all agents with postmortem therapeutic concentrations) was not a major contributing factor for the fatal outcome. A selective liquid chromatography-mass spectrometry (MS)-MS method detected AH-7921 concentrations, ranging from 0.03 to 0.99 mcg/g blood. Metabolites of the parent compound, found in the blood, included 2 N-demethylated that dominated and 4 mono-hydroxylated in trace amounts. Death occurred both at low and high concentrations of AH-7921, probably due to the differences in tolerance of the drug (Kronstrand et al, 2014).
    3) 6-ACETYLMORPHINE - A metabolite of heroin, can be detected in urine using a gas chromatography/electron impact mass fragmentometric determination (Paul et al, 1989).
    4) Quantifying 6-monoacetylmorphine in serum, a good indicator of heroin intake, is possible using GC/MS and RIA methods (Moeller & Mueller, 1995).
    5) PLASMA - A flame-ionization gas chromatographic method for alphaprodine in plasma ) had a detectability limit of 0.03 mg/L (Fung et al, 1980).
    6) REMIFENTANIL - Normal toxicological screening does not reveal the presence of remifentanil or its major metabolite. A very short elimination half-life (3 to 10 minutes) makes it almost impossible to detect remifentanil in serum. However, its metabolite, GR90291, has been detected in whole blood, liver, kidney and lung following a fatal overdose by means of a GC/MS-EI method (Asselborn et al, 2002).
    7) KETOBEMIDONE - A gas chromatographic/mass spectrometry method has been described for the detection and quantification of ketobemidone and its major metabolite, norketobemidone. A cut off of 0.25 mcg/mL was used for both ketobemidone and its metabolite (Angelo & Hausner, 2002).
    8) BUTYRFENTANYL: Butyrfentanyl, a potent short-acting opioid and an analog of fentanyl, is only 0.13 the potency of fentanyl, but 7 times more potent than morphine. Cross-reactivity of fentanyl and butyrfentanyl on immunoassay is very high (Cole et al, 2015).
    a) CASE REPORT: An 18-year-old boy with a past medical history of IV heroin abuse, developed hemoptysis, acute lung injury, hypoxic respiratory failure, and diffuse alveolar hemorrhage, after snorting an unknown amount of butyrfentanyl instead of acetyl fentanyl. Fentanyl and opiates immunoassays were both positive, but fentanyl was not present in high-performance liquid chromatography and mass spectrometry. Butyrfentanyl was detected using a high-performance gas chromatography obtained from the Bureau of Criminal Apprehension. Following further supportive care, his condition improved gradually and he was extubated on day 4 and discharged 3 days later after receiving treatment for ventilator-associated pneumonia (Cole et al, 2015).
    D) OTHER
    1) HAIR - Human hair analysis has been explored as a way to test the drug history of an individual from sampling time to several months previous (Cone, 1990).
    a) Hair is analyzed by radioimmunoassay, and specific substances can be quantitated using gas chromatography/mass spectrometry.
    b) GC/MS in SIM mode has been described for analyzing and quantifying heroin and cocaine metabolites in urine (Bermejo-Barrera & Strano-Rossi, 1995).
    c) In experimental studies on human heroin abusers, both morphine and codeine were easily detected in beard hair 7 to 8 days after drug administration, long after clinical signs and drug levels in urine, plasma, and saliva had abated (Cone, 1990).
    d) Drug levels in beard hair appear to be dose-related, and it can be assumed that hair grows at a rate of 1 cm/month (Cone, 1990). These data allow for an approximation of time and dose of drug exposure (Strang et al, 1990).
    e) Mean morphine hair content in addicts who died as a result of heroin was 1.15 ng/mg (SD 2.35 ng/mg; range 0 to 12.25 ng/mg), while active heroin addicts had mean levels of 6.07 ng/mg and abstinent former addicts had mean levels of 0.74 ng/mg (Tagliaro et al, 1998).
    f) Hair analysis has been used as evidence in courts of law (Strang et al, 1990; Brewer, 1990).
    2) FINGERNAILS - Fingernail clippings may be an alternative to hair sampling for the detection of past heroin use. In 25 out of 26 heroin users, positive RIA results were obtained with nail clippings (mean morphine concentration of 1.67 ng/mg). In 22 out of 26 heroin users, positive HPLC results were obtained (mean morphine concentration of 2.11 ng/mg) (Lemos et al, 2000).
    3) BONE/BONE MARROW - In a fatal heroin addiction case, post-mortem examination in bone and bone marrow was performed, which included immediately after death and one year after burial of the samples, via gas-liquid chromatographic methodology. Morphine concentrations of 195, 340 and 155 ng/g for bone marrow, bone and buried bone, respectively, were reported. No bone marrow remained after 1 year. Interpretation of morphine concentrations in bone and bone marrow is not yet possible (Raikos et al, 2001).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) Patients with significant, persistent central nervous system depression should be admitted to the hospital. A patient needing more than 2 doses of naloxone should be admitted as a longer-acting opioid has likely been taken; additional doses may be needed. Patients with coma, seizures, dysrhythmias, delirium, and those needing a naloxone infusion or who are intubated should be admitted to an intensive care setting.
    6.3.1.2) HOME CRITERIA/ORAL
    A) Respiratory depression may occur at doses just above the therapeutic dose. Children should be observed and evaluated in the hospital as they are generally opioid naive and may develop respiratory depression. Adults should be evaluated by a health care professional if they have received a higher than recommended (therapeutic) dose, especially if opioid naive.
    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) SUMMARY: Patients with deliberate ingestions or a pediatric ingestion should be sent to a health care facility for observation for at least 4 hours, to ensure that peak plasma levels have been reached and there has been sufficient time for symptoms to develop. Patients that have ingested a sustained release or long acting product have the potential to manifest symptoms in a delayed fashion and should be observed for 24 hours. Patients who are treated with naloxone should be observed for 4 to 6 hours after the last dose, for recurrent CNS depression or acute lung injury.
    B) Patients should be observed for return of respiratory depression and resedation after naloxone administration. The duration of action for naloxone is approximately 20 to 90 minutes, depending on the dose, route and the opioid agonist ingested (Howland, 2006).
    C) Patients with intravenous heroin overdose requiring naloxone reversal should be monitored for 4 hours after the last dose of naloxone to observe for evidence of pulmonary edema (Duberstein & Kaufman, 1971; Steinberg & Karlinger, 1968). Patients with oral opioid overdose should be monitored for 6 hours and admitted if signs or symptoms develop. Patients with overdose of long acting opioids, or controlled-release dosage forms, require admission as clinical effects may be delayed (Wilen et al, 1975; Sey et al, 1971; Geller & Garrettson, 1994; Westerling et al, 1998).
    D) In a retrospective review of 726 patients meeting criteria for opioid overdoses, all were treated by paramedics or in ED with naloxone and bag-valve-mask ventilation. Less than 5%, excepting those that expired first, required hospital admission. Patients were admitted for the following reasons: noncardiogenic pulmonary edema, pneumonia or other infections, persistent respiratory depression and persistent alteration in mental status. Those patients found in cardiopulmonary arrest were not observed to have long-term survival. The out-of-hospital treatment with naloxone and bag-valve-mask ventilation was all that the majority of patients required (Sporer et al, 1996).

Monitoring

    A) Monitor vital signs frequently, pulse oximetry, and continuous cardiac monitoring.
    B) Monitor for CNS and respiratory depression.
    C) Opioid plasma levels are not clinically useful or readily available. Urine toxicology screens may confirm exposure, but are rarely useful in guiding therapy; urine toxicology immunoassays may also miss synthetic opioids (eg, fentanyl and methadone).
    D) Obtain acetaminophen and salicylate levels in patients with a suspected overdose that may include combination products.
    E) Routine lab work is usually not indicated, unless it is helpful to rule out other causes or if the diagnosis of opioid toxicity is uncertain.
    F) Obtain a chest x-ray for persistent hypoxia. Consider a head CT and/or lumbar puncture to rule out an intracranial mass, bleeding or infection, if the diagnosis is uncertain.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) SUMMARY
    1) Opioid overdoses are life threatening. Activated charcoal should be considered early after a significant oral ingestion, if a patient can protect their airway and is without significant signs of toxicity. If a patient is displaying signs of moderate to severe toxicity, do NOT administer activated charcoal because of the risk of aspiration. Search for and remove any fentanyl patches on the patient's body.
    2) 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)
    3) 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).
    B) ACTIVATED CHARCOAL
    1) PREHOSPITAL ACTIVATED CHARCOAL ADMINISTRATION
    a) Consider prehospital administration of activated charcoal as an aqueous slurry in patients with a potentially toxic ingestion who are awake and able to protect their airway. Activated charcoal is most effective when administered within one hour of ingestion. Administration in the prehospital setting has the potential to significantly decrease the time from toxin ingestion to activated charcoal administration, although it has not been shown to affect outcome (Alaspaa et al, 2005; Thakore & Murphy, 2002; Spiller & Rogers, 2002).
    1) In patients who are at risk for the abrupt onset of seizures or mental status depression, activated charcoal should not be administered in the prehospital setting, due to the risk of aspiration in the event of spontaneous emesis.
    2) The addition of flavoring agents (cola drinks, chocolate milk, cherry syrup) to activated charcoal improves the palatability for children and may facilitate successful administration (Guenther Skokan et al, 2001; Dagnone et al, 2002).
    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).
    6.5.2) PREVENTION OF ABSORPTION
    A) SUMMARY
    1) Consider activated charcoal if a patient presents soon after an ingestion and is not manifesting signs and symptoms of toxicity. Activated charcoal is generally not recommended in patients with significant signs of toxicity because of the risk of aspiration. Gastric lavage is not recommended as patients usually do well with supportive care.
    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).
    3) VOLUNTEER STUDIES demonstrate that activated charcoal can decrease opioid absorption (Laine et al, 1997).
    C) BODY PACKERS/BODY STUFFERS
    1) Refer to "BODY PACKERS" and "BODY STUFFERS" managements for further information.
    6.5.3) TREATMENT
    A) MONITORING OF PATIENT
    1) Monitor vital signs frequently, pulse oximetry, and continuous cardiac monitoring.
    2) Monitor for CNS and respiratory depression.
    3) Opioid plasma levels are not clinically useful or readily available. Urine toxicology screens may confirm exposure, but are rarely useful in guiding therapy; urine toxicology immunoassays may also miss synthetic opioids (e.g., fentanyl and methadone).
    4) Obtain acetaminophen and salicylate concentrations if ingestion of a combination product is possible.
    5) Routine lab work is usually not indicated, unless it is helpful to rule out other causes or if the diagnosis of opioid toxicity is uncertain.
    6) Obtain a chest x-ray for persistent hypoxia. Consider a head CT and/or lumbar puncture to rule out an intracranial mass, bleeding or infection, if the diagnosis is uncertain.
    B) 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).
    C) NALMEFENE
    1) MANAGEMENT OF OPIOID OVERDOSE: Nalmefene is rarely used for acute opioid overdose. The initial dose for non-opioid dependent patients is 0.5 mg/70 kg. If needed, a second dose of 1.0 mg/70 kg may be given 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 be effective. The reversal effect of nalmefene varies. If a partial reversing dose is used (1 mcg/kg), reversal may only last 30 to 60 minutes due to redistribution of the drug or persistent opioid effect. At higher doses, which is usually the full reversal (1 mg/70kg) dose, the duration of action for nalmefene may last several hours. Repeated dosing may be indicated for a recurrence of respiratory depression; the dose should be titrated to response (Prod Info REVEX(R) injection, 2006).
    a) ALTERNATIVE ROUTES: If IV access is unavailable, the intramuscular or subcutaneous route may be used. However, the effect of a 1 mg dose may delayed by 5 to 15 minutes (Howland, 2006a).
    b) WITHDRAWAL SYNDROME: Using higher dosages or shorter intervals is likely to increase the risk of symptoms related to acute withdrawal (i.e., nausea, vomiting, elevated blood pressure, and anxiety) (Prod Info REVEX(R) injection, 2006).
    D) NALTREXONE
    1) OPIOID ADDICTION: Naltrexone is an effective narcotic antagonist in the management of opiate addiction/dependence, but is NOT used for opioid overdose. Studies have shown that 50 mg of naltrexone will block the effect of 25 mg of heroin for up to 24 hours. Data have also shown that doubling the dose of naltrexone will double the duration of action and tripling the dose will provide blockade for up to 72 hours (Prod Info VIVITROL(TM) extended-release injectable suspension, 2006).
    E) AIRWAY MANAGEMENT
    1) Administer oxygen and assist ventilation for respiratory depression. Orotracheal intubation for airway protection should be performed early in cases of obtundation and/or respiratory depression that do not respond to naloxone, or in patients who develop severe acute lung injury.
    2) REFRACTORY: A case of massive oxycodone controlled release ingestion refractory to naloxone (188 mg naloxone given over 14 hours) has been reported. In this case, naloxone was discontinued and the patient's trachea was intubated in order to maintain adequate ventilation. Mechanical ventilation was required for about 72 hours (Schneir et al, 2002).
    F) ACUTE LUNG INJURY
    1) ONSET: Onset of acute lung injury after toxic exposure may be delayed up to 24 to 72 hours after exposure in some cases.
    2) 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)
    3) 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).
    4) ANTIBIOTICS: Indicated only when there is evidence of infection (Artigas et al, 1998).
    5) EXPERIMENTAL THERAPY: Partial liquid ventilation has shown promise in preliminary studies (Kollef & Schuster, 1995).
    6) 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).
    7) 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).
    G) SEIZURE
    1) Seizures are rare, but may be a result of hypoxia or due to properties of certain agents (eg, meperidine, tramadol, propoxyphene). If CNS depression is present, administer naloxone and correct hypoxia. Administer intravenous, benzodiazepines, barbiturates or propofol if seizures persist.
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2009; Chin et al, 2008).
    5) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    6) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    H) HYPOTENSIVE EPISODE
    1) If CNS depression is present, administer naloxone and correct hypoxia. Hypotension should initially be treated with a saline bolus, if patient can tolerate a fluid load, then adrenergic vasopressors to raise mean arterial pressure.
    2) 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.
    3) 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).
    4) 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) RHABDOMYOLYSIS
    1) SUMMARY: Early aggressive fluid replacement is the mainstay of therapy and may help prevent renal insufficiency. Diuretics such as mannitol or furosemide may be added if necessary to maintain urine output but only after volume status has been restored as hypovolemia will increase renal tubular damage. Urinary alkalinization is NOT routinely recommended.
    2) Initial treatment should be directed towards controlling acute metabolic disturbances such as hyperkalemia, hyperthermia, and hypovolemia. Control seizures, agitation, and muscle contractions (Erdman & Dart, 2004).
    3) FLUID REPLACEMENT: Early and aggressive fluid replacement is the mainstay of therapy to prevent renal failure. Vigorous fluid replacement with 0.9% saline (10 to 15 mL/kg/hour) is necessary even if there is no evidence of dehydration. Several liters of fluid may be needed within the first 24 hours (Walter & Catenacci, 2008; Camp, 2009; Huerta-Alardin et al, 2005; Criddle, 2003; Polderman, 2004). Hypovolemia, increased insensible losses, and third spacing of fluid commonly increase fluid requirements. Strive to maintain a urine output of at least 1 to 2 mL/kg/hour (or greater than 150 to 300 mL/hour) (Walter & Catenacci, 2008; Camp, 2009; Erdman & Dart, 2004; Criddle, 2003). To maintain a urine output this high, 500 to 1000 mL of fluid per hour may be required (Criddle, 2003). Monitor fluid input and urine output, plus insensible losses. Monitor for evidence of fluid overload and compartment syndrome; monitor serum electrolytes, CK, and renal function tests.
    4) DIURETICS: Diuretics (eg, mannitol or furosemide) may be needed to ensure adequate urine output and to prevent acute renal failure when used in combination with aggressive fluid therapy. Loop diuretics increase tubular flow and decrease deposition of myoglobin. These agents should be used only after volume status has been restored, as hypovolemia will increase renal tubular damage. If the patient is maintaining adequate urine output, loop diuretics are not necessary (Vanholder et al, 2000).
    5) URINARY ALKALINIZATION: Alkalinization of the urine is not routinely recommended, as it has never been documented to reduce nephrotoxicity, and may cause complications such as hypocalcemia and hypokalemia (Walter & Catenacci, 2008; Huerta-Alardin et al, 2005; Brown et al, 2004; Polderman, 2004). Retrospective studies have failed to demonstrate any clinical benefit from the use of urinary alkalinization (Brown et al, 2004; Polderman, 2004; Homsi et al, 1997).
    6) Continuous venovenous hemodiafiltration (CVVH-D) and fasciotomy for patients with severe compartments syndrome complicated by rhabdomyolysis has been described. This aggressive method may improve renal function in the face of acute renal failure (Russo et al, 1995).
    J) SEROTONIN SYNDROME
    1) GENERAL: Serotonin syndrome may be precipitated by an acute mixed ingestion (eg, heroin).
    2) SUMMARY
    a) Benzodiazepines are the mainstay of therapy. Cyproheptadine, a 5-HT antagonist, is also commonly used. Severe cases have been managed with benzodiazepine sedation and neuromuscular paralysis with non-depolarizing agents(Claassen & Gelissen, 2005).
    3) HYPERTHERMIA
    a) Control agitation and muscle activity. Undress patient and enhance evaporative heat loss by keeping skin damp and using cooling fans.
    b) MUSCLE ACTIVITY: Benzodiazepines are the drug of choice to control agitation and muscle activity. DIAZEPAM: ADULT: 5 to 10 mg IV every 5 to 10 minutes as needed, monitor for respiratory depression and need for intubation. CHILD: 0.25 mg/kg IV every 5 to 10 minutes; monitor for respiratory depression and need for intubation.
    c) Non-depolarizing paralytics may be used in severe cases.
    4) CYPROHEPTADINE
    a) Cyproheptadine is a non-specific 5-HT antagonist that has been shown to block development of serotonin syndrome in animals (Sternbach, 1991). Cyproheptadine has been used in the treatment of serotonin syndrome (Mills, 1997; Goldberg & Huk, 1992). There are no controlled human trials substantiating its efficacy.
    b) ADULT: 12 mg initially followed by 2 mg every 2 hours if symptoms persist, up to a maximum of 32 mg in 24 hours. Maintenance dose 8 mg orally repeated every 6 hours (Boyer & Shannon, 2005).
    c) CHILD: 0.25 mg/kg/day divided every 6 hours, maximum dose 12 mg/day (Mills, 1997).
    5) HYPERTENSION
    a) Monitor vital signs regularly. For mild/moderate asymptomatic hypertension, pharmacologic intervention is usually not necessary.
    6) HYPOTENSION
    a) Administer 10 to 20 mL/kg 0.9% saline bolus and place patient supine. Further fluid therapy should be guided by central venous pressure or right heart catheterization to avoid volume overload.
    b) Pressor agents with dopaminergic effects may theoretically worsen serotonin syndrome and should be used with caution. Direct acting agents (norepinephrine, epinephrine, phentolamine) are theoretically preferred.
    c) NOREPINEPHRINE
    1) PREPARATION: Add 4 mL of 0.1% solution to 1000 mL of dextrose 5% in water to produce 4 mcg/mL.
    2) INITIAL DOSE
    a) ADULT: 2 to 3 mL (8 to 12 mcg)/minute.
    b) ADULT or CHILD: 0.1 to 0.2 mcg/kg/min. Titrate to maintain adequate blood pressure.
    3) MAINTENANCE DOSE
    a) 0.5 to 1 mL (2 to 4 mcg)/minute.
    7) SEIZURES
    a) DIAZEPAM
    1) MAXIMUM RATE: Administer diazepam IV over 2 to 3 minutes (maximum rate: 5 mg/min).
    2) ADULT DIAZEPAM DOSE: 5 to 10 mg initially, repeat every 5 to 10 minutes as needed. Monitor for hypotension, respiratory depression and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after diazepam 30 milligrams.
    3) PEDIATRIC DIAZEPAM DOSE: 0.2 to 0.5 mg/kg, repeat every 5 minutes as needed. Monitor for hypotension, respiratory depression and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after diazepam 10 milligrams in children over 5 years or 5 milligrams in children under 5 years of age.
    4) RECTAL USE: If an intravenous line cannot be established, diazepam may be given per rectum (not FDA approved), or lorazepam may be given intramuscularly.
    b) LORAZEPAM
    1) MAXIMUM RATE: The rate of IV administration of lorazepam should not exceed 2 mg/min (Prod Info Ativan(R), 1991).
    2) ADULT LORAZEPAM DOSE: 2 to 4 mg IV. Initial doses may be repeated in 10 to 15 minutes, if seizures persist (Prod Info ATIVAN(R) injection, 2003).
    3) PEDIATRIC LORAZEPAM DOSE: 0.1 mg/kg IV push (range: 0.05 to 0.1 mg/kg; maximum dose 4 mg); may repeat dose in 5 to 10 minutes if seizures continue. It has also been given rectally at the same dose in children with no IV access (Sreenath et al, 2009; Chin et al, 2008; Wheless, 2004; Qureshi et al, 2002; De Negri & Baglietto, 2001; Mitchell, 1996; Appleton, 1995; Giang & McBride, 1988).
    c) RECURRING SEIZURES
    1) If seizures cannot be controlled with diazepam or recur, give phenobarbital or propofol.
    d) PHENOBARBITAL
    1) SERUM LEVEL MONITORING: Monitor serum levels over next 12 to 24 hours for maintenance of therapeutic levels (15 to 25 mcg/mL).
    2) ADULT PHENOBARBITAL LOADING DOSE: 600 to 1200 mg of phenobarbital IV initially (10 to 20 mg/kg) diluted in 60 mL of 0.9% saline given at 25 to 50 mg/minute.
    3) ADULT PHENOBARBITAL MAINTENANCE DOSE: Additional doses of 120 to 240 mg may be given every 20 minutes.
    4) MAXIMUM SAFE ADULT PHENOBARBITAL DOSE: No maximum safe dose has been established. Patients in status epilepticus have received as much as 100 mg/min until seizure control was achieved or a total dose of 10 mg/kg.
    5) PEDIATRIC PHENOBARBITAL LOADING DOSE: 15 to 20 mg/kg of phenobarbital intravenously at a rate of 25 to 50 mg/min.
    6) PEDIATRIC PHENOBARBITAL MAINTENANCE DOSE: Repeat doses of 5 to 10 mg/kg may be given every 20 minutes.
    7) MAXIMUM SAFE PEDIATRIC PHENOBARBITAL DOSE: No maximum safe dose has been established. Children in status epilepticus have received doses of 30 to 120 mg/kg within 24 hours. Vasopressors and mechanical ventilation were needed in some patients receiving these doses.
    8) NEONATAL PHENOBARBITAL LOADING DOSE: 20 to 30 mg/kg IV at a rate of no more than 1 mg/kg/min in patients with no preexisting phenobarbital serum levels.
    9) NEONATAL PHENOBARBITAL MAINTENANCE DOSE: Repeat doses of 2.5 mg/kg every 12 hours may be given; adjust dosage to maintain serum levels of 20 to 40 mcg/mL.
    10) MAXIMUM SAFE NEONATAL PHENOBARBITAL DOSE: Doses of up to 20 mg/kg/min up to a total of 30 mg/kg have been tolerated in neonates.
    11) CAUTION: Adequacy of ventilation must be continuously monitored in children and adults. Intubation may be necessary with increased doses.
    8) CHLORPROMAZINE
    a) Chlorpromazine is a 5-HT2 receptor antagonist that has been used to treat cases of serotonin syndrome (Graham, 1997; Gillman, 1996). Controlled human trial documenting its efficacy are lacking.
    b) ADULT: 25 to 100 mg intramuscularly repeated in 1 hour if necessary.
    c) CHILD: 0.5 to 1 mg/kg repeated as needed every 6 to 12 hours not to exceed 2 mg/kg/day.
    9) NOT RECOMMENDED
    a) BROMOCRIPTINE: It has been used in the treatment of neuroleptic malignant syndrome but is NOT RECOMMENDED in the treatment of serotonin syndrome as it has serotonergic effects (Gillman, 1997). In one case the use of bromocriptine was associated with a fatal outcome (Kline et al, 1989).

Inhalation Exposure

    6.7.2) TREATMENT
    A) LEUKOENCEPHALOPATHY
    1) Coenzyme Q has been postulated as a possible therapy for spongiform leukoencephalopathy induced by inhalation of heroin pyrolysate (Long et al, 2003). One suggested regimen is 300 mg four times daily. These are no controlled studies to support its efficacy.
    B) Treatment should include recommendations listed in the ORAL EXPOSURE section when appropriate.

Case Reports

    A) ADULT
    1) CASE REPORT: A 33-year-old woman in active labor was inadvertently given 45 mcg (approximately 9-fold greater than normal dosing) of sufentanil into the intrathecal space and complained of pruritus within 2 minutes of receiving the dose and some difficulty swallowing. No respiratory depression or alteration in vital signs were noted. During delivery the patient had inadequate pain control and reported pain of 7/10, an epidural infusion of bupivacaine was started for pain control. The infant's heart rate dropped which resulted in rapid delivery with an assistive device within 12 minutes of the event. Apgar scores of 8 and 9 were reported at 1 and 5 minutes, respectively. The woman received repeated doses of naloxone (total dose: 520 mcg over 60 minutes) shortly after delivery for complaints of severe pruritus. Symptoms markedly improved but only lasted 7 to 10 minutes. The patient and infant were discharged to home the following day (Coleman et al, 2009). The authors suggested that repeated doses of naloxone may have prevented the development of other adverse events (respiratory depression, sedation, nausea) associated with intrathecal exposure. However, the patient did not receive her first dose until 55 minutes after exposure.

Summary

    A) TOXICITY: The toxicity of an opioid varies with the agent, as well as with tolerance developed from habitual use. Infants and children have unusual sensitivity to opioid agents.

Therapeutic Dose

    7.2.1) ADULT
    A) GENERAL
    1) Opioids are subject to dosage variability based on the intended indication and the patient's ability to handle the drug.
    a) The goal of therapy for analgesia is to use the smallest effective dose. The following summarizes usual adult dosages.
    b) Please refer to the manufacturer's information for more specific information on individual agents.
    B) SPECIFIC SUBSTANCE
    1) ALVIMOPAN
    a) To accelerate the time to upper and lower gastrointestinal recovery following partial large or small bowel resection surgery or primary anastomosis: 12 mg (1 capsule) given 30 minutes to 5 hours prior to surgery followed by 12 mg twice daily beginning the day after surgery for a maximum of 7 days; MAXIMUM: 15 doses. Alvimopan is only distributed to hospitals that have registered and are participating in the ENTEREG(R) Access Support and Education (EASE) program (Call 1-877-282-4786 for further information) (Prod Info ENTEREG(R) oral capsules, 2012).
    2) ELUXADOLINE
    a) ORAL: USUAL DOSE: 100 mg orally 2 times daily (Prod Info VIBERZI oral tablets, 2015).
    b) ORAL: DOSE IN PATIENTS WHO DO NOT HAVE A GALLBLADDER: 75 mg orally 2 times daily (Prod Info VIBERZI oral tablets, 2015).
    3) OPIUM
    a) ORAL: 10% tincture, 0.6 mL 4 times daily; maximum 6 mL/day (Prod Info opium tincture, 2003)
    b) ORAL: Paregoric (2 mg morphine per 5 mL), 5-10 mL 1 to 4 times daily. Maximum 40 mL/day (Prod Info Paregoric, 1998).
    c) RECTAL: 30 to 60 mg once to twice daily not to exceed 4 doses daily (Prod Info B&O SUPPRETTES(R) No. 15A and No. 16A rectal suppositories, 2002).
    4) REMIFENTANIL
    a) NOTE: Remifentanil is not recommended as the sole agent in general anesthesia because loss of consciousness cannot be assured and because of a high incidence of apnea, muscle rigidity, and tachycardia. It is synergistic with other anesthetics, and the doses of thiopental, propofol, isoflurane, and midazolam may need to be reduced by up to 75% with concomitant administration of remifentanil. Administration must be individualized based on patient response (Prod Info Ultiva(R), remifentanil hydrochloride, 1999).
    b) PARENTERAL: IV infusion of 0.5 to 1 mcg/kg/min, with an initial dose of 1 mcg/kg administered over 30 to 60 seconds for general anesthesia and continuing as an analgesic (Prod Info Ultiva(R), remifentanil hydrochloride, 1996).
    5) SUFENTANIL
    a) PARENTERAL: 8 to 30 mcg/kg IV with 100% oxygen and a muscle relaxant, then 0.5 to 10 mcg/kg as needed in response to signs of lightening of anesthesia. Maximum dose of 30 mcg/kg/procedure (Prod Info sufentanil citrate injection, 2003).
    6) EQUIVALENT DOSES
    a) The following table presents approximate equivalent analgesic doses for various opioids. These doses are primarily based on single-dose studies in healthy adults and do not reflect individual variation due to tolerance, weight or age.
    EQUIANALGESIC (THERAPEUTIC) DOSES
    OPIATEIM OR IV (mg)ORAL (mg)
    Alfentanilno data availableno data available
    Alphaprodine40 to 60no data available
    Buprenorphine0.40.4 to 0.8 SL
    Butorphanol28
    Codeine120200
    Dihydrocodeine10 to 5010 to 30
    Ethoheptazineno data available1000
    Fentanyl0.1 to 0.2no data available
    Hydrocodone5 to 1025 to 30
    Hydromorphone1.57.5
    Heroin3 to 560
    Levorphanol24
    Meperidine75 to 100300
    Morphine1060
    Nalbuphine1050 to 60
    Oxycodone1520 to 30
    Oxymorphone1.56
    Pentazocine30 to 60180 to 250
    Propoxypheneno data available130
    Sufentanil0.01 to 0.02no data available
    (Inturrisi, 1982; Kastrup, 1988)

    7.2.2) PEDIATRIC
    A) SUMMARY
    1) Opioids are subject to dosage variability based on the intended indication and the patient's ability to handle the drug.
    a) The goal of therapy for analgesia is to use the smallest effective dose. The following summarizes usual pediatric dosages.
    b) Please refer to manufacturer's information for more specific information on individual agents.
    B) SPECIFIC SUBSTANCE
    1) ELUXADOLINE
    a) Safety and effectiveness have not been established in pediatric patients (Prod Info VIBERZI oral tablets, 2015).
    2) REMIFENTANIL
    a) PARENTERAL (Initial): 0.25 micrograms/kilogram/minute (Prod Info Ultiva(R), 2004)
    b) PARENTERAL (Maintenance): 0.05 to 1.3 micrograms/kilogram/minute; supplemental doses of 1 micrograms/kilogram every 2 to 5 min if needed (Prod Info Ultiva(R), 2004)
    3) SUFENTANIL
    a) PARENTERAL: 10 to 25 micrograms/kilogram with 100% oxygen, additional doses of up to 25 to 50 micrograms for maintenance of anesthesia (Prod Info sufentanil citrate injection, 2003).

Minimum Lethal Exposure

    A) SPECIFIC SUBSTANCE
    1) ETORPHINE
    a) Death has been reported in humans following a total dose of 0.03 to 0.12 milligram of etorphine.
    2) HEROIN
    a) Fatal heroin overdoses have been reported following the parenteral route, ingestions, inhalation, and nasal administration. No difference between median blood morphine concentrations of injecting and non-injecting users is apparent (Darke & Ross, 2000). Risk factors contributing to fatalities following heroin overdose include loss of tolerance, concomitant use of alcohol and other CNS depressants, and pulmonary and hepatic dysfunction resulting from systemic disease (Warner-Smith et al, 2001).

Maximum Tolerated Exposure

    A) SPECIFIC SUBSTANCE
    1) ALVIMOPAN
    a) During clinical trials, healthy adults tolerated single doses up to 120 mg and multiple doses up to 48 mg for 7 days (Prod Info ENTEREG(R) oral capsules, 2008).
    2) DIAMORPHINE
    a) CASE REPORT: A 52-year-old woman developed only nausea and mild drowsiness after inadvertently receiving 3 mL (3 mg) of diamorphine intrathecally instead of 0.5% bupivacaine for spinal anesthesia for a total knee replacement surgery. She recovered after 48 hours of observation (Jafar & Chowdhry, 2014).
    3) NALBUPHINE
    a) Single 72 milligram intramuscular dose of nalbuphine in normal subjects produced minimal toxic effects of drowsiness and dysphoria (Prod Info Nubain(R), 1983) .
    4) REMIFENTANIL
    a) A primiparous woman who had received epidural fentanyl with bupivacaine (three 10-mL dose of bupivacaine 0.1% with fentanyl 2 mcg/mL) for labor pain, became unconscious and developed respiratory arrest within 5 minutes of self-administering the first dose of remifentanil (programmed regimen: 40 mcg/mL, 1-mL bolus [40 mcg], 2-min lockout) patient-controlled analgesia (PCA). She recovered within 30 to 60 seconds of supportive care. The PCA machine was examined and it was determined that 160 mcg was inadvertently delivered over 4 attempts (Pruefer & Bewlay, 2012).

Serum Plasma Blood Concentrations

    7.5.1) THERAPEUTIC CONCENTRATIONS
    A) THERAPEUTIC CONCENTRATION LEVELS
    1) SPECIFIC SUBSTANCE
    a) REMIFENTANIL
    1) Whole blood therapeutic concentration range of remifentanil varies between 20 to 60 nanograms/milliliter (Asselborn et al, 2002).
    7.5.2) TOXIC CONCENTRATIONS
    A) TOXIC CONCENTRATION LEVELS
    1) SPECIFIC SUBSTANCE
    a) AH-7921
    1) Nine patients were found dead after using AH-7921, a designer opioid with about 80% of morphine's mu-agonist activity. Six patients also used other drugs, such as benzodiazepines, antidepressants, and analgesics; however, it was concluded that poly-drug use (all agents with postmortem therapeutic concentrations) was not a major contributing factor for the fatal outcome. A selective liquid chromatography-mass spectrometry (MS)-MS method detected AH-7921 concentrations, ranging from 0.03 to 0.99 mcg/g blood. Metabolites of the parent compound, found in the blood, included 2 N-demethylated that dominated and 4 mono-hydroxylated in trace amounts. Death occurred both at low and high concentrations of AH-7921, probably due to the differences in tolerance of the drug (Kronstrand et al, 2014).
    b) ALFENTANIL
    1) A perimortem blood sample, taken during resuscitation, was reported to contain 79 micrograms/liter of alfentanil which was self-administered parenterally in a 27-year-old for recreational use (Bralthwaite et al, 2000).
    c) ALPHAPRODINE
    1) 20 milligrams intravenously in a 215 pound man who died one hour later produced blood concentrations of 0.62 milligrams/liter by gas chromatography in postmortem specimens (Baselt, 1982).
    d) ANILERIDINE
    1) Therapeutic blood or plasma concentrations have not been established (Baselt, 1982). Postmortem blood anileridine concentrations in 2 cases of intentional ingestion were 0.9 milligram/liter and 2 milligrams/liter, respectively (Baselt, 1982).
    e) HEROIN
    1) Addicts self-administering intravenous heroin in doses of 150 to 200 milligrams developed plasma morphine levels as high as 0.3 milligram/liter (Bolelli et al, 1979).
    2) In five cases of fatal heroin overdose, the minimal lethal blood concentration was 0.021 micrograms morphine per milliliter (Kintz et al, 1989).
    3) The tissue/blood ratio of heroin metabolites in a series of 25 heroin deaths were as follows (Wyman & Bultman, 2004):
      Vitreous Humor (VH)/Blood Ratio CSF/Blood Ratio Liver/Blood Ratio
    6-Monoacetylmorphine (6-MAM) 11.3 +/- 9.4 (range 1.7 to 27) n=13 6.6 +/- 4.4 (2.6 to 17.3) n=13 Not detected in liver
    Free Morphine0.36 +/- 0.18 (0.11 to 0.78) n=25 0.64 +/- 0.27 (0.25 to 1.56)n=25 2.99 +/- 2.12(1.6 to 11.9)n=25
    Codeine 1.03 +/- 0.39(0.50 to 2.0)n=25 0.94 +/- 0.26(0.55 to 2.00)n=25 2.67 +/- 0.79(1.57 to 4.00)n=25

    a) The results show that postmortem analysis of the vitreous humor may be useful in determining the presence of 6-MAM, a heroin metabolite that is often used to distinguish between heroin and morphine exposures.
    f) METHYLFENTANYL
    1) Blood concentrations found in 8 adults who died after intravenous overdosage ranged from 2 to 11 micrograms/liter (Reed, 1981; Gillespie et al, 1982).
    g) SUFENTANIL
    1) Postmortem sufentanil blood concentrations were 2.6 mcg/L and 3.0 mcg/L in two men who were found dead after apparent intravenous abuse of sufentanil (Baselt, 2004).

Pharmacologic Mechanism

    A) Opioids interact with stereospecific and saturable binding sites in the CNS and other tissues, with the highest concentration in the limbic system, thalamus, striatum, hypothalamus, mid brain and spinal cord (Snyder et al, 1974; Simon & Hiller, 1978; Gilman et al, 1980). Their effects may result from mimicking the actions of enkephalins, beta-endorphin and other exogenous ligands which have been found to occupy the same binding sites (Gilman et al, 1980).
    B) These depress the central nervous system and may produce coma and cessation of respiration. Addiction following chronic usage of these agents is common and results in a withdrawal state upon their termination.
    C) Opioids act by binding to several receptor types within the central nervous system, as well as in other areas of the body (Seifert, 2004). A nomenclature change for these receptors has been recommend and its been suggested to move away from the Greek terminology. The suggested nomenclature is thought to be more consistent with other neurotransmitters. The term opiate peptide (OP) is used to define the receptor types (Nelson, 2006).
    D) Multiple opioid receptor types, (e.g., mu (OP3), kappa (OP2), delta (OP1)), appear to subserve different physiologic functions (Jaffe & Martin, 1990).
    1) Mu (OP3) receptor stimulation will produce supraspinal (brain) analgesia, respiratory depression, gastrointestinal dysmotility, sedation, bradycardia, euphoria and physical dependence (Nelson, 2006).
    2) Delta (OP1) receptor stimulation appears to interact with Mu (OP3) receptors to produce spinal and supraspinal analgesia. These receptors are also mediate dopamine release (Seifert, 2004).
    3) Kappa (OP2) receptor stimulation will produce spinal analgesia, miosis, diuresis and dysphoria (Nelson, 2006).
    E) Opioid drugs are categorized according to their capacity to bind and activate the various receptor sites. Agonists are those opioids that bind and activate a receptor. Antagonists are those opioids that bind but do not activate a receptor.

Toxicologic Mechanism

    A) RESPIRATORY DEPRESSION - Respiration, which is controlled mainly through medullary respiratory centers with peripheral input from chemoreceptors and other sources, is affected by opioids which produce inhibition at chemoreceptors via Mu (OP3) opioid receptors and in the medulla via mu and delta receptors. Tolerance develops more quickly to euphoria and other effects than to respiratory effects (White & Irvine, 1999).
    B) PRIAPISM - may be induced by opiates due to their sympatholytic actions which may induce an unopposed cholinergic state that could result in Ach-induced vasorelaxation involving NO pathways. Alpha-blockade has also been postulated as a mechanism. Autonomic system dysregulation appears not to be dose-specific (Nocchi & D'az, 1999).

Physical Characteristics

    A) ALFENTANIL HYDROCHLORIDE is a sterile, nonpyrogenic, preservative-free aqueous solution (Prod Info alfentanil HCl intravenous injection, 2013), which is soluble in water; freely soluble in alcohol, in chloroform, and in methanol; and sparingly soluble in acetone (Prod Info Alfenta(R), 1995).
    B) OPIUM is a light brown to yellow brown powder with characteristic odor (Prod Info opium tincture (DEODORIZED) oral solution, 2008).
    C) REMIFENTANIL is a white to off-white lyophilized powder (Prod Info ULTIVA(R) IV injection, 2007).
    D) SUFENTANIL is a nonpyrogenic, aqueous solution (Prod Info sufentanil citrate intravenous injection, epidural injection, 2011).
    E) The following compounds are soluble in alcohol: alphaprodine hydrochloride, ethoheptazine citrate (Prod Info EMBEDA(R) oral extended-release capsules, 2009; Prod Info AVINZA(R) extended-release oral capsules, 2008; Prod Info OPANA(R)ER extended release oral tablets, 2007).

Ph

    A) ALFENTANIL HYDROCHLORIDE: 4 to 6 (Prod Info alfentanil HCl intravenous injection, 2013)
    B) REMIFENTANIL: 2.5 to 3.5 (Prod Info ULTIVA(R) IV injection, 2007)
    C) SUFENTANIL: 3.5 to 6 (Prod Info sufentanil citrate intravenous injection, epidural injection, 2011)

Molecular Weight

    A) ALFENTANIL HYDROCHLORIDE: 452.98 (Prod Info alfentanil HCl intravenous injection, 2013)
    B) ELUXADOLINE: 569.65 (Prod Info VIBERZI oral tablets, 2015)
    C) NALBUPHINE HYDROCHLORIDE: 393.91 (Prod Info NUBAIN(R) injection, 2005)
    D) REMIFENTANIL: 412.91 (Prod Info ULTIVA(R) IV injection, 2007)
    E) SUFENTANIL CITRATE: 578.68 (Prod Info sufentanil citrate intravenous injection, epidural injection, 2011)

Clinical Effects

    11.1.3) CANINE/DOG
    A) Signs may include: drowsiness, ataxia, vomiting, seizures, miosis, coma, respiratory depression, and hypotension. Death can occur within 12 hours (Dumonceaux & Beasley, 1990).
    11.1.5) EQUINE/HORSE
    A) Opioids and their derivatives have an excitatory effect on the CNS of horses.
    11.1.6) FELINE/CAT
    A) Opioids and their derivatives have an excitatory effect on the CNS of cats.

Treatment

    11.2.1) SUMMARY
    A) GENERAL TREATMENT
    1) SUMMARY
    a) Begin treatment immediately.
    b) Keep animal warm and do not handle unnecessarily.
    c) Remove the patient and other animals from the source of contamination or remove dietary sources.
    2) Treatment should always be done on the advice and with the consultation of a veterinarian.
    3) Additional information regarding treatment of poisoned animals may be obtained from a Veterinary Toxicologist or the National Animal Poison Control Center.
    4) ASPCA ANIMAL POISON CONTROL CENTER
    a) ASPCA Animal Poison Control Center, 1717 S Philo Road, Suite 36 Urbana, IL 61802
    b) It is an emergency telephone service which provides toxicology information to veterinarians, animal owners, universities, extension personnel and poison center staff for a fee. A veterinary toxicologist is available for consultation.
    c) Contact information: (888) 426-4435 (hotline) or www.aspca.org (A fee may apply. Please inquire with the poison center). The agency will make follow-up calls as needed in critical cases at no extra charge.
    11.2.2) LIFE SUPPORT
    A) GENERAL
    1) MAINTAIN VITAL FUNCTIONS: Secure airway, supply oxygen, and begin supportive fluid therapy if necessary.
    11.2.4) DECONTAMINATION
    A) GASTRIC DECONTAMINATION
    1) GENERAL TREATMENT
    a) EMESIS/GASTRIC LAVAGE -
    1) CAUTION: Carefully examine patients with chemical exposure before inducing emesis. If signs of oral, pharyngeal, or esophageal irritation, a depressed gag reflex, or central nervous system excitation or depression are present, EMESIS SHOULD NOT BE INDUCED.
    2) HORSES OR CATTLE: DO NOT attempt to induce emesis in ruminants (cattle) or equids (horses).
    3) DOGS AND CATS
    a) IPECAC: If within 2 hours of exposure: induce emesis with 1 to 2 milliliters/kilogram syrup of ipecac per os.
    b) APOMORPHINE: Dogs may vomit more readily with 1 tablet (6 milligrams) apomorphine diluted in 3 to 5 milliliters water and instilled into the conjunctival sac or per os.
    1) Dogs may also be given apomorphine intravenously at 40 micrograms/kilogram, although this route may not be as effective.
    4) LAVAGE: In the absence of a gag reflex or if vomiting cannot be induced, place a cuffed endotracheal tube and begin gastric lavage.
    a) Pass large bore stomach tube and instill 5 to 10 milliliters/kilogram water or lavage solution, then aspirate. Repeat 10 times.
    b) ACTIVATED CHARCOAL/CATHARTIC -
    1) ACTIVATED CHARCOAL: Administer activated charcoal. Dose: 2 grams/kilogram per os or via stomach tube. Avoid aspiration by proper restraint, careful technique, and if necessary tracheal intubation.
    2) CATHARTIC: Administer a dose of a saline or sorbitol cathartic such as magnesium or sodium sulfate (sodium sulfate dose is 1 gram/kilogram). If access to these agents is limited, give 5 to 15 milliliters magnesium oxide (Milk of Magnesia) per os for dilution.
    3) ACTIVATED CHARCOAL/HORSES: Administer 0.5 to 1 kilogram of activated charcoal in up to 1 gallon warm water via nasogastric tube. Neonates: administer 250 grams (one-half pound) activated charcoal in up to 2 quarts water.
    4) ACTIVATED CHARCOAL/RUMINANTS: Administer 2 to 9 grams/ kilogram of activated charcoal in a slurry of 1 gram charcoal/3 to 5 milliliters warm water via stomach tube. Sheep may be given 0.5 kilogram charcoal in slurry.
    5) CATHARTICS/HORSES: Mineral oil is administered 30 minutes after activated charcoal. DOSE: 4 to 6 liters in adult horses and 1 to 4 liters in neonates or foals.
    a) Magnesium sulfate: 0.2 to 0.9 grams/kilogram (500 grams for adults).
    b) The sulfate laxatives are especially effective when given 30 to 45 minutes after mineral oil administration.
    c) Carbachol (lentin): administer 1 milligram to an adult.
    6) CATHARTICS/RUMINANTS & SWINE: Adult cattle: administer 500 grams sodium or magnesium sulfate. Other ruminants and swine: administer 1 to 2 grams/kilogram.
    a) The sulfate laxatives are especially effective when given 30 to 45 minutes after cathartic administration.
    b) Mineral oil: Do not administer within 30 minutes of activated charcoal. DOSE: small ruminants and swine, 60 to 200 milliliters; cattle, 0.5 to 1 gallon.
    c) Magnesium oxide: (Milk of Magnesia) Small ruminants, up to 0.25 gram/kilogram in 1 to 3 gallons warm water; adult cattle up to 1 gram/kilogram in 1 to 3 gallons warm water or 2 to 4 boluses MgOH per os.
    d) Give these solutions via stomach tube and monitor for aspiration.
    11.2.5) TREATMENT
    A) GENERAL TREATMENT
    1) NALOXONE -
    a) Administer naloxone 0.02 milligram/kilogram (1 milliliter per 40 pounds body weight) intramuscularly, or intravenously if patient is in shock or severe respiratory depression.
    2) SEIZURES -
    a) SEIZURES/LARGE ANIMALS: May be controlled with diazepam.
    1) HORSES/DIAZEPAM: Neonates: 0.05 to 0.4 milligrams/kilogram; Adults: 25 to 50 milligrams. Give slowly intravenously to effect; repeat in 30 minutes if necessary.
    2) CATTLE, SHEEP AND SWINE/DIAZEPAM: 0.5 to 1.5 milligrams/kilogram intravenously to effect.
    b) SEIZURES/DOGS & CATS:
    1) DIAZEPAM: 0.5 to 2 milligrams/kilogram intravenous bolus; may repeat dose every ten minutes for four total doses. Give slowly over 1 to 2 minutes to effect.
    2) PHENOBARBITAL: 5 to 30 milligrams/kilogram over 5 to 10 minutes intravenously to effect.
    3) REFRACTORY SEIZURES: Consider anaesthesia or heavy sedation. Administer pentobarbital to DOGS & CATS at a dose of 3 to 15 milligrams/kilogram intravenously slowly to effect. May need to repeat in 4 to 8 hours. Be sure to protect the airway.
    3) HYPOTENSION -
    a) FLUIDS: Begin intravenous administration of lactated ringers or other solution at a rate of up to 60 milliliters/kilograms/hour.
    1) If packed cell volume is less than 25 percent or total protein is less than 3.5 grams/deciliter, give 10 to 20 milliliters/kilogram of the appropriate solution (whole blood, plasma, packed cells or dextran).
    b) DOPAMINE: If the patient does not respond to volume loading, administer dopamine (DOGS & CATS) at a dose not to exceed 2 to 5 micrograms/kilogram/minute.
    c) DOBUTAMINE: If inadequate cardiac function is contributing to poor peripheral perfusion, administer dobutamine (DOGS & CATS): 3 to 10 micrograms/kilogram/minute.
    d) NOREPINEPHRINE:
    1) NOREPINEPHRINE is the drug of choice to combat hypotension. Do NOT use epinephrine.
    2) Dosage: Levarteranol bitartrate, in ampules of 2 milligrams/milliliter: Dilute 1 to 2 milliliters in 250 milliliters intravenous solution. Give as IV drip to effect.
    3) Reported dosages are for small animals; modify as needed.
    4) MONITORING -
    a) Admit all symptomatic patients and begin treatment.
    b) Keep the patient for 8 hours after cessation of clinical signs in case of relapse.

Continuing Care

    11.4.1) SUMMARY
    11.4.1.2) DECONTAMINATION/TREATMENT
    A) GENERAL TREATMENT
    1) SUMMARY
    a) Begin treatment immediately.
    b) Keep animal warm and do not handle unnecessarily.
    c) Remove the patient and other animals from the source of contamination or remove dietary sources.
    2) Treatment should always be done on the advice and with the consultation of a veterinarian.
    3) Additional information regarding treatment of poisoned animals may be obtained from a Veterinary Toxicologist or the National Animal Poison Control Center.
    4) ASPCA ANIMAL POISON CONTROL CENTER
    a) ASPCA Animal Poison Control Center, 1717 S Philo Road, Suite 36 Urbana, IL 61802
    b) It is an emergency telephone service which provides toxicology information to veterinarians, animal owners, universities, extension personnel and poison center staff for a fee. A veterinary toxicologist is available for consultation.
    c) Contact information: (888) 426-4435 (hotline) or www.aspca.org (A fee may apply. Please inquire with the poison center). The agency will make follow-up calls as needed in critical cases at no extra charge.
    11.4.2) DECONTAMINATION
    11.4.2.2) GASTRIC DECONTAMINATION
    A) GASTRIC DECONTAMINATION
    1) GENERAL TREATMENT
    a) EMESIS/GASTRIC LAVAGE -
    1) CAUTION: Carefully examine patients with chemical exposure before inducing emesis. If signs of oral, pharyngeal, or esophageal irritation, a depressed gag reflex, or central nervous system excitation or depression are present, EMESIS SHOULD NOT BE INDUCED.
    2) HORSES OR CATTLE: DO NOT attempt to induce emesis in ruminants (cattle) or equids (horses).
    3) DOGS AND CATS
    a) IPECAC: If within 2 hours of exposure: induce emesis with 1 to 2 milliliters/kilogram syrup of ipecac per os.
    b) APOMORPHINE: Dogs may vomit more readily with 1 tablet (6 milligrams) apomorphine diluted in 3 to 5 milliliters water and instilled into the conjunctival sac or per os.
    1) Dogs may also be given apomorphine intravenously at 40 micrograms/kilogram, although this route may not be as effective.
    4) LAVAGE: In the absence of a gag reflex or if vomiting cannot be induced, place a cuffed endotracheal tube and begin gastric lavage.
    a) Pass large bore stomach tube and instill 5 to 10 milliliters/kilogram water or lavage solution, then aspirate. Repeat 10 times.
    b) ACTIVATED CHARCOAL/CATHARTIC -
    1) ACTIVATED CHARCOAL: Administer activated charcoal. Dose: 2 grams/kilogram per os or via stomach tube. Avoid aspiration by proper restraint, careful technique, and if necessary tracheal intubation.
    2) CATHARTIC: Administer a dose of a saline or sorbitol cathartic such as magnesium or sodium sulfate (sodium sulfate dose is 1 gram/kilogram). If access to these agents is limited, give 5 to 15 milliliters magnesium oxide (Milk of Magnesia) per os for dilution.
    3) ACTIVATED CHARCOAL/HORSES: Administer 0.5 to 1 kilogram of activated charcoal in up to 1 gallon warm water via nasogastric tube. Neonates: administer 250 grams (one-half pound) activated charcoal in up to 2 quarts water.
    4) ACTIVATED CHARCOAL/RUMINANTS: Administer 2 to 9 grams/ kilogram of activated charcoal in a slurry of 1 gram charcoal/3 to 5 milliliters warm water via stomach tube. Sheep may be given 0.5 kilogram charcoal in slurry.
    5) CATHARTICS/HORSES: Mineral oil is administered 30 minutes after activated charcoal. DOSE: 4 to 6 liters in adult horses and 1 to 4 liters in neonates or foals.
    a) Magnesium sulfate: 0.2 to 0.9 grams/kilogram (500 grams for adults).
    b) The sulfate laxatives are especially effective when given 30 to 45 minutes after mineral oil administration.
    c) Carbachol (lentin): administer 1 milligram to an adult.
    6) CATHARTICS/RUMINANTS & SWINE: Adult cattle: administer 500 grams sodium or magnesium sulfate. Other ruminants and swine: administer 1 to 2 grams/kilogram.
    a) The sulfate laxatives are especially effective when given 30 to 45 minutes after cathartic administration.
    b) Mineral oil: Do not administer within 30 minutes of activated charcoal. DOSE: small ruminants and swine, 60 to 200 milliliters; cattle, 0.5 to 1 gallon.
    c) Magnesium oxide: (Milk of Magnesia) Small ruminants, up to 0.25 gram/kilogram in 1 to 3 gallons warm water; adult cattle up to 1 gram/kilogram in 1 to 3 gallons warm water or 2 to 4 boluses MgOH per os.
    d) Give these solutions via stomach tube and monitor for aspiration.

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) Abelson JL: Urine drug testing: watch what you eat! (letter). JAMA 1991; 266:3130-3131.
    4) Addiego JE, Ridgway D, & Bleyer WA: The acute management of intrathecal methotrexate overdose: pharmacologic rationale and guidelines. J Pediatr 1981; 98(5):825-828.
    5) Aderjan RE & Skopp G: Formation and clearance of active and inactive metabolites of opiates in humans. Ther Drug Monitor 1998; 20:561-569.
    6) Aitkenhead AR, Lin ES, & Achola KJ: The pharmacokinetics and intravenous nalbuphine in healthy volunteers. Br J Clin Pharmacol 1988; 25:264-268.
    7) Alaspaa AO, Kuisma MJ, Hoppu K, et al: Out-of-hospital administration of activated charcoal by emergency medical services. Ann Emerg Med 2005; 45:207-12.
    8) Amin HM, Sopchak AM, & Esposito BF: Naloxone-induced and spontaneous reversal of depressed ventilatory responses to hypoxia during and after continuous infusion of remifentanil or alfentanil. J Pharmacol Exp Therapeut 1995; 274:34-39.
    9) Anderson K: Bronchospasm and intravenous street heroin (letter). Lancet 1986; 1:1208.
    10) Angelo HR & Hausner B: Testing for ketobemidone and its main metabolites in urine from drug addicts using GC/MS (abstract). Clin Toxicol 2002; 40:390.
    11) Anon: American academy of pediatrics committee on drugs: transfer of drugs and other chemicals into human milk. Pediatrics 2001; 108(3):776-789.
    12) Antonopoulos S, Balatsouras DG, Kanakaki S, et al: Bilateral sudden sensorineural hearing loss caused by alcohol abuse and heroin sniffing. Auris Nasus Larynx 2012; 39(3):305-309.
    13) Appleton R: Lorazepam vs diazepam in the acute treatment of epileptic seizures and status epilepticus.. Dev Med Child Neuro 1995; 37:682-688.
    14) 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.
    15) Arvanitis ML & Satonik RC: Transdermal fentanyl abuse and misuse (letter). Am J Emerg Med 2002; 20:58-59.
    16) Asselborn G, Yegles M, & Wennig R: Suicide with remifentanil and midazolam: a case report. Acta Clinica Belgica 2002; (Suppl 1):54-57.
    17) Baden LR, Horowitz G, & Jacoby H: Quinolones and false-positive urine screening for opiates by immunoassay technology. J Amer Med Assc 2001; 286:3115-3119.
    18) Balikova M & Maresova V: Fatal opiates overdose. Toxicological identification of various metabolites in a blood sample by GC-MS after silylation. Forensic Sci Int 1998; 94:201-209.
    19) Baselt RC: Disposition of Toxic Drugs and Chemicals in Man, 2nd ed, Biomedical Publications, Davis, CA, 1982.
    20) Baselt RC: Disposition of Toxic Drugs and Chemicals in Man, 5th ed, Chemical Toxicology Institute, Foster City, CA, 2000.
    21) Baselt RC: Disposition of Toxic Drugs and Chemicals in Man, 7th ed. Biomedical Publications, Foster City, CA, 2004, pp -.
    22) Batora I, Kukumberg P, & Plackova S: Atypical clinical picture and course of heroin intoxication (abstract). Clin Toxicol 2002; 40:326-327.
    23) Beck O, Vitols S, & Stensio M: Positive urine screening for opiates after consumption of sandwich bread with poppy seed flavoring (letter). Ther Drug Monit 1990; 12:585-586.
    24) Bermejo-Barrera AM & Strano-Rossi S: Hair and urine analysis: relative distribution of drugs and their metabolites. Forensic Science International 1995; 70:203-210.
    25) Biasi D, Carletto A, & Caramaschi P: Can talc mixed with heroin induce systemic sclerosis?. Expansion Scientific Pub 1998; 157-158.
    26) Blaney SM, Poplack DG, Godwin K, et al: Effect of body position on ventricular CSF methotrexate concentration following intralumbar administration. J Clin Oncol 1995; 13(1):177-179.
    27) Blinick G, Jerez E, & Wallach RC: Methadone maintenance, pregnancy, and progeny. JAMA 1973; 225:477-479.
    28) Bolelli G, Lafisca S, & Flamigni C: Heroin addiction: relationship between the plasma levels of testosterone, dihydrotestosterone, androstenedione, LH, FSH, and the plasma concentration of heroin. Toxicology 1979; 15:19-29.
    29) Bovill JG, Sebel PS, & Blackburn CL: The pharmacokinetics of sufentanil in surgical patients. Anesthesiology 1984; 61:502-506.
    30) Bovill JG: The pharmacokinetics of sufentanil. 55th Congress of IARS (March 8-12), 1981.
    31) Boyer EW & Shannon M: The serotonin syndrome. N Eng J Med 2005; 352(11):1112-1120.
    32) Bralthwaite RA, Elliott SP, & Hale KA: An unusual fatality due to abuse of alfentanil/midazolam mixture (abstract), EAPCCT XX International Congress, Amsterdam, The Netherlands, 2000.
    33) Brewer C: Hair analysis for drugs of abuse (letter). Lancet 1990; 335:980.
    34) 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.
    35) Briggs GG, Freeman RK, & Yaffe SJBriggs GG, Freeman RK, & Yaffe SJ (Eds): Drugs in Pregnancy and Lactation, 5th. Williams & Wilkins, Baltimore, MD, 1998.
    36) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    37) 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.
    38) Brown CV, Rhee P, Chan L, et al: Preventing renal failure in patients with rhabdomyolysis: do bicarbonate and mannitol make a difference?. J Trauma 2004; 56(6):1191-1196.
    39) Bui DH , Pace J , Manjila S , et al: Heroin inhalation complicated by refractory hydrocephalus: A novel presentation. Neurology 2015; 84(20):2093-2095.
    40) Butch AJ, Yokel RA, & Sigell LT: Abuse and pulmonary complications of injecting pentazocine and tripelennamine tablets. Clin Toxicol 1979; 14:301-306.
    41) Camp NE: Drug- and toxin-induced Rhabdomyolysis. J Emerg Nurs 2009; 35(5):481-482.
    42) 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.
    43) 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.
    44) Chan P, Lin TH, & Luo JP: Acute heroin intoxication with complications of acute pulmonary edema, acute renal failure, rhabdomyolysis and lumbosacral plexitis: a case report. Chin Med J 1995a; 55:397-400.
    45) Chan P, Lin TH, & Tsai CW: Unique features of acute heroin intoxication in an epidemic area (abstract 8). Ann Emerg Med 1995; 26:714.
    46) Chan YF, Wong PK, & Chow TC: Acute myoglobinuria as a fatal complication of heroin addiction. Am J Forensic Med Pathol 1990; 11:160-164.
    47) Chen CY, Lee KW, & Lee CC: Heroin-induced spongiform leukoencephalopathy: value of diffusion MR imaging. J Computer Assist Tomography 2000; 24:735-737.
    48) 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.
    49) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    50) Choudry N & Doe J: Inadvertent abuse of amphetamines in street heroin (letter). Lancet 1986; 2:817.
    51) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    52) Claassen JAHR & Gelissen HPMM: The serotonin syndrome (letter). N Eng J Med 2005; 352(23):2455.
    53) Cole JB, Dunbar JF, McIntire SA, et al: Butyrfentanyl overdose resulting in diffuse alveolar hemorrhage. Pediatrics 2015; 135(3):e740-e743.
    54) Coleman L, Carvalho B, Lipman S, et al: Accidental intrathecal sufentanil overdose during combined spinal-epidural analgesia for labor. Int J Obstet Anesth 2009; 18(1):78-80.
    55) Collignon PJ & Sorrell T: Candidiasis in heroin abusers. J Infect Dis 1987; 155:595.
    56) Cone EJ, Welch P, & Paul BD: Forensic drug testing for opiates, III. Urinary excretion rates of morphine and codeine following codeine administration. J Anal Toxicol 1991; 15:161-166.
    57) Cone EJ: Testing human hair for drugs of abuse. I. Individual dose and time profiles of morphine and codeine in plasma, saliva, urine, and beard compared to drug-induced effects on pupils and behavior. J Anal Toxicol 1990; 14:1-7.
    58) Conway BR, Fogarty DG, Nelson WE, et al: Opiate toxicity in patients with renal failure. BMJ 2006; 332(7537):345-346.
    59) Cook S, Moeschler O, & Michaud K: Acute opiate overdose: characteristics of 190 consecutive cases. Addiction 1998; 93:1559-1565.
    60) Cooley S, Lalchandani S, & Keane D: Heroin overdose in pregnancy: an unusual case report. J Obstet Gynaecol 2002; 22:219-220.
    61) Criddle LM: Rhabdomyolysis. Pathophysiology, recognition, and management. Crit Care Nurse 2003; 23(6):14-22, 24-26, 28.
    62) Cunningham EE, Zielezny MA, & Venuto RC: Heroin-associated nephropathy. JAMA 1983; 250:2935-2936.
    63) Cygan J, Trunsky M, & Corbridge T: Inhaled heroin-induced status asthmaticus. Five cases and a review of the literature. Chest 2000; 117:272-275.
    64) Dagnone D, Matsui D, & Rieder MJ: Assessment of the palatability of vehicles for activated charcoal in pediatric volunteers. Pediatr Emerg Care 2002; 18:19-21.
    65) Dan U, Rabinovici Y, Barkai G, et al: Intravenous pethidine and nalbuphine during labor: a prospective double-blind comparative study. Gynecol Obstet Invest 1991; 32(1):39-43.
    66) Dancer SJ, McNair D, & Finn P: Bacillus cereus cellulitis from contaminated heroin. J Med Microbiol 2002; 51:278-281.
    67) Darke S & Ross J: Fatal heroin overdoses resulting from non-injecting routes of administration, NSW, Australia, 1992-1996. Addiction 2000; 95:569-573.
    68) Darke S, Hall W, & Kaye S: Hair morphine concentrations of fatal heroin overdose cases and living heroin users. Addiction 2002; 97:977-984.
    69) Dastur CK & Chang GY: Heroin inhalation complicated by refractory hydrocephalus: A novel presentation. Neurology 2015; 85(23):2083.
    70) Davison AG, Collinson PO, & Assefi AR: Meptazinol overdose producing near fatal respiratory depression. Human Toxicol 1987; 6:331.
    71) De Gans J, Stam J, & Van Wijngaarden GK: Rhabdomyolysis and concomitant neurological lesions after intravenous heroin abuse. J Neurol Neurosurg Psychiatry 1985; 48:1057-1059.
    72) De Negri M & Baglietto MG: Treatment of status epilepticus in children. Paediatr Drugs 2001; 3(6):411-420.
    73) Del Los Santos-Sastre S, Capote-Gil F, & Gonzales-Castro A: Airway obstruction and heroin inhalation. Lancet 1986; 2:1158.
    74) DelGiudice P, Vandenbos F, Boissy C, et al: Cutaneous complications of direct intra-arterial injections in drug addicts. Acta Derm Venereol 2005; 85(5):451-452.
    75) Dettmeyer R, Schmidt P, & Musshoff F: Pulmonary edema in fatal heroin overdose: immunohistological investigations with IgE, collagen IV and laminin - no increase of defects of alveolar-capillary membranes. Forensic Sci Intl 2000; 110:87-96.
    76) Diamant-Berger O, Gherardi R, & Baud F: Intracorporeal concealment of narcotics. Experience at the medico-judical emergency center of the Hotel-Dieu hospital: one hundred cases. Presse Med 1988; 17:107-109.
    77) Duberstein JL & Kaufman DM: A clinical study of an epidemic of heroin intoxication and heroin-induced pulmonary edema. Am J Med 1971; 51:704-714.
    78) Dubrow A, Mittman N, & Ghali V: The changing spectrum of heroin-associated nephropathy. Am J Kidney Dis 1985; 5:36-41.
    79) Dumonceaux GA & Beasley VR: Emergency treatments for police dogs used for illicit drug detection. J Am Vet Med Assoc 1990; 197:185-187.
    80) Egan TD, Lemmens HJM, & Fiset P: The pharmacokinetics of the new short-acting opioid remifentanil (GI7084B) in healthy volunteers. Anesthesiology 1993; 79:881-892.
    81) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    82) Erdman AR & Dart RC: Rhabdomyolysis. In: Dart RC, Caravati EM, McGuigan MA, et al, eds. Medical Toxicology, 3rd ed. Lippincott Williams & Wilkins, Philadelphia, PA, 2004, pp 123-127.
    83) Esposito FM & Winek CL: The synthetic drug 3-methylfentanyl: identification and quantitation of powdered samples. J Forensic Sci 1991; 36:86-92.
    84) Evans LE, Swainson CP, & Roscoe P: Treatment of drug overdosage with naloxone, a specific narcotic antagonist. Lancet 1973; 1:452-455.
    85) FDA: MedWatch Safety Alert: FDA Public Health Advisory Suspended Marketing of Palladone (hydromorphone hydrochloride, extended release capsules). U.S. Food and Drug Administration, Center for Drug Evaluation and Research. Rockville, MDAvailable from URL: http://www.fda.gov/medwatch/SAFETY/2005/safety05.htm#Palladone. As accessed 08/11/2005.
    86) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    87) Farber HW, Mathers JA, & Glauser FL: Gallium scans and serum angiotensin converting enzyme levels in talc granulomatosis and lymphocytic interstitial pneumonitis. South Med J 1980; 73:1663-1666.
    88) Firn S: Accidental poisoning by an animal immobilizing agent. Lancet 1973; 2:95-96.
    89) Fischer CG & Cook DR: The respiratory and narcotic antagonistic effects of naloxone in infants. Anesth Analg 1974; 53:849-852.
    90) Ford M, Hoffman RS, & Goldfrank LR: Opioids and designer drugs. Emerg Med Clin North Am 1990; 8:495-511.
    91) Fragen RJ, Randel GI, & Librojo ES: The interaction of remifentanil and propofol to prevent tracheal response to intubation and the start of surgery for outpatient knee arthroscopy (abstract). Anesthesiology 1994; 81:A376.
    92) Fragen RJ: Implications of the use of remifentanil on patient outcomes. Eur J Anaesthesiol 1995; 12(Suppl):75-76.
    93) Franc-Law JM, Rossignol M, & Vernec A: Poisoning-induced acute atraumatic compartment syndrome. Am J Emerg Med 2000; 18:616-621.
    94) Fung DL, Asling JH, & Eisele JH: A comparison of alphaprodine and meperidine pharmacokinetics. J Clin Pharm 1980; 20:37-41.
    95) Funk L, Grover D, & deSilva H: Compartment syndrome of the hand following intra-arterial injection of heroin. J Hand Surg [Br] 1999; 24(3):366-367.
    96) G Greenberg : Scopolamine poisoning among heroin users - City, Newark, Philadelphia, and Baltimore, 1995 and 1996. Duke University. Durham, NC. 1996. Available from URL: www.Duke.edu.
    97) Gahr M, Freudenmann RW, Hiemke C, et al: "Krokodil":revival of an old drug with new problems. Subst Use Misuse 2012a; 47(7):861-863.
    98) Gahr M, Freudenmann RW, Hiemke C, et al: Desomorphine goes "crocodile". J Addict Dis 2012; 31(4):407-412.
    99) Gal TJ: Naloxone reversal of buprenorphine-induced respiratory depression. Clin Pharmacol Ther 1989; 45:66-71.
    100) Gaspar MR & Hare RR: Gangrene due to intra-arterial injection of drugs by drug addicts. Surgery 1972; 72:573-577.
    101) Gaspar MR & Hare RR: Gangrene due to intra-arterial injection of drugs by drug addicts. Surgery 1972a; 72:573.
    102) Geller RJ & Garrettson LK: Delayed onset of toxicity after methadone ingestion due to therapeutic error (abstract). Vet Hum Toxicol 1994; 36:367.
    103) Giang DW & McBride MC : Lorazepam versus diazepam for the treatment of status epilepticus. Pediatr Neurol 1988; 4(6):358-361.
    104) Gibbs J, Newson T, & Williams J: Naloxone hazard in infant of opioid abuser (letter). Lancet 1989; 2:159-160.
    105) Gillespie TJ, Gandolfi AJ, & Davis TP: Identification and quantification of methylfentanyl in postmortem specimens. J Anal Toxicol 1982; 6:139-142.
    106) Gillman PK: Ecstasy, serotonin syndrome and the treatment of hyperpyrexia (letter). MJA 1997; 167:109-111.
    107) Gillman PK: Successful treatment of serotonin syndrome with chlorpromazine (letter). MJA 1996; 165:345.
    108) Gilman AG, Goodman LS, & Gilman AGilman AG, Goodman LS, & Gilman A (Eds): Goodman and Gilman's The Pharmacological Basis of Therapeutics, 6th. Macmillan Publishing Co, New York, NY, 1980.
    109) Glass PSA, Hardman D, & Kamiyama Y: Preliminary pharmacokinetics and pharmacodynamics of an ultra-short-acting opioid: remifentanil (GI87084B). Anesth Analg 1993; 77:1031-1040.
    110) Glass PSA: Pharmacology of remifentanil. Eur J Anesthesiol 1995; 12 (Suppl):73-74.
    111) Glassroth J, Adams GD, & Schnoll S: The impact of substance abuse on the respiratory system. Chest 1987; 91:596-602.
    112) Gober AE, Kearns GL, & Yokel RA: Repeated naloxone administration for morphine overdose in a 1-month-old infant. Pediatrics 1979; 63:606-608.
    113) Goldberg RJ & Huk M: Serotonin syndrome from trazodone and buspirone (letter). Psychosomatics 1992; 33:235-236.
    114) Goldstein DS, Karpel JP, & Appel D: Bullous pulmonary damage in users of intravenous drugs. Chest 1986; 89:266-269.
    115) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    116) Gomez J & Rodriguez A: An evaluation of the results of a drug sample analysis. Bull Narcotics 1989; 41:121-126.
    117) Gosselin S & Isbister GK: Re: Treatment of accidental intrathecal methotrexate overdose. J Natl Cancer Inst 2005; 97(8):609-610.
    118) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    119) Graham PM: Successful treatment of the toxic serotonin syndrome with chlorpromazine (letter). Med J Australia 1997; 166:166-167.
    120) Griffiths B, Willms L, & Jayathissa S: AV conduction block and proximal myopathy induced by Gee's cough linctus. N Z Med J 2009; 122(1293):3557-.
    121) Guenther Skokan E, Junkins EP, & Corneli HM: Taste test: children rate flavoring agents used with activated charcoal. Arch Pediatr Adolesc Med 2001; 155:683-686.
    122) Guillonneau M, Jacqz-Aigrain E, De-Crepy A, et al: Perinatal adverse effects of nalbuphine given during parturition. Lancet 1990; 335:1588.
    123) Haas CF: Mechanical ventilation with lung protective strategies: what works?. Crit Care Clin 2011; 27(3):469-486.
    124) Hamilton RJ, Perrone J, & Hoffman R: A descriptive study of an epidemic of poisoning caused by heroin adulterated with scopolamine. Clin Toxicol 2000; 38:597-608.
    125) Harchelroad F: Identification of orally ingested cocaine by CT scan (Abstract). Vet Human Toxicol 1992; 34:350.
    126) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    127) Harrison AM, Joo YD, & Israel JR: Intra-arterial administration of oral pentazocine. JAMA 1970; 214:914.
    128) Harrison DW & Walls RM: "Cotton Fever": a benign febrile syndrome in intravenous drug abusers. J Emerg Med 1990; 8:135-139.
    129) Hartoko TJ, Demey HE, & De Schepper AMA: The body packer syndrome-cocaine smuggling in the gastro-intestinal tract. Klin Wochenschr 1988; 66:1116-1120.
    130) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    131) Helander A , Backberg M , & Beck O : MT-45, a new psychoactive substance associated with hearing loss and unconsciousness. Clin Toxicol (Phila) 2014; 52(8):901-904.
    132) Hendrickson RG: Painful and petechial rash after injecting black tar heroin. Clin Toxicol (Phila) 2015; 53(2):134-135.
    133) Herschel M, Khoshnood B, & Lass NA: Role of naloxone in newborn resuscitation. Pediatrics 2000; 106:831-835.
    134) Hewer T, Rose E, & Ghadirian P: Ingested mutagens from opium and tobacco pyrolysis products and cancer of the oesophagus. Lancet 1978; 494-496.
    135) Hibbard R, Wahl M, & Kirshenbaum M: Spiral CT imaging of ingested foreign bodies wrapped in plastic: a pilot study designed to mimic cocaine bodystuffers (abstract). J Toxicol-Clin Toxicol 1999; 37:644.
    136) Hibbs J, Perper J, & Winek CL: An outbreak of designer drug-related deaths in Pennsylvania. JAMA 1991; 265:1011-1013.
    137) Hieger MA, Troendle MM, Kevin MF, et al: Surreptitious adulteration of heroin: a case series of clenbuterol toxicity: 2015 Annual Meeting of the North American Congress of Clinical Toxicology (NACCT). Clin Toxicol 2015; 53(7):655-656.
    138) Hill MD, Cooper PW, & Perry JR: Chasing the dragon - neurological toxicity associated with inhalation of heroin vapour: case report. Can Med Assc J 2000; 162:236-238.
    139) Hock MO, Ooi SB, Saw SM, et al: A randomized controlled trial comparing the hair apposition technique with tissue glue to standard suturing in scalp lacerations (HAT study).. Ann Emerg Med 2002; 40(1):19-26.
    140) 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.
    141) Homsi E, Barreiro MF, Orlando JM, et al: Prophylaxis of acute renal failure in patients with rhabdomyolysis. Ren Fail 1997; 19(2):283-288.
    142) 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.
    143) Howland MA: Antidotes in Depth. In: Goldfrank LR, Flomenbaum N, Hoffman RS, et al, eds. Goldfrank's Toxicologic Emergencies. 8th ed., 8th ed. McGraw-Hill, New York, NY, 2006a, pp 826-828.
    144) Howland MA: Opioid Antagonists. In: Goldfrank LR, Flomenbaum N, eds. Goldfrank's Toxicologic Emergencies. 8th ed., 8th ed. McGraw-Hill, New York, NY, 2006, pp -.
    145) Hsu C, Hung D, & Yang D: The effect of codeine in anti-cough syrup on morphine screen (abstract). J Toxicol-Clin Toxicol 2000; 38:533.
    146) Huerta-Alardin AL, Varon J, & Marik PE: Bench-to-bedside review: Rhabdomyolysis -- an overview for clinicians. Crit Care 2005; 9(2):158-169.
    147) Hughes S & Calverley PMA: Heroin inhalation and asthma. Br Med J 1988; 297:1511-1512.
    148) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    149) Infante F, Dominguez E, & Trujillo D: Metal contamination in illicit samples of heroin. J Forensic Sci 1999; 44:110-113.
    150) Inturrisi CE, Max BM, & Foley KM: The pharmacokinetics of heroin in patients with chronic pain. N Engl J Med 1984; 310:1213-1217.
    151) Iqbal N: Tetanus in IV heroin users. Ann Saudi Med 2001; 21:296-299.
    152) JEF Reynolds : Martindale: The Extra Pharmacopeia (CD-ROM version). The Pharmaceutical Press. London, UK (Internet Version). Edition expires 1989; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    153) Jafar MF & Chowdhry P: Accidental intrathecal administration of high dose diamorphine. J Pak Med Assoc 2014; 64(11):1318-1319.
    154) Jaffe JH & Martin WR: Opioid analgesics and antagonists. Goodman & Gilman's the pharmacological basis of therapeutics 8th ed, Pergamon Press, New York, NY, 1990, pp 485-521.
    155) Jasinski DR, Pevnick JS, & Griffith ID: Human pharmacology and abuse potential of analgesic buprenorphine. Arch Gen Psychiatry 1978; 35:501-516.
    156) Jehanli A, Brannan S, & Moore L: Blind trials of an onsite saliva drug test for marijuana and opiates. J Forensic Sci 2001; 46:1214-1220.
    157) Jolley CJ , Bell J , Rafferty GF , et al: Understanding heroin overdose: a study of the acute respiratory depressant effects of injected pharmaceutical heroin. PLoS One 2015; 10(10):1-.
    158) Jurado C, Gimenez MP, & Menendez M: Simultaneous quantification of opiates, cocaine and cannabinoids in hair. Foren Sci Internat 1995; 70:165-174.
    159) Kaiser KG & Bainton CR: Treatment of intrathecal morphine overdose by aspiration of cerebrospinal fluid. Anesth Analg 1987; 66:475-477.
    160) Kao H, Hung D, & Chang C: Hypoglycemia caused by heroin: a case report (abstract), EAPCCT XVI Int Congress, Vienna, Austria, 1994.
    161) Kelly AM & Koutsogiannis Z: Intranasal naloxone for life threatening opioid toxicity. Emerg Med J 2002; 19:375.
    162) 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.
    163) Kersschot E, Beaucourt L, & Degryse H: Roentgenographical detection of cocaine smuggling in the alimentary tract. Forthschr Rontgenstri 1985; 42:295-298.
    164) Kintz P & Mangin P: Toxicological findings in a death involving dextromethorphan and terfenadine. Amer J Forensic Med Pathol 1992; 13:351-352.
    165) Kintz P & Mangin P: What constitutes a positive result in hair analysis: proposal for the establisment of cut-off values. Forens Sci Internat 1995; 70:3-11.
    166) Kintz P, Mangin P, & Lugnier AA: Toxicological data after heroin overdose. Human Toxicol 1989; 8:487-489.
    167) 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.
    168) Kline SS, Mauro LS, & Scala-Barnett DM: Serotonin syndrome versus neuroleptic malignant syndrome as a cause of death. Clin Pharmac 1989; 8:510-514.
    169) Kofke WA, Garman RH, & Janosky J: Opioid neurotoxicity: neuropathologic effects in rats of different fentanyl congeners and the effects of hexamethonium-induced normotension. Anesth Analg 1996; 83:141-146.
    170) Kollef MH & Schuster DP: The acute respiratory distress syndrome. N Engl J Med 1995; 332:27-37.
    171) Krauss B & Green SM: Procedural sedation and analgesia in children. Lancet 2006; 367(9512):766-780.
    172) Kriegstein AR, Armitage BA, & Kim PY: Heroin inhalation and progressive spongiform leukoencephalopathy (letter). New Engl J Med 1997; 336:589-590.
    173) Krige LP, Milne FJ, & Margolius KA: Rhabdomyolysis and renal failure - unusual complications of drug abuse. S Afr Med J 1983; 64:253-254.
    174) Kronstrand R, Thelander G, Lindstedt D, et al: Fatal intoxications associated with the designer opioid AH-7921. J Anal Toxicol 2014; 38(8):599-604.
    175) Kumar N, Bhalla MC, Frey JA, et al: Intraparenchymal hemorrhage after heroin use. Am J Emerg Med 2015; 33(8):1109.e3-1109.e4.
    176) Kumar R, West DM, & Jingree M: Unusual consequences of heroin overdose: rhabdomyolysis, acute renal failure, paraplegia and hypercalcaemia. Br J Anaesthesia 1999; 83:496-498.
    177) LaBarbera M & Wolfe T: Characteristics, attitudes and implications of fentanyl use based on reports from self-identified fentanyl users. J Psychoactive Drugs 1983; 15:293-301.
    178) Laine K, Kivisto KT, & Ojala-Karlsson P: Effect of activated charcoal on the pharmacokinetics of pholcodine, with special reference to delayed charcoal ingestion. Ther Drug Monitor 1997; 19:46-50.
    179) Larbi EB: Drug induced rhabdomyolysis: case report. East Afr Med J 1997; 74:829-831.
    180) Larijani GE, Goldberg ME, & Rogers KH: Treatment of opioid-induced pruritus with ondansetron: report of four patients. Pharmacotherapy 1996; 16(5):958-960.
    181) 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.
    182) Lawrenson JB, Potgieter PD, & Commerford PJ: Opiate intoxication secondary to the smoking of crude opiates (letter). SAMJ 1993; 83(1):67-68.
    183) Leclerc G, Weber M, & Contet-Audonneau N: Candida folliculitis in heroin addicts. Int J Dermatol 1986; 25:100-102.
    184) Lemmens HJM: Pharmacokinetic-pharmacodynamic relationships for opioids in balanced anaesthesia. Clin Pharmacokinet 1995; 29:231-242.
    185) Lemos NP, Anderson RA, & Valentini R: Analysis of morphine by RIA and HPLC in fingernail clippings obtained from heroin users. J Forensic Sci 2000; 45:407-412.
    186) Levine B & Smialek JE: Considerations in the interpretation of urine analyses in suspected opiate intoxications. J Forensic Sci 1998; 43:388-389.
    187) Lewis JW, Groux N, & Elliot P: Complications of attempted central venous injections performed by drug abusers. Chest 1980; 78:4.
    188) Lin TJ, Ger J, & Yang GC: Dihydroetorphine addiction (abstract 9). Ann Emerg Med 1995; 26:715.
    189) Lloyd WK, Porter JM, & Lindell TD: Accidental intra-arterial injection in drug abuse. Am J Roentgenol 1973; 117:892.
    190) Lo DST & Chua TH: Poppy seeds: implications of consumption. Med Sci Law 1992; 32:296-302.
    191) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    192) Long H, Deore K, & Hoffman RS: Heroin-induced leukoencephalopathy due to chasing the dragon (abstract). Clin Toxicol 2002; 40:654.
    193) Long H, Deore K, Hoffman RS, et al: A fatal case of spongiform leukoencephalopathy linked to "chasing the dragon". J Toxicol Clin Toxicol 2003; 41:887-891.
    194) Maio RF, Gaukel B, & Freeman B: Intralingual naloxone injection for narcotic-induced respiratory depression. Ann Emerg Med 1987; 16:572-573.
    195) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    196) Marc B, Baud FJ, & Aelion MJ: The cocaine body-packer syndrome: evaluation of a method of contrast study of the bowel. J Forens Sci 1990; 35:345-355.
    197) Maxwell TM, Olcott C, & Blaisdell FW: Vascular complications of drug abuse. Arch Surg 1972; 105:875.
    198) McCann B, Hunter R, & McCann J: Cocaine/heroin induced rhabdomyolysis and ventricular fibrillation. Emerg Med J 2002; 19:264-265.
    199) McCann B, Hunter R, & McCann J: Cocaine/heroin induced rhabdomyolysis and ventricular fibrillation. Emerg Med J 2002a; 19(3):264-265.
    200) McCarron MM & Wood JD: The cocaine body packer syndrome. JAMA 1983; 250:1417-1420.
    201) McCreary M, Emerman C, & Hanna J: Acute myelopathy following intranasal insufflation of heroin: a case report. Neurology 2000; 55:316-317.
    202) McDonald FW, Cienki JJ, & Horowitz RS: Delayed post-anoxic encephalopathy after heroin use, EAPCCT XVII Int Congress, Krakow, Poland, 1995.
    203) McDonald RD, Evans FT, & Weise VK: Effect of morphine and nalorphine on plasma hydrocortisone levels in man. J Pharmacol Exp Ther 1959; 125:241-247.
    204) Meatherall R & Dai J: False-positive EMIT II opiates from ofloxacin. Ther Drug Monitor 1997; 19:98-99.
    205) Melandri R, Re G, & Lanzarini C: Myocardial damage and rhabdomyolysis associated with prolonged hypoxic coma following opiate overdose. Clin Tox 1996; 34:199-203.
    206) Melandri R, Re G, & Lanzarini C: Myocardial damage and rhabdomyolysis associated with prolonged hypoxic coma following opiate overdose. Clin Toxicol 1996a; 34:199-203.
    207) Mikkelsen SL & Ash KO: Adulterants causing false negatives in illicit drug testing. Clin Chem 1988; 34:2333-2336.
    208) Miller M, Barber CW, Leatherman S, et al: Prescription opioid duration of action and the risk of unintentional overdose among patients receiving opioid therapy. JAMA Intern Med 2015; 175(4):608-615.
    209) Miller RR: Clinical effects of parenteral narcotics in hospitalized medical patients. J Clin Pharm 1980; 20:165-171.
    210) Mills KC: Serotonin syndrome: a clinical update. Med Toxicol 1997; 13:763-783.
    211) Mitchell PA & Pons PT: Wound botulism associated with black tar heroin and lower extremity cellulitis. J Emerg Med 2001; 20:371-375.
    212) Mitchell WG: Status epilepticus and acute repetitive seizures in children, adolescents, and young adults: etiology, outcome, and treatment. Epilepsia 1996; 37(S1):S74-S80.
    213) Moeller MR & Mueller C: The detection of 6-monoacetylmorphine in urine, serum and hair by GC/MS and RIA. Forensic Science International 1995; 70:125-133.
    214) Mofenson HC & Caraccio TR: Continuous infusion of intravenous naloxone (letter). Ann Emerg Med 1987; 16:374-375.
    215) Molina DK & Dimaio VJ: The reliability of immunoassay for determining the presence of opiates in the forensic setting. Am J Forensic Med Pathol 2005; 26(4):303-304.
    216) Moore KA, Addison J, & Levine B: Applicability of opiate cutoffs to opiate intoxication cases (letter). J Analy Toxicol 2001; 25:657-658.
    217) Murray S & Wooltorton E: Alcohol-associated rapid release of a long-acting opioid. CMAJ 2005; 173(7):756.
    218) 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.
    219) Nair EL, Cienkowski KM, & Michaelides E: The impact of sudden hearing loss secondary to heroin overdose on fitting outcomes. Am J Audiol 2010; 19(2):86-90.
    220) Nelson L: Opioids. In: Goldfrank LR, Flomenbaum N, eds. Goldfrank's Toxicologic Emergencies. 8th ed., 8th ed. McGraw-Hill, New York, NY, 2006, pp -.
    221) Nelson LS: Opioids, in Goldfrank LR, Flomenbaum NE, Lewin NA et al (eds): Goldfrank's Toxicologic Emergencies, 6th ed, Appleton & Lange, Stamford, CT, 1998.
    222) Neugarten J, Gallo GR, Buxbaum J, et al: Amyloidosis in subcutaneous heroin abusers ("skin poppers' amyloidosis"). Am J Med 1986; 81(4):635-640.
    223) Nicholls K, Niall JF, & Moran JE: Rhabdomyolysis and renal failure. Med J Aust 1982; 2:387-389.
    224) Nocchi D & D'az JE: Heroin-induced priapism (abstract). J Toxicol-Clin Toxicol 1999; 37:646-647.
    225) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    226) Ochoa-Gomez J, Villar-Arias A, & Aresti I: A case of severe hyperkalaemia and compartment syndrome due to rhabdomyolysis after drugs abuse. Resuscitation 2002; 54:103-105.
    227) Oh AK, Ishiyama A, & Baloh RW: Deafness associated with abuse of hydrocodone/acetaminophen. Neurology 2000; 54:2345.
    228) Oliver RM: Bronchospasm and heroin inhalation (letter). Lancet 1986; 1:915.
    229) Olives T & Cole J: Systemic inflammatory response syndrome following injection of boiled poppy seed tea to avoid withdrawal. Clin Toxicol (Phila) 2015; 53(7):678.
    230) Onesti MG, Fioramonti P, Fino P, et al: Skin ulcer caused by venous extravasation of heroin. Int Wound J 2014; 11(4):409-411.
    231) Osterhoudt KC & Perrone J: Induced hypothermia for drug overdose (letter). Acad Emerg Med 2002; 9:962.
    232) Parkinson SK, Bailey SL, & Little WL: Myoclonic seizure activity with chronic high-dose spinal opioid administration. Anesthesiology 1990; 72:743-745.
    233) Passaro DJ, Werner SB, & McGee J: Wound botulism associated with black tar heroin among injecting drug users. J Am Med Assoc 1998; 279:859-863.
    234) Patrick SW , Davis MM , Lehmann CU , et al: Increasing incidence and geographic distribution of neonatal abstinence syndrome: United States 2009 to 2012. J Perinatol 2015; 35(8):650-655.
    235) Paul BD, Mitchell JM, & Mell LD Jr: Gas chromatography/electron impact mass fragmentometric determination of urinary 6-acetylmorphine, a metabolite of heroin. J Anal Toxicol 1989; 13:2-7.
    236) 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.
    237) Penn RD & Kroin JS: Treatment of intrathecal morphine overdose. J Neurosurg 1995; 82(1):147-148.
    238) Perrone J, Shaw L, & DeRoos F: Laboratory confirmation of scopolamine co-intoxication in patients using tainted heroin. Clin Toxicol 1999; 37:491-496.
    239) Podzamczer D, Ribera M, & Gudiol F: Skin abscesses caused by Candida albicans in heroin abusers. J Am Acad Dermatol 1987; 16:386-387.
    240) Polderman KH: Acute renal failure and rhabdomyolysis. Int J Artif Organs 2004; 27(12):1030-1033.
    241) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    242) Porter JM, Wesche DH, & Rosch J: Intra-arterial sympathetic blockade in the treatment of clinical frostbite. Am J Surg 1976; 132:625-630.
    243) Prachand N, Krantz A, & Franklin C: Severe asthma associated with heroin insufflation (abstract). J Toxicol-Clin Toxicol 1999; 37:646.
    244) Product Information: ALFENTA(R) IV injection, alfentanil HCl IV injection. Akorn, Inc., Lake Forest, IL, 2008.
    245) Product Information: ATIVAN(R) injection, lorazepam injection. Baxter Healthcare Corporation, Deerfield, IL, 2003.
    246) Product Information: AVINZA(R) extended-release oral capsules, morphine sulfate extended-release oral capsules. Ligand Pharmaceuticals Incorporated, San Diego, CA, 2005.
    247) Product Information: AVINZA(R) extended-release oral capsules, morphine sulfate extended-release oral capsules. King Pharmaceuticals Inc, Bristol, TN, 2008.
    248) Product Information: Alfenta(R), alfenanil hydrochloride injection. Janssen Pharmaceutica Inc, Titusville, NJ, USA, 1995.
    249) Product Information: Alfenta(R), alfentanil. Janssen Pharmaceutica, Inc, Piscataway, NJ, 1987.
    250) Product Information: B&O SUPPRETTES(R) No. 15A and No. 16A rectal suppositories, belladonna, opium rectal suppositories. PolyMedica, Woburn, MA, 2002.
    251) Product Information: Belladonna & Opium rectal suppositories, Belladonna & Opium rectal suppositories. Paddock Laboratories, Inc, Minneapolis, MN, 2005.
    252) Product Information: Duragesic(R), fentanyl transdermal system. Janssen Pharmaceutica Products, Titusville, NJ, 2001.
    253) Product Information: EMBEDA(R) oral extended-release capsules, morphine sulfate and naltrexone hcl oral extended-release capsules. King Pharmaceuticals, Inc., Bristol, TN, 2009.
    254) Product Information: ENTEREG(R) oral capsules, alvimopan oral capsules. Adolor Corp, Exton, PA, 2008.
    255) Product Information: ENTEREG(R) oral capsules, alvimopan oral capsules. Cubist Pharmaceuticals, Inc. (per FDA), Lexington, MA, 2012.
    256) Product Information: ENTEREG(R) oral capsules, alvimopan oral capsules. Cubist Pharmaceuticals, Inc. (per manufacturer), Lexington, MA, 2013.
    257) Product Information: EVZIO(TM) injection solution, naloxone HCl injection solution. Kaleo, Inc. (per FDA), Richmond, VA, 2014.
    258) Product Information: EXALGO(R) extended release oral tablets, hydromorphone hydrochloride extended release oral tablets. ALZA Corporation, Vacaville, CA, 2010.
    259) Product Information: Infumorph(R), morphine sulfate. Elkins-Sinn, Cherry Hill, NJ, 1993.
    260) Product Information: NARCAN(R) nasal spray, naloxone HCl nasal spray. Adapt Pharma (per FDA), Radnor, PA, 2015.
    261) Product Information: NUBAIN(R) injection, nalbuphine hcl injection. Endo Pharmaceuticals, Chadds Ford, PA, 2005.
    262) Product Information: Nubain(R), nalbuphine. Endo Laboratories, Garden City, NY, 1983.
    263) Product Information: OPANA(R)ER extended release oral tablets, oxymorphone hcl extended release oral tablets. Endo Pharmaceuticals, Chadds Ford, PA, 2007.
    264) Product Information: Paregoric, anhydrous morphine. Geneva Generics, Broomfield, CO, 1998.
    265) Product Information: REVEX(R) injection, nalmefene hcl injection. Baxter Healthcare Corporation, Deerfield, IL, 2006.
    266) Product Information: Sufenta(R), sufentanil citrate. Janssen Pharmaceutica, Titusville, New Jersey, 1987a.
    267) Product Information: ULTIVA(R) IV injection, remifentanil hydrochloride IV injection. Abbott Laboratories, North Chicago, IL, 2007.
    268) Product Information: Ultiva(R), remifentanil hydrochloride. Abbott Laboratories, North Chicago, IL, 2004.
    269) Product Information: Ultiva(R), remifentanil hydrochloride. Glaxo Wellcome Inc, Research Triangle Park, North Carolina, 1996.
    270) Product Information: Ultiva(R), remifentanil hydrochloride. Glaxo Wellcome Inc, Research Triangle Park, North Carolina, 1999.
    271) Product Information: VIBERZI oral tablets, eluxadoline oral tablets. Patheon Pharmaceuticals, Inc. (per Manufacturer), Cincinnati, OH, 2015.
    272) Product Information: VIVITROL(TM) extended-release injectable suspension, naltrexone extended-release injectable suspension. Cephalon,Inc, Frazer, PA, 2006.
    273) Product Information: alfentanil HCl intravenous injection, alfentanil HCl intravenous injection. Akorn, Inc. (per DailyMed), Lake Forest, IL, 2013.
    274) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    275) Product Information: dopamine hcl, 5% dextrose IV injection, dopamine hcl, 5% dextrose IV injection. Hospira,Inc, Lake Forest, IL, 2004.
    276) Product Information: hydromorphone hcl oral tablets, hydromorphone hcl oral tablets. Mallinckrodt Inc, St. Louis, MO, 2003.
    277) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    278) Product Information: nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution, nalbuphine hydrochloride subcutaneous injection, IM injection, IV injection, solution. Hospira Inc., Lake Forest, IL, 2007.
    279) Product Information: naloxone HCl IV, IM, subcutaneous injection solution, naloxone HCl IV, IM, subcutaneous injection solution. Hospira, Inc (per DailyMed), Lake Forest, IL, 2008.
    280) Product Information: norepinephrine bitartrate injection, norepinephrine bitartrate injection. Sicor Pharmaceuticals,Inc, Irvine, CA, 2005.
    281) Product Information: opium tincture (DEODORIZED) oral solution, opium tincture (DEODORIZED) oral solution. Marathon Pharmaceuticals, LLC, Deerfield, IL, 2008.
    282) Product Information: opium tincture, opium tincture. Ranbaxy Pharmaceuticals,Inc., Gloversville, NY, 2003.
    283) Product Information: paregoric oral solution, paregoric oral solution. Barre-National Inc, Baltimore, MD, 1996.
    284) Product Information: sufentanil citrate injection, sufentanil citrate injection. Abbott Laboratories, North Chicago, IL, 2003.
    285) Product Information: sufentanil citrate intravenous injection, epidural injection, sufentanil citrate intravenous injection, epidural injection. West-ward Pharmaceutical Corp. (per DailyMed), Eatontown, NJ, 2011.
    286) Product Information: tapentadol immediate release oral tablets, tapentadol immediate release oral tablets. Ortho-McNeil-Janssen Pharmaceuticals Inc, Raritan, NJ, 2008.
    287) Pruefer C & Bewlay A: Respiratory arrest with remifentanil patient-controlled analgesia--another case. Anaesthesia 2012; 67(9):1044-1045.
    288) Pullan PT, Watson FE, & Seow SSW: Methadone-induced hypoadrenalism. Lancet 1983; 1:714.
    289) Qureshi A, Wassmer E, Davies P, et al: Comparative audit of intravenous lorazepam and diazepam in the emergency treatment of convulsive status epilepticus in children. Seizure 2002; 11(3):141-144.
    290) Raikos N, Tsoukali H, & Njau SN: Determination of opiates in postmortem bone and bone marrow. Forensic Sci Intl 2001; 123:140-141.
    291) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    292) Redfern N: Dihydrocodeine overdose treated with naloxone infusion. Br Med J 1983; 287:751-752.
    293) Reed D: Presented at the quarterly meeting of the California Association of Toxicologists, San Jose, CA, 1981.
    294) Reents SB: Paradoxical response to high-dose nalbuphine. Clin Pharm 1986; 5:359-370.
    295) Remskar M, Noc M, & Leskovsek B: Profound circulatory shock following heroin overdose. Resuscitation 1998; 38:51-53.
    296) Rice EK, Isbel NM, & Becker GJ: Heroin overdose and myoglobinuric acute renal failure. Clin Nephrol 2000; 54:449-454.
    297) Rice TB: Paregoric intoxication with pulmonary edema in infancy. Clin Pediatr 1984; 23:101-103.
    298) Ringertz SH, Hoiby EA, & Jensenius M: Injectional anthrax in a heroin skin-popper. Lancet 2000; 356:1574-1575.
    299) Riva N, Morana P, Cerri F, et al: Acute myelopathy selectively involving lumbar anterior horns following intranasal insufflation of ecstasy and heroin. J Neurol Neurosurg Psychiatry 2007; 78(8):908-909.
    300) Ropper AH & Blair R: Symmetric deep cerebellar lesions after smoking heroin. Arch Neurol 2003; 60:1605-1606.
    301) Rosow CE: Remifentanil: a unique opioid analgesic. Anesthesiology 1993; 79:875-876.
    302) Rosow CE: Sufentanil citrate: a new opioid analgesic for use in anesthesia. Pharmacotherapy 1984; 4:11-19.
    303) Russo A, Ciocca F, & Vivino G: Severe rhabdomyloysis and compartment syndrome in poisoned patients (abstract 48). Ann Emerg Med 1995; 26:726.
    304) Rutili A, Maggiani M, Bertelloni C, et al: Persistent overdose caused by a very small dose of intrathecal morphine in an elderly patient undergoing vaginal hysterectomy: a case report. Minerva Anestesiol 2007; 73(7-8):433-436.
    305) Saba GP, James AE, & Johnson BA: Pulmonary complications of narcotic abuse. Am J Roentgenol Radium Ther Nucl Med 1974; 122:733-739.
    306) Sadeghi A, Behmard S, & Vesselinovitch SD: Opium: a potential urinary bladder carcinogen in man. Cancer 1979; 43:2315-2321.
    307) Schiffer D, Brignolio F, & Giordana MT: Spongiform encephalopathy in addicts inhaling pre-heated heroin. Clin Neuropathol 1985; 4:174-180.
    308) Schneir AB, Vadeboncoeur TF, & Offerman SR: Massive Oxycontin(R) ingestion refractory to naloxone (abstract). Clin Toxicol 2002; 40:617.
    309) Schweitzer VG, Darrat I, Stach BA, et al: Sudden bilateral sensorineural hearing loss following polysubstance narcotic overdose. J Am Acad Audiol 2011; 22(4):208-214.
    310) Scott JC, Ponganis KV, & Stanski DR: EEG quantitation of narcotic effect: the comparative pharmacodynamics of fentanyl and alfentanil. Anesthesiology 1985; 62:234-241.
    311) 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.
    312) Seifert S: Opioid Medications. In: Dart RC, ed. Medical toxicology. 3rd ed., 3rd ed. Lippincott, Williams & Wilkins, Philadelphia, PA, 2004, pp -.
    313) Selavka CM: Poppy seed ingestion as a contributing factor to opiate-positive urinalysis results: the Pacific perspective. J Forensic Sci 1991; 36:685-696.
    314) Sempere AP, Posada I, & Ramo C: Spongiform leucoencephalopathy after inhaling heroin. Lancet 1991; 338:320.
    315) Sey MJ, Rubinstein D, & Smith DS: Accidental methadone intoxication in a child. Pediatrics 1971; 48:294-296.
    316) Sgro C, Escousse A, Tennenbaum D, et al: Perinatal adverse effects of nalbuphine given during labour. Lancet 1990; 336:1070.
    317) Shakeri R, Malekzadeh R, Etemadi A, et al: Opium: an emerging risk factor for gastric adenocarcinoma. Int J Cancer 2013; 133(2):455-461.
    318) Shesser R, Jotte R, & Olshaker J: The contribution of impurities to the acute morbidity of illegal drug use. Am J Emerg Med 1991; 9:336-342.
    319) Simon EJ & Hiller JMI: The opiate receptors. Ann Rev Pharmacol Toxicol 1978; 18:371-394.
    320) Skowronek R, Celinski R, & Chowaniec C: "Crocodile"--new dangerous designer drug of abuse from the East. Clin Toxicol (Phila) 2012; 50(4):269-269.
    321) Smith RM & Nelsen LA: Hmong folk remedies: limited acetylation of opium by aspirin and acetaminophen. J Forensic Sci 1991; 36:280-287.
    322) Snow J, Tormoehlen LM, & Hetrick A: Delayed Post-Hypoxic Leukoencephalopathy following Heroin Abuse with Serial Neuropsychometric Testing. Clin Toxicol (Phila) 2015; 53(7):679.
    323) Snyder SH, Pert CB, & Posternak GW: The opiate receptor. Ann Intern Med 1974; 81:534-540.
    324) Speckl IM, Hallbach J, & Guder WG: Opiate detection in saliva and urine- a prospective comparison by gas chromatography-mass spectrometry. Clin Toxicol 1999; 37:441-445.
    325) Spiller HA & Rogers GC: Evaluation of administration of activated charcoal in the home. Pediatrics 2002; 108:E100.
    326) Sporer KA & Dorn E: Heroin-related noncardiogenic pulmonary edema. Chest 2001; 120:1628-1632.
    327) Sporer KA, Firestone J, & Isaacs SM: Out-of-hospital treatment of opioid overdoses in an urban setting. Acad Emerg Med 1996; 3:660-667.
    328) Sporer KA: Acute heroin overdose. Ann Intern Med 1999; 130:584-590.
    329) 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.
    330) Stamboulis E, Psimaras A, & Malliara-Loulakaki S: Brachial and lumbar plexitis as a reaction to heroin. Drug Alcohol Depend 1988; 22:205-207.
    331) Staub C: Hair analysis: its importance for the diagnosis of poisoning associated with opiate addiction. Forensic Science International 1993; 63(1-3):69-75.
    332) Steinberg AD & Karlinger JS: The clinical spectrum of heroin pulmonary edema. Arch Intern Med 1968; 122:122-127.
    333) Sternbach H: The serotonin syndrome. Am J Psychiatr 1991; 148:705-713.
    334) Sterrett C, Brownfield J, Korn CS, et al: Patterns of presentation in heroin overdose resulting in pulmonary edema. Am J Emerg Med 2003; 21:32-34.
    335) 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.
    336) Strang J, Marsh A, & Desouza N: Hair analysis for drugs of abuse (letter). Lancet 1990; 335:740.
    337) Strong WE & Matson M: Probable seizure after alfentanil. Anesth Analg 1989; 68:692-693.
    338) Struempler RE: Excretion of codeine and morphine following ingestion of poppy seeds. J Analytical Toxicol 1987; 11:97-99.
    339) Stueber K: The treatment of intraarterial pentazocine injection injuries with intraarterial reserpine. Ann Plastic Surg 1987; 18:41-46.
    340) Tagliaro F, De Battisti Z, & Smith FP: Death from heroin overdose: findings from hair analysis. Lancet 1998; 351:1923-1925.
    341) Tagliaro F, Dorizzi R, & Lafisca S: Calcitonin serum levels in heroin addicts: effects of methadone and clonidine detoxication treatments. Drug Alcohol Depend 1985; 16:181-183.
    342) Tandberg D & Abercrombie D: Treatment of heroin overdose with endotracheal naloxone. Ann Emerg Med 1982; 11:443-445.
    343) Tenenbein M: Continuous naloxone infusion for opiate poisoning in infancy. J Pediatr 1984; 105:645-648.
    344) Thakore S & Murphy N: The potential role of prehospital administration of activated charcoal. Emerg Med J 2002; 19:63-65.
    345) U.S. Drug Enforcement Administration: Clenbuterol (Street Names: Clen). U.S. Drug Enforcement Administration. Springfield, VA. 2009. Available from URL: http://www.deadiversion.usdoj.gov/drugs_concern/clenbuterol.htm. As accessed 2012-03-06.
    346) USP: Drug Information for the Health Care Professional, 8th ed. United States Pharmacopeial Convention, Inc, 1988.
    347) Umans JG & Szeto HH: Precipitated opiate abstinence in utero. Am J Obstet Gynecol 1985; 151:441-444.
    348) Utecht MJ, Stone AF, & McCarron MM: Heroin body packers. J Emerg Med 1993; 11:33-40.
    349) Van Nesselrooij BPM, Roumen FJME, & Da Costa AJ: (Post-asphyctic encephalopathy of the neonate following administration of nalbuphine during childbirth). Ned Tijdschr Genesskd 1992; 136:1073-1076.
    350) 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.
    351) 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.
    352) Vanholder R, Sever MS, Erek E, et al: Rhabdomyolysis. J Am Soc Nephrol 2000; 11(8):1553-1561.
    353) Vila N & Chamorro A: Ballistic movements due to ischemic infarcts after intravenous heroin overdose: report of two cases. Clin Neurol & Neurosurg 1997; 99:259-262.
    354) Walter LA & Catenacci MH: Rhabdomyolysis. Hosp Physician 2008; 44(1):25-31.
    355) 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.
    356) Warner-Smith M, Darke S, & Day C: Morbidity associated with non-fatal heroin overdose. Addiction 2002; 97:963-967.
    357) Warner-Smith M, Darke S, & Lynskey M: Heroin overdose: causes and consequences (review). Addiction 2001; 96:1113-1125.
    358) Watson WA, Steele MT, & Muelleman RL: Opioid toxicity recurrence after an initial response to naloxone. Clin Toxicol 1998; 36:11-17.
    359) 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.
    360) Weber W, Henkes H, & Moller P: Toxic spongiform leucoencephalopathy after inhaling heroin vapour. Eur Radiol 1998; 8:749-755.
    361) 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.
    362) Westerling D, Sawe J, & Eklundh G: Near fatal intoxication with controlled- release morphine tablets in a depressed woman. Acta Anaesthesiol Scand 1998; 42:586-589.
    363) Westmoreland CL, Hoke JF, & Sebel PS: Pharmacokinetics of remifentanil (GI87084B) and its major metabolite (GI90291) in patients undergoing elective inpatient surgery. Anesthesiology 1993; 79:893-903.
    364) Wheless JW : Treatment of status epilepticus in children. Pediatr Ann 2004; 33(6):376-383.
    365) Whipple JK, Quebbeman EJ, & Lewis KS: Difficulties in diagnosing narcotic overdoses in hospitalized patients. Ann Pharmacotherapy 1994; 28:446-450.
    366) White JM & Irvine RJ: Mechanisms of fatal opioid overdose (review). Addiction 1999; 94:961-972.
    367) Wichmann S, Nielsen SL, Siersma VD, et al: Risk factors for 48-hours mortality after prehospital treatment of opioid overdose. Emerg Med J 2012; Epub:Epub.
    368) Wiener PC, Hogg MIJ, & Rosen M: Effects of naloxone on pethidine-induced neonatal depression. Part I. Br Med J 1977; 2:228-229.
    369) Wilen SB, Ulreich S, & Rabinowitz JG: Roentgenographic manifestations of methodone-induced pulmonary edema. Radiology 1975; 114:51-55.
    370) Wilkes TCR, Davies DP, & Dar MF: Apnea in a 3 month old baby prescribed compound linctus containing codeine. Lancet 1981; 1:1166-1167.
    371) Willson DF, Truwit JD, Conaway MR, et al: The adult calfactant in acute respiratory distress syndrome (CARDS) trial. Chest 2015; 148(2):356-364.
    372) 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.
    373) Wilson SC, Stiller RL, & Davis PJ: Fentanyl and alfentanil plasma protein binding in preterm and term neonates. Anesth Analg 1997; 84:315-318.
    374) Wolff AP, Hasenbos MA, & Liem TH: Accidental overdose of epidural bupivacaine and sufentanil. Regional Anesthesia 1992; 17:237-238.
    375) Wolters EC, Stam FC, & Lousberg RJ: Leucoencephalopathy after inhaling heroin pyrolysate. Lancet 1982; 2:1233-1237.
    376) Wyman J & Bultman S: Postmortem distribution of heroin metabolites in femoral blood, liver, cerebrospinal fluid, and vitreous humor. J Analyt Toxicol 2004; 28:260-263.
    377) Zebelman AM, Troyer BL, & Randall GL: Detection of morphine and codeine following consumption of poppy seeds. J Analytical Toxicol 1987; 11:131-132.
    378) Zuckerman GB, Ruiz DC, Keller IA, et al: Neurologic complications following intranasal administration of heroin in an adolescent. Ann Pharmacother 1996; 30(7-8):778-781.
    379) Zuckerman M, Weisberg SN, & Boyer EW: Pitfalls of intranasal naloxone. Prehosp Emerg Care 2014; 18(4):550-554.
    380) Zuspan GP, Gumpel JA, & Mejia-Zelaya A: Fetal stress from methadone withdrawal. Am J Obstet Gynecol 1975; 122:43-46.
    381) deGans J, Stam J, & vanWijngaarden GK: Rhabdomyolysis and concomitant neurological lesions after intravenous heroin abuse. J Neurol Neurosurg Psychiatry 1985; 48(10):1057-1059.
    382) doSameiroFaria M, Sampaio S, Faria V, et al: Nephropathy associated with heroin abuse in Caucasian patients. Nephrol Dial Transplant 2003; 18(11):2308-2313.